1
|
Archavlis E, Palombi D, Konstantinidis D, Carvi y Nievas M, Trobisch P, Stoyanova II. Pathophysiologic Mechanisms of Severe Spinal Cord Injury and Neuroplasticity Following Decompressive Laminectomy and Expansive Duraplasty: A Systematic Review. Neurol Int 2025; 17:57. [PMID: 40278428 PMCID: PMC12030159 DOI: 10.3390/neurolint17040057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/06/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Severe spinal cord injury (SCI) represents a debilitating condition with long-term physical and socioeconomic impacts. Understanding the pathophysiology of SCI and therapeutic interventions such as decompressive laminectomy and expansive duraplasty is crucial for optimizing patient outcomes. OBJECTIVE This systematic review explores the pathophysiology of SCI and evaluates evidence linking decompressive laminectomy and duraplasty to improved neuroplasticity and recovery. METHODS A comprehensive search was conducted in PubMed, Web of Science, and Cochrane Library for studies on decompressive surgery in SCI. Inclusion criteria were original articles investigating pathophysiology, neuroplasticity mechanisms, or surgical outcomes. Data on pathophysiological changes, molecular markers, and functional outcomes were extracted. RESULTS From 1240 initial articles, 43 studies were included, encompassing both animal models and human clinical data. Findings highlighted the role of inflammatory cascades, blood-spinal cord barrier disruption, and neurotrophic factor modulation in recovery. Decompressive duraplasty was associated with improved intrathecal pressure (ITP) management and neuroplasticity markers, such as BDNF and GAP-43. CONCLUSIONS This review underscores the therapeutic potential of decompressive laminectomy and duraplasty in SCI. While evidence suggests benefits in promoting neuroplasticity, further research is needed to elucidate molecular mechanisms and refine interventions.
Collapse
Affiliation(s)
- Eleftherios Archavlis
- Interdisciplinary Spine Center and Department of Neurosurgery, Elisabethen Hospital, 60487 Frankfurt, Germany
- School of Health, IU University of Applied Sciences, 53604 Bad Honnef, Germany
- School of Medicine, Frankfurt Branch, European University Cyprus, 60487 Frankfurt, Germany (I.I.S.)
| | - Davide Palombi
- Neurosurgery Section, Department of Neuroscience, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00136 Rom, Italy
| | - Dimitrios Konstantinidis
- Interdisciplinary Spine Center and Department of Neurosurgery, Elisabethen Hospital, 60487 Frankfurt, Germany
| | - Mario Carvi y Nievas
- School of Medicine, Frankfurt Branch, European University Cyprus, 60487 Frankfurt, Germany (I.I.S.)
| | - Per Trobisch
- Department of Spine Surgery, Eifelklinik St. Brigida, 52152 Simmerath, Germany
| | - Irina I. Stoyanova
- School of Medicine, Frankfurt Branch, European University Cyprus, 60487 Frankfurt, Germany (I.I.S.)
| |
Collapse
|
2
|
Luo Y, Liu R, Yuan G, Pan Y. Polyphenols for stroke therapy: the role of oxidative stress regulation. Food Funct 2024; 15:11383-11399. [PMID: 39497601 DOI: 10.1039/d4fo01900h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Stroke is associated with a high incidence and disability rate, which seriously endangers human health. Oxidative stress (OS) plays a crucial role in the underlying pathologic progression of cerebral damage in stroke. Emerging experimental studies suggest that polyphenols have antioxidant potential and express protective effects after different types of strokes, but no breakthrough has been achieved in clinical studies. Nanomaterials, due to small characteristic sizes, can be used to deliver drugs, and have shown excellent performance in the treatment of various diseases. The drug delivery capability of nanomaterials has significant implications for the clinical translation and application of polyphenols. This comprehensive review introduces the mechanism of oxidative stress in stroke, and also summarizes the antioxidant effects of polyphenols on reactive oxygen species generation and oxidative stress after stroke. Also, the application characteristics and research progress of nanomaterials in the treatment of stroke with antioxidants are presented.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
3
|
Barata P, Camacho O, Lima CG, Pereira AC. The Role of Hyperbaric Oxygen Therapy in Neuroregeneration and Neuroprotection: A Review. Cureus 2024; 16:e62067. [PMID: 38989389 PMCID: PMC11235151 DOI: 10.7759/cureus.62067] [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] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Neurogenesis is a high energy-demanding process, which is why blood vessels are an active part of the neurogenic niche since they allow the much-needed oxygenation of progenitor cells. In this regard, although neglected for a long time, the "oxygen niche" should be considered an important intervenient in adult neurogenesis. One possible hypothesis for the failure of numerous neuroprotective trials is that they relied on compounds that target a highly specific neuroprotective pathway. This approach may be too limited, given the complexity of the processes that lead to cell death. Therefore, research should adopt a more multifactorial approach. Among the limited range of agents with multimodal neuromodulatory capabilities, hyperbaric oxygen therapy has demonstrated effectiveness in reducing secondary brain damage in various brain injury models. This therapy functions not only as a neuroprotective mechanism but also as a powerful neuroregenerative mechanism.
Collapse
Affiliation(s)
- Pedro Barata
- Pathology and Laboratory Medicine, Centro Hospitalar Universitário do Porto, Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| | - Oscar Camacho
- Hyperbaric Medicine Unit, Unidade Local de Saúde de Matosinhos, Matosinhos, PRT
| | - Clara G Lima
- Anesthesiology, Hospital Pedro Hispano, Matosinhos, PRT
| | - Ana Claudia Pereira
- Faculty of Health Sciences, Universidade Fernando Pessoa (UFP), Porto, PRT
- CECLIN (Center for Clinical Studies), Hospital-Escola da Universidade Fernando Pessoa (HE-UFP), Porto, PRT
| |
Collapse
|
4
|
Wan R, Su Y, Zhu M, Huang Y. Elevated blood malondialdehyde level contributed to a high stroke risk in a Chinese elderly population from rural areas: a cross-sectional study. Sci Rep 2024; 14:4325. [PMID: 38383846 PMCID: PMC10881991 DOI: 10.1038/s41598-024-54419-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 02/13/2024] [Indexed: 02/23/2024] Open
Abstract
Individuals living in rural areas have a higher incidence rate of stroke than their urban counterparts in China. However, few studies have investigated the association between blood malondialdehyde (MDA), an end product of lipid oxidation caused by reactive oxygen species (ROS), and stroke risk in rural populations. We aimed to investigate whether blood MDA levels contribute to a higher stroke risk in a Chinese elderly population from rural areas. Data from 2011 to 2012 from the Chinese Longitudinal Healthy Longevity Survey (CLHLS), a national cohort of older adults in China, were analyzed. Smooth curve and multivariable correction analyses were used to evaluate the association between blood MDA levels and stroke risk in elderly populations from rural and urban areas, respectively. The median age of all included participants (N = 1598) was 84.04 years. The results of the smooth curve model revealed a gradual upward trend in the association of blood MDA levels with stroke risk in rural participants but not in urban participants. Similarly, the conditional logistic regression analysis suggested a significant association between MDA levels and stroke risk in rural participants but not in urban participants after adjustments for related confounding factors (age, sex, current smoker, current drinker, regular exercise, BMI and cardiovascular diseases (hypertension, heart disease, atrial fibrillation and diabetes)) were made. In brief, among the elderly population in China, elevated blood MDA levels were associated with increased stroke risk in rural participants but not in urban participants.
Collapse
Affiliation(s)
- Rong Wan
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yuhao Su
- Cardiovascular Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Meilan Zhu
- Rehabilitation Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ying Huang
- Rehabilitation Department, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
5
|
Yu M, Zhang M, Fu P, Wu M, Yin X, Chen Z. Research progress of mitophagy in chronic cerebral ischemia. Front Aging Neurosci 2023; 15:1224633. [PMID: 37600521 PMCID: PMC10434995 DOI: 10.3389/fnagi.2023.1224633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Chronic cerebral ischemia (CCI), a condition that can result in headaches, dizziness, cognitive decline, and stroke, is caused by a sustained decrease in cerebral blood flow. Statistics show that 70% of patients with CCI are aged > 80 years and approximately 30% are 45-50 years. The incidence of CCI tends to be lower, and treatment for CCI is urgent. Studies have confirmed that CCI can activate the corresponding mechanisms that lead to mitochondrial dysfunction, which, in turn, can induce mitophagy to maintain mitochondrial homeostasis. Simultaneously, mitochondrial dysfunction can aggravate the insufficient energy supply to cells and various diseases caused by CCI. Regulation of mitophagy has become a promising therapeutic target for the treatment of CCI. This article reviews the latest progress in the important role of mitophagy in CCI and discusses the induction pathways of mitophagy in CCI, including ATP synthesis disorder, oxidative stress injury, induction of reactive oxygen species, and Ca2+ homeostasis disorder, as well as the role of drugs in CCI by regulating mitophagy.
Collapse
Affiliation(s)
- Mayue Yu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Peijie Fu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Moxin Wu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
6
|
Büttiker P, Weissenberger S, Esch T, Anders M, Raboch J, Ptacek R, Kream RM, Stefano GB. Dysfunctional mitochondrial processes contribute to energy perturbations in the brain and neuropsychiatric symptoms. Front Pharmacol 2023; 13:1095923. [PMID: 36686690 PMCID: PMC9849387 DOI: 10.3389/fphar.2022.1095923] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
Mitochondria are complex endosymbionts that evolved from primordial purple nonsulfur bacteria. The incorporation of bacteria-derived mitochondria facilitates a more efficient and effective production of energy than what could be achieved based on previous processes alone. In this case, endosymbiosis has resulted in the seamless coupling of cytochrome c oxidase and F-ATPase to maximize energy production. However, this mechanism also results in the generation of reactive oxygen species (ROS), a phenomenon that can have both positive and negative ramifications on the host. Recent studies have revealed that neuropsychiatric disorders have a pro-inflammatory component in which ROS is capable of initiating damage and cognitive malfunction. Our current understanding of cognition suggests that it is the product of a neuronal network that consumes a substantial amount of energy. Thus, alterations or perturbations of mitochondrial function may alter not only brain energy supply and metabolite generation, but also thought processes and behavior. Mitochondrial abnormalities and oxidative stress have been implicated in several well-known psychiatric disorders, including schizophrenia (SCZ) and bipolar disorder (BPD). As cognition is highly energy-dependent, we propose that the neuronal pathways underlying maladaptive cognitive processing and psychiatric symptoms are most likely dependent on mitochondrial function, and thus involve brain energy translocation and the accumulation of the byproducts of oxidative stress. We also hypothesize that neuropsychiatric symptoms (e.g., disrupted emotional processing) may represent the vestiges of an ancient masked evolutionary response that can be used by both hosts and pathogens to promote self-repair and proliferation via parasitic and/or symbiotic pathways.
Collapse
Affiliation(s)
- Pascal Büttiker
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic, Prague, Czechia
| | - Simon Weissenberger
- Department of Psychology, University of New York in Prague, Czech Republic, Prague, Czechia
| | - Tobias Esch
- Institute for Integrative Health Care and Health Promotion, School of Medicine, Witten/Herdecke University, Witten, Germany
| | - Martin Anders
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic, Prague, Czechia
| | - Jiri Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic, Prague, Czechia
| | - Radek Ptacek
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic, Prague, Czechia
| | - Richard M. Kream
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic, Prague, Czechia
| | - George B. Stefano
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic, Prague, Czechia,*Correspondence: George B. Stefano,
| |
Collapse
|
7
|
Metabolic Features of Brain Function with Relevance to Clinical Features of Alzheimer and Parkinson Diseases. Molecules 2022; 27:molecules27030951. [PMID: 35164216 PMCID: PMC8839962 DOI: 10.3390/molecules27030951] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/04/2022] Open
Abstract
Brain metabolism is comprised in Alzheimer’s disease (AD) and Parkinson’s disease (PD). Since the brain primarily relies on metabolism of glucose, ketone bodies, and amino acids, aspects of these metabolic processes in these disorders—and particularly how these altered metabolic processes are related to oxidative and/or nitrosative stress and the resulting damaged targets—are reviewed in this paper. Greater understanding of the decreased functions in brain metabolism in AD and PD is posited to lead to potentially important therapeutic strategies to address both of these disorders, which cause relatively long-lasting decreased quality of life in patients.
Collapse
|
8
|
Nath AK, Dey SG. Simultaneous Binding of Heme and Cu to Amyloid β Peptides: Active Site and Reactivities. Dalton Trans 2022; 51:4986-4999. [DOI: 10.1039/d2dt00162d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amyloid imbalance and Aβ plaque formation are key histopathological features of Alzheimer’s disease (AD). These amyloid plaques observed in post-mortem AD brains have been found to contain increased levels of...
Collapse
|
9
|
Kim KH, Kim SJ, Singha S, Yang YJ, Park SK, Ahn KH. Ratiometric Detection of Hypochlorous Acid in Brain Tissues of Neuroinflammation and Maternal Immune Activation Models with a Deep-Red/Near-Infrared Emitting Probe. ACS Sens 2021; 6:3253-3261. [PMID: 34467757 DOI: 10.1021/acssensors.1c00930] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) produced by an inflammatory response in the brain are associated with various neurological disorders. To investigate ROS-associated neuroinflammatory diseases, fluorescent probes with practicality are in demand. We have investigated hypochlorous acid, an important ROS, in the brain tissues of neuroinflammation and maternal immune activation (MIA) model mice, using a new fluorescent probe. The probe has outstanding features over many known probes, such as providing two bright ratio signals in cells and tissues in deep-red/near-infrared wavelength regions with a large spectral separation, in addition to being strongly fluorescent, photo- and chemo-stable, highly selective and sensitive, fast responding, and biocompatible. We have found that the level of hypochlorous acid in the brain tissue of a neuroinflammatory mouse model was higher (2.7-4.0-fold) compared with that in normal brain tissue. Furthermore, the level of hypochlorous acid in the brain tissue of a MIA mouse model was higher (1.2-1.3-fold) compared with that in the normal brain tissue. The "robust" probe provides a practical tool for studying ROS-associated neurological disorders.
Collapse
Affiliation(s)
- Kyeong Hwan Kim
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic of Korea
| | - Soo Jeong Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic of Korea
| | - Subhankar Singha
- Institute of Advanced Studies and Research, JIS University, Kolkata 700091, India
| | - Yun Jae Yang
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic of Korea
| | - Kyo Han Ahn
- Department of Chemistry, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic of Korea
| |
Collapse
|
10
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and remyelination. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 11/09/2023] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
11
|
Rupnik M, Baker D, Selwood DL. Oligodendrocytes, BK channels and the preservation of myelin. F1000Res 2021; 10:781. [PMID: 34909188 PMCID: PMC8596180 DOI: 10.12688/f1000research.53422.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2021] [Indexed: 11/20/2022] Open
Abstract
Oligodendrocytes wrap multiple lamellae of their membrane, myelin, around axons of the central nervous system (CNS), to improve impulse conduction. Myelin synthesis is specialised and dynamic, responsive to local neuronal excitation. Subtle pathological insults are sufficient to cause significant neuronal metabolic impairment, so myelin preservation is necessary to safeguard neural networks. Multiple sclerosis (MS) is the most prevalent demyelinating disease of the CNS. In MS, inflammatory attacks against myelin, proposed to be autoimmune, cause myelin decay and oligodendrocyte loss, leaving neurons vulnerable. Current therapies target the prominent neuroinflammation but are mostly ineffective in protecting from neurodegeneration and the progressive neurological disability. People with MS have substantially higher levels of extracellular glutamate, the main excitatory neurotransmitter. This impairs cellular homeostasis to cause excitotoxic stress. Large conductance Ca2 +-activated K + channels (BK channels) could preserve myelin or allow its recovery by protecting cells from the resulting excessive excitability. This review evaluates the role of excitotoxic stress, myelination and BK channels in MS pathology, and explores the hypothesis that BK channel activation could be a therapeutic strategy to protect oligodendrocytes from excitotoxic stress in MS. This could reduce progression of neurological disability if used in parallel to immunomodulatory therapies.
Collapse
Affiliation(s)
- Maddalena Rupnik
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| | - David Baker
- Centre for Neuroscience and Trauma, Blizard Institute, Queen Mary University of London, London, WC1E 6BT, UK
| | - David L. Selwood
- Wolfson Insitute for Biomedical Research, University College London, London, WC1E 6BT, UK
| |
Collapse
|
12
|
Ren JX, Li C, Yan XL, Qu Y, Yang Y, Guo ZN. Crosstalk between Oxidative Stress and Ferroptosis/Oxytosis in Ischemic Stroke: Possible Targets and Molecular Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6643382. [PMID: 34055196 PMCID: PMC8133868 DOI: 10.1155/2021/6643382] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/23/2021] [Indexed: 01/21/2023]
Abstract
Oxidative stress is a key cause of ischemic stroke and an initiator of neuronal dysfunction and death, mainly through the overproduction of peroxides and the depletion of antioxidants. Ferroptosis/oxytosis is a unique, oxidative stress-induced cell death pathway characterized by lipid peroxidation and glutathione depletion. Both oxidative stress and ferroptosis/oxytosis have common molecular pathways. This review summarizes the possible targets and the mechanisms underlying the crosstalk between oxidative stress and ferroptosis/oxytosis in ischemic stroke. This knowledge might help to further understand the pathophysiology of ischemic stroke and open new perspectives for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jia-Xin Ren
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
- China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
| | - Chao Li
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
- China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
| | - Xiu-Li Yan
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
- China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
| | - Yang Qu
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
- China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
| | - Yi Yang
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
- China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, No. 1 Xinmin Street, Changchun 130021, China
| | - Zhen-Ni Guo
- Stroke Center & Clinical Trial and Research Center for Stroke, Department of Neurology, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
- China National Comprehensive Stroke Center, No. 1 Xinmin Street, Changchun 130021, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, No. 1 Xinmin Street, Changchun 130021, China
| |
Collapse
|
13
|
Faborode OS, Yusuf IO, Okpe PO, Okudaje AO, Onasanwo SA. Exposure to prolonged unpredictable light impairs spatial memory via induction of oxidative stress and tumor necrosis factor-alpha in rats. J Basic Clin Physiol Pharmacol 2021; 33:355-362. [PMID: 33873260 DOI: 10.1515/jbcpp-2020-0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 01/23/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The human body physiology rapidly changes and adapt to several environmental stimuli, including light. Abnormal artificial light exposures have been shown to affect sleep cycle, cognition, and mood. Although studies have reported inconsistent effects of short-term or constant long-term light exposures, human exposures to artificial lights occur at varying, unpredictable times and duration daily. Here, we studied the effects of long-term unpredictable light exposure on learning, memory, oxidative status, and associated cytokines in rats. METHODS Artificial lighting was provided using an array of white light-emitting diodes coupled to a microcontroller that switches them on or off at unpredictable times and duration (light intensity = 200 ± 20 lx). Within the last eight days of 40 days exposure, animals were subjected to open field test, Morris water maze, and novel object recognition behavioral paradigms. Brain levels of malondialdehyde (MDA), superoxide dismutase (SOD), catalase, reduced glutathione (GSH), glutathione S-transferase (GST), tumor necrosis factor-alpha (TNF-α), and vascular endothelial growth factor (VEGF) were assayed. RESULTS Exposed rats showed impaired spatial learning and memory (p<0.05), but no changes in object recognition memory or locomotor activity. Oxidative stress analyses also revealed significant changes in the concentrations of MDA, SOD, catalase, and GSH levels (p<0.05), not GST. Similarly, there was an increased TNF-α expression (p<0.05), not VEGF. CONCLUSIONS We conclude that oxidative stress is involved in memory impairment in rats exposed to prolonged unpredictable lights, which again suggests the detrimental effects of extended light exposure on the nervous system.
Collapse
Affiliation(s)
- Oluwaseun S Faborode
- Department of Human Physiology, College of Health Sciences, Bingham University, Karu, Nigeria.,Neuroscience and Oral Physiology Unit, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| | - Issa O Yusuf
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester,MA, USA
| | - Paschal O Okpe
- Department of Human Physiology, College of Health Sciences, Bingham University, Karu, Nigeria
| | - Ann O Okudaje
- Department of Human Physiology, College of Health Sciences, Bingham University, Karu, Nigeria
| | - Samuel A Onasanwo
- Neuroscience and Oral Physiology Unit, Department of Physiology, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
14
|
Mitochondria focused neurotherapeutics for spinal cord injury. Exp Neurol 2020; 330:113332. [DOI: 10.1016/j.expneurol.2020.113332] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/21/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023]
|
15
|
Trigiani LJ, Lacalle-Aurioles M, Bourourou M, Li L, Greenhalgh AD, Zarruk JG, David S, Fehlings MG, Hamel E. Benefits of physical exercise on cognition and glial white matter pathology in a mouse model of vascular cognitive impairment and dementia. Glia 2020; 68:1925-1940. [PMID: 32154952 DOI: 10.1002/glia.23815] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/06/2020] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
White matter (WM) pathology is a clinically predictive feature of vascular cognitive impairment and dementia (VCID). Mice overexpressing transforming growth factor-β1 (TGF) with an underlying cerebrovascular pathology when fed a high cholesterol diet (HCD) develop cognitive deficits (VCID mice) that we recently found could be prevented by physical exercise (EX). Here, we further investigated cognitive and WM pathology in VCID mice and examined the cellular substrates of the protective effects of moderate aerobic EX focusing on WM alterations. Six groups were studied: Wild-type (WT) and TGF mice (n = 20-24/group) fed standard lab chow or a 2% HCD, with two HCD-fed groups given concurrent access to running wheels. HCD had a significant negative effect in TGF mice that was prevented by EX on working and object recognition memory, the latter also altered in WT HCD mice. Whisker-evoked increases in cerebral blood flow (CBF) were reduced in HCD-fed mice, deficits that were countered by EX, and baseline WM CBF was similarly affected. VCID mice displayed WM functional deficits characterized by lower compound action potential amplitude not found in EX groups. Moreover, there was an increased number of collapsing capillaries, galectin-3-expressing microglial cells, as well as a reduced number of oligodendrocytes in the WM of VCID mice; all of which were prevented by EX. Our findings indicate that a compromised cerebral circulation precedes reduced WM vascularization, enhanced WM inflammation and impaired oligodendrogenesis that all likely account for the increased susceptibility to memory impairments in VCID mice, which can be prevented by EX. MAIN POINTS: A compromised cerebral circulation increases susceptibility to anatomical and functional white matter changes that develop alongside cognitive deficits when challenged with a high cholesterol diet; preventable by a translational regimen of exercise.
Collapse
Affiliation(s)
- Lianne J Trigiani
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - María Lacalle-Aurioles
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Miled Bourourou
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Lijun Li
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Andrew D Greenhalgh
- Center for Research in Neuroscience, The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Juan G Zarruk
- Center for Research in Neuroscience, The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Samuel David
- Center for Research in Neuroscience, The Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
16
|
Langley MR, Yoon H, Kim HN, Choi CI, Simon W, Kleppe L, Lanza IR, LeBrasseur NK, Matveyenko A, Scarisbrick IA. High fat diet consumption results in mitochondrial dysfunction, oxidative stress, and oligodendrocyte loss in the central nervous system. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165630. [PMID: 31816440 PMCID: PMC7982965 DOI: 10.1016/j.bbadis.2019.165630] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/14/2019] [Accepted: 12/02/2019] [Indexed: 02/07/2023]
Abstract
Metabolic syndrome is a key risk factor and co-morbidity in multiple sclerosis (MS) and other neurological conditions, such that a better understanding of how a high fat diet contributes to oligodendrocyte loss and the capacity for myelin regeneration has the potential to highlight new treatment targets. Results demonstrate that modeling metabolic dysfunction in mice with chronic high fat diet (HFD) consumption promotes loss of oligodendrocyte progenitors across the brain and spinal cord. A number of transcriptomic and metabolomic changes in ER stress, mitochondrial dysfunction, and oxidative stress pathways in HFD-fed mouse spinal cords were also identified. Moreover, deficits in TCA cycle intermediates and mitochondrial respiration were observed in the chronic HFD spinal cord tissue. Oligodendrocytes are known to be particularly vulnerable to oxidative damage, and we observed increased markers of oxidative stress in both the brain and spinal cord of HFD-fed mice. We additionally identified that increased apoptotic cell death signaling is underway in oligodendrocytes from mice chronically fed a HFD. When cultured under high saturated fat conditions, oligodendrocytes decreased both mitochondrial function and differentiation. Overall, our findings show that HFD-related changes in metabolic regulators, decreased mitochondrial function, and oxidative stress contribute to a loss of myelinating cells. These studies identify HFD consumption as a key modifiable lifestyle factor for improved myelin integrity in the adult central nervous system and in addition new tractable metabolic targets for myelin protection and repair strategies.
Collapse
Affiliation(s)
- Monica R Langley
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Hyesook Yoon
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Ha Neui Kim
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Chan-Il Choi
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Whitney Simon
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Laurel Kleppe
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA
| | - Ian R Lanza
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Nathan K LeBrasseur
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Isobel A Scarisbrick
- Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research Center, Mayo Clinic, Rochester, MN 55905, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
17
|
Ji P, B Nonnecke E, Doan N, Lönnerdal B, Tan B. Excess Iron Enhances Purine Catabolism Through Activation of Xanthine Oxidase and Impairs Myelination in the Hippocampus of Nursing Piglets. J Nutr 2019; 149:1911-1919. [PMID: 31373370 DOI: 10.1093/jn/nxz166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/17/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Few studies have addressed the risk of nutritional iron overexposure in infancy. We previously found that excess dietary iron in nursing piglets resulted in iron overload in the liver and hippocampus and diminished socialization with novel conspecifics in a test for social novelty preference. OBJECTIVES This experiment aimed to identify metabolites and metabolic pathways affected by iron overload in the liver and hippocampus of nursing piglets. METHODS Liver and hippocampal tissues collected from 22-d-old piglets (Hampshire × Yorkshire crossbreed; 5.28 ± 0.53 kg body weight; 50% male) that received orally 0 (NI group) or 50 mg iron/(d · kg body weight) (HI group) from postnatal day (PD) 2 to PD21 were analyzed for mRNA and protein expression and enzyme activity of xanthine oxidase (XO). Untargeted metabolomics was performed using GC-MS. Expression of myelin basic protein (MBP) in the hippocampus was determined using western blot. RESULTS There were 108 and 126 metabolites identified in the hippocampus and liver, respectively. Compared with NI, HI altered 15 metabolites (P < 0.05, q < 0.2) in the hippocampus, including a reduction in myo-inositol (0.86-fold) and N-acetylaspartic acid (0.84-fold), 2 metabolites important for neuronal function and myelination. Seven metabolites involved in purine and pyrimidine metabolism (e.g., hypoxanthine, xanthine, and β-alanine) were coordinately changed in the hippocampus (P < 0.05, q < 0.2), suggesting that iron excess enhanced purine catabolism. The mRNA expression (2.3-fold) (P < 0.05) and activity of XO, a rate-limiting enzyme in purine degradation, was increased. Excess iron increased hippocampal lipid peroxidation by 74% (P < 0.05) and decreased MBP by 44% (P = 0.053). The hepatic metabolome was unaffected. CONCLUSIONS In nursing piglets, excess iron enhances hippocampal purine degradation through activation of XO, which may induce oxidative stress and alter energy metabolism in the developing brain.
Collapse
Affiliation(s)
- Peng Ji
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Eric B Nonnecke
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Nicole Doan
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, USA
| | - Bie Tan
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| |
Collapse
|
18
|
Hill RL, Singh IN, Wang JA, Hall ED. Effects of Phenelzine Administration on Mitochondrial Function, Calcium Handling, and Cytoskeletal Degradation after Experimental Traumatic Brain Injury. J Neurotrauma 2019; 36:1231-1251. [PMID: 30358485 PMCID: PMC6479250 DOI: 10.1089/neu.2018.5946] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) results in the production of peroxynitrite (PN), leading to oxidative damage of lipids and protein. PN-mediated lipid peroxidation (LP) results in production of reactive aldehydes 4-hydroxynonenal (4-HNE) and acrolein. The goal of these studies was to explore the hypothesis that interrupting secondary oxidative damage following a TBI via phenelzine (PZ), analdehyde scavenger, would protect against LP-mediated mitochondrial and neuronal damage. Male Sprague-Dawley rats received a severe (2.2 mm) controlled cortical impact (CCI)-TBI. PZ was administered subcutaneously (s.c.) at 15 min (10 mg/kg) and 12 h (5 mg/kg) post-injury and for the therapeutic window/delay study, PZ was administered at 1 h (10 mg/kg) and 24 h (5 mg/kg). Mitochondrial and cellular protein samples were obtained at 24 and 72 h post-injury (hpi). Administration of PZ significantly improved mitochondrial respiration at 24 and 72 h compared with vehicle-treated animals. These results demonstrate that PZ administration preserves mitochondrial bioenergetics at 24 h and that this protection is maintained out to 72 hpi. Additionally, delaying the administration still elicited significant protective effects. PZ administration also improved mitochondrial Ca2+ buffering (CB) capacity and mitochondrial membrane potential parameters compared with vehicle-treated animals at 24 h. Although PZ treatment attenuated aldehyde accumulation post-injury, the effects were insignificant. The amount of α-spectrin breakdown in cortical tissue was reduced by PZ administration at 24 h, but not at 72 hpi compared with vehicle-treated animals. In conclusion, these results indicate that acute PZ treatment successfully attenuates LP-mediated oxidative damage eliciting multiple neuroprotective effects following TBI.
Collapse
Affiliation(s)
- Rachel L. Hill
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky
| | - Indrapal N. Singh
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Juan A. Wang
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky
| | - Edward D. Hall
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, Kentucky
| |
Collapse
|
19
|
Foliaki ST, Lewis V, Islam AMT, Ellett LJ, Senesi M, Finkelstein DI, Roberts B, Lawson VA, Adlard PA, Collins SJ. Early existence and biochemical evolution characterise acutely synaptotoxic PrPSc. PLoS Pathog 2019; 15:e1007712. [PMID: 30970042 PMCID: PMC6490942 DOI: 10.1371/journal.ppat.1007712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 04/30/2019] [Accepted: 03/18/2019] [Indexed: 11/19/2022] Open
Abstract
Although considerable evidence supports that misfolded prion protein (PrPSc) is the principal component of “prions”, underpinning both transmissibility and neurotoxicity, clear consensus around a number of fundamental aspects of pathogenesis has not been achieved, including the time of appearance of neurotoxic species during disease evolution. Utilizing a recently reported electrophysiology paradigm, we assessed the acute synaptotoxicity of ex vivo PrPSc prepared as crude homogenates from brains of M1000 infected wild-type mice (cM1000) harvested at time-points representing 30%, 50%, 70% and 100% of the terminal stage of disease (TSD). Acute synaptotoxicity was assessed by measuring the capacity of cM1000 to impair hippocampal CA1 region long-term potentiation (LTP) and post-tetanic potentiation (PTP) in explant slices. Of particular note, cM1000 from 30% of the TSD was able to cause significant impairment of LTP and PTP, with the induced failure of LTP increasing over subsequent time-points while the capacity of cM1000 to induce PTP failure appeared maximal even at this early stage of disease progression. Evidence that the synaptotoxicity directly related to PrP species was demonstrated by the significant rescue of LTP dysfunction at each time-point through immuno-depletion of >50% of total PrP species from cM1000 preparations. Moreover, similar to our previous observations at the terminal stage of M1000 prion disease, size fractionation chromatography revealed that capacity for acute synpatotoxicity correlated with predominance of oligomeric PrP species in infected brains across all time points, with the profile appearing maximised by 50% of the TSD. Using enhanced sensitivity western blotting, modestly proteinase K (PK)-resistant PrPSc was detectable at very low levels in cM1000 at 30% of the TSD, becoming robustly detectable by 70% of the TSD at which time substantial levels of highly PK-resistant PrPSc was also evident. Further illustrating the biochemical evolution of acutely synaptotoxic species the synaptotoxicity of cM1000 from 30%, 50% and 70% of the TSD, but not at 100% TSD, was abolished by digestion of immuno-captured PrP species with mild PK treatment (5μg/ml for an hour at 37°C), demonstrating that the predominant synaptotoxic PrPSc species up to and including 70% of the TSD were proteinase-sensitive. Overall, these findings in combination with our previous assessments of transmitting prions support that synaptotoxic and infectious M1000 PrPSc species co-exist from at least 30% of the TSD, simultaneously increasing thereafter, albeit with eventual plateauing of transmitting conformers. Although evidence clearly supports that misfolded prion protein (PrPSc) is the principal component of “prions”, underpinning both transmissibility and neurotoxicity, consensus is lacking around the time of appearance and biochemical profile of neurotoxic species during disease evolution. Employing an electrophysiology model, measuring the capacity of brain homogenates derived from across the disease time-course to impair CA1 region long-term potentiation (LTP) and post-tetanic potentiation (PTP) in hippocampal slices, we observed that synaptotoxic species were present from 30% of the terminal stage of disease (TSD). Evidence that synaptotoxicity directly related to PrP species was demonstrated by significant rescue of LTP dysfunction at each time-point through immuno-depleting >~50% of total PrP species from cM1000 preparations. Moreover, size fractionation chromatography revealed that acute synpatotoxicity correlated with predominance of oligomeric PrP species in infected brains across all time points, while additional characterisation of cM1000 demonstrated that the predominant synaptotoxic PrPSc species up to and including 70% of the TSD were quite proteinase-sensitive. These findings in combination with our previous assessments of transmitting prions support that synaptotoxic and infectious M1000 PrPSc species co-exist from at least 30% of the TSD, simultaneously increasing thereafter, with biochemical transformation of synaptotoxic conformers continuing until late in disease.
Collapse
Affiliation(s)
- Simote Totauhelotu Foliaki
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
| | - Victoria Lewis
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | | | - Laura Jane Ellett
- Department of Pathology The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | - Matteo Senesi
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
| | | | - Blaine Roberts
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Victoria A. Lawson
- Department of Pathology The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Anthony Adlard
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Steven John Collins
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
20
|
Evaluation of Pathological Association between Stroke-Related QTL and Salt-Induced Renal Injury in Stroke-Prone Spontaneously Hypertensive Rat. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5049746. [PMID: 30792992 PMCID: PMC6354164 DOI: 10.1155/2019/5049746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/28/2018] [Indexed: 01/13/2023]
Abstract
The stroke-prone spontaneously hypertensive rat (SHRSP) suffers from severe hypertension and hypertensive organ damage such as cerebral stroke and kidney injury under salt-loading. By a quantitative trait locus (QTL) analysis between SHRSP and SHR (the stroke-resistant parental strain of SHRSP), two major QTLs for stroke susceptibility were identified on chromosomes 1 and 18 of SHRSP, which were confirmed in congenic strains constructed between SHRSP and SHR. As the progression of renal dysfunction was suggested to be one of the key factors inducing stroke in SHRSP, we examined effects of the stroke-related QTLs on kidney injury using two congenic strains harboring either of SHRSP-derived fragments of chromosomes 1 and 18 in the SHR genome. The congenic strains were challenged with 1% NaCl solution for 4 weeks; measurement of systolic blood pressure and urinary isoprostane level (a marker for oxidative stress) and evaluation of renal injury by quantification of genetic marker expression and histological examination were performed. We found that the congenic rats with SHRSP-derived fragment of chromosome 18 showed more severe renal damage with higher expression of Col1α-1 (a genetic marker for renal fibrosis) and higher urinary isoprostane level. In contrast, the fragment of chromosome 1 from SHRSP did not give such effects on SHR. Blood pressure was not greater in either of the congenic strains when compared with SHR. We concluded that the QTL region on chromosome 18 might deteriorate salt-induced renal injury in SHR through a blood pressure-independent mechanism.
Collapse
|
21
|
Zhao XY, Lu MH, Yuan DJ, Xu DE, Yao PP, Ji WL, Chen H, Liu WL, Yan CX, Xia YY, Li S, Tao J, Ma QH. Mitochondrial Dysfunction in Neural Injury. Front Neurosci 2019; 13:30. [PMID: 30778282 PMCID: PMC6369908 DOI: 10.3389/fnins.2019.00030] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are the double membrane organelles providing most of the energy for cells. In addition, mitochondria also play essential roles in various cellular biological processes such as calcium signaling, apoptosis, ROS generation, cell growth, and cell cycle. Mitochondrial dysfunction is observed in various neurological disorders which harbor acute and chronic neural injury such as neurodegenerative diseases and ischemia, hypoxia-induced brain injury. In this review, we describe how mitochondrial dysfunction contributes to the pathogenesis of neurological disorders which manifest chronic or acute neural injury.
Collapse
Affiliation(s)
- Xiu-Yun Zhao
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Mei-Hong Lu
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - De-Juan Yuan
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases, Dalian Medical University, Dalian, China
| | - De-En Xu
- Wuxi No. 2 People’s Hospital, Wuxi, China
| | - Pei-Pei Yao
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wen-Li Ji
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong Chen
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wen-Long Liu
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen-Xiao Yan
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi-Yuan Xia
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shao Li
- Department of Physiology, Liaoning Provincial Key Laboratory of Cerebral Diseases, Dalian Medical University, Dalian, China
| | - Jin Tao
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
- Department of Physiology and Neurobiology and Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou, China
| | - Quan-Hong Ma
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
22
|
Neuroprotective Effect and Mechanism of Action of Tetramethylpyrazine Nitrone for Ischemic Stroke Therapy. Neuromolecular Med 2018; 20:97-111. [DOI: 10.1007/s12017-018-8478-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 01/18/2018] [Indexed: 10/18/2022]
|
23
|
Zarkovic K, Jakovcevic A, Zarkovic N. Contribution of the HNE-immunohistochemistry to modern pathological concepts of major human diseases. Free Radic Biol Med 2017; 111:110-126. [PMID: 27993730 DOI: 10.1016/j.freeradbiomed.2016.12.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/05/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
Abstract
Excessive production of reactive oxygen species can induce peroxidation of the polyunsaturated fatty acids thus generating reactive aldehydes like 4-hydroxy-2-nonenal (HNE), denoted as "the second messenger of free radicals". Because HNE has high binding affinity for cysteine, histidine and lysine it forms relatively stable and hardly metabolized protein adducts. By changing structure and function of diverse structural and regulatory proteins, HNE achieves not only cytotoxic, but also regulatory functions in various pathophysiological processes. Numerous animal model studies and clinical trials confirmed HNE as one of the crucial factors in development and progression of many disorders, in particular of cancer, (neuro)degenerative, metabolic and inflammatory diseases. Since HNE has multiple biological effects and is in the living system usually bound to proteins and peptides, many research groups work on development of specific immunochemical methods targeting the HNE-histidine adducts as major bioactive marker of lipid peroxidation, following the research pathway initiated by Hermann Esterbauer, who discovered HNE in 60's. Such immunohistochemical studies did not only prove the high biomedical importance of HNE, but have also given new insights into major diseases of the modern man. Immunohistochemical studies have shown reversibility of formation of the HNE-protein adducts, as well as differential onset of the HNE-mediated lipid peroxidation between age- associated atherosclerosis and photoaging, revealing eventually selective anti-cancer effects of HNE produced by non-malignant cells in vicinity of cancer. This review summarizes some of the HNE-histidine immunohistochemistry findings we believe are of broad biomedical interest and could inspire new studies in the field.
Collapse
Affiliation(s)
- Kamelija Zarkovic
- University of Zagreb, School of Medicine, Clinical Hospital Centre Zagreb, Croatia.
| | - Antonia Jakovcevic
- University of Zagreb, School of Medicine, Clinical Hospital Centre Zagreb, Croatia
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Zagreb, Croatia
| |
Collapse
|
24
|
O'Hare Doig RL, Chiha W, Giacci MK, Yates NJ, Bartlett CA, Smith NM, Hodgetts SI, Harvey AR, Fitzgerald M. Specific ion channels contribute to key elements of pathology during secondary degeneration following neurotrauma. BMC Neurosci 2017; 18:62. [PMID: 28806920 PMCID: PMC5557315 DOI: 10.1186/s12868-017-0380-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 08/05/2017] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Following partial injury to the central nervous system, cells beyond the initial injury site undergo secondary degeneration, exacerbating loss of neurons, compact myelin and function. Changes in Ca2+ flux are associated with metabolic and structural changes, but it is not yet clear how flux through specific ion channels contributes to the various pathologies. Here, partial optic nerve transection in adult female rats was used to model secondary degeneration. Treatment with combinations of three ion channel inhibitors was used as a tool to investigate which elements of oxidative and structural damage related to long term functional outcomes. The inhibitors employed were the voltage gated Ca2+ channel inhibitor Lomerizine (Lom), the Ca2+ permeable AMPA receptor inhibitor YM872 and the P2X7 receptor inhibitor oxATP. RESULTS Following partial optic nerve transection, hyper-phosphorylation of Tau and acetylated tubulin immunoreactivity were increased, and Nogo-A immunoreactivity was decreased, indicating that axonal changes occurred acutely. All combinations of ion channel inhibitors reduced hyper-phosphorylation of Tau and increased Nogo-A immunoreactivity at day 3 after injury. However, only Lom/oxATP or all three inhibitors in combination significantly reduced acetylated tubulin immunoreactivity. Most combinations of ion channel inhibitors were effective in restoring the lengths of the paranode and the paranodal gap, indicative of the length of the node of Ranvier, following injury. However, only all three inhibitors in combination restored to normal Ankyrin G length at the node of Ranvier. Similarly, HNE immunoreactivity and loss of oligodendrocyte precursor cells were only limited by treatment with all three ion channel inhibitors in combination. CONCLUSIONS Data indicate that inhibiting any of a range of ion channels preserves certain elements of axon and node structure and limits some oxidative damage following injury, whereas ionic flux through all three channels must be inhibited to prevent lipid peroxidation and preserve Ankyrin G distribution and OPCs.
Collapse
Affiliation(s)
- Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia.,Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Wissam Chiha
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia.,Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Marcus K Giacci
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Nathanael J Yates
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Nicole M Smith
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia.,Experimental and Regenerative Neurosciences, School of Chemistry and Biochemistry, The University of Western Australia, Crawley, WA, 6009, Australia
| | - Stuart I Hodgetts
- Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia.,Perron Institute for Neurological and Translational Science, Verdun St, Nedlands, WA, 6009, Australia
| | - Alan R Harvey
- Experimental and Regenerative Neurosciences, School of Human Sciences, The University of Western Australia, Crawley, WA, 6009, Australia.,Perron Institute for Neurological and Translational Science, Verdun St, Nedlands, WA, 6009, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Biological Sciences, The University of Western Australia, Crawley, WA, 6009, Australia. .,Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia. .,Perron Institute for Neurological and Translational Science, Verdun St, Nedlands, WA, 6009, Australia.
| |
Collapse
|
25
|
Ćurko-Cofek B, Grubić Kezele T, Barac-Latas V. Hepcidin and metallothioneins as molecular base for sex-dependent differences in clinical course of experimental autoimmune encephalomyelitis in chronic iron overload. Med Hypotheses 2017; 107:51-54. [PMID: 28915963 DOI: 10.1016/j.mehy.2017.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 07/12/2017] [Accepted: 07/15/2017] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis is a chronic demyelinating disease of the central nervous system characterised by inflammatory and degenerative changes. It is considered that disease arises from the influence of environmental factors on genetically susceptible individuals. Recent researches, using magnetic resonance imaging, connected iron deposits in different brain regions with demyelinating process in multiple sclerosis patients. Although iron is an essential trace element important for many biological functions it could be harmful because iron excess can induce the production of reactive oxygen species, development of oxidative stress and lipid peroxidation which leads to demyelination. In experimental autoimmune encephalomyelitis model, the most common experimental animal model for multiple sclerosis, we recently found that chronic iron overload influences the clinical course of disease in Dark Agouti rats. In female rats iron overload accelerated the onset of disease, while in male rats it accelerated the progression of disease and increased mortality rate. We hypothesize that those differences arise on molecular level in different expression of stress response proteins hepcidin and metallothioneins in male and female iron overloaded rats. They are both upregulated by metal ions in both sexes. Hepcidin is additionally upregulated by estrogen in female rats and therefore causes higher degradation of iron exporter ferroportin and sequestration of iron in the cells, lowering the possibility for the development of oxidative stress. Antioxidative effect of metallothioneins could be increased in female rats because of their ability to reversibly exchange metal ions with the estrogen receptor. In case of iron excess metallothioneins release zinc, which is normally bound to them. Zinc binds to estrogen receptor and leaves metallothioneins binding domains free for iron, causing at least provisional cytoprotective effect. To test this hypothesis, we propose to determine and compare serum levels of hepcidin and estrogen using ELISA essay as well as expression and distribution of acute stress response proteins hepcidin and metallothioneins, iron and estrogen receptor in the brain and spinal cord tissue using immunohistochemistry in control and chronic iron overloaded male and female rats in experimental autoimmune encephalomyelitis model. It would be also possible to perform the same immunohistochemistry in the brain tissue of multiple sclerosis patients post mortem. The results of experiments could contribute to better understanding of cytoprotective mechanisms in chronic iron overload that could have possible therapeutic applications in iron disturbances. In order to elucidate whether common measure of systemic iron status, like ferritin, haemoglobin concentration and transferrin saturation levels, may be used to distinguish physiologic from potentially harmful iron levels in local disease, for example multiple sclerosis and Still's disease, well-designed clinical trials would be of great interest.
Collapse
Affiliation(s)
- Božena Ćurko-Cofek
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia.
| | - Tanja Grubić Kezele
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Vesna Barac-Latas
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| |
Collapse
|
26
|
Ng GJ, Quek AM, Cheung C, Arumugam TV, Seet RC. Stroke biomarkers in clinical practice: A critical appraisal. Neurochem Int 2017; 107:11-22. [DOI: 10.1016/j.neuint.2017.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/05/2017] [Accepted: 01/08/2017] [Indexed: 02/04/2023]
|
27
|
Neurodegeneration and Glial Response after Acute Striatal Stroke: Histological Basis for Neuroprotective Studies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3173564. [PMID: 28090244 PMCID: PMC5165163 DOI: 10.1155/2016/3173564] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/26/2016] [Accepted: 10/12/2016] [Indexed: 11/24/2022]
Abstract
Stroke is a leading cause of death and neurological disability worldwide and striatal ischemic stroke is frequent in humans due to obstruction of middle cerebral artery. Several pathological events underlie damage progression and a comprehensive description of the pathological features following experimental stroke in both acute and chronic survival times is a necessary step for further functional studies. Here, we explored the patterns of microglial activation, astrocytosis, oligodendrocyte damage, myelin impairment, and Nogo-A immunoreactivity between 3 and 30 postlesion days (PLDs) after experimental striatal stroke in adult rats induced by microinjections of endothelin-1 (ET-1). The focal ischemia induced tissue loss concomitant with intense microglia activation between 3 and 14 PLDs (maximum at 7 PLDs), decreasing afterward. Astrocytosis was maximum around 7 PLDs. Oligodendrocyte damage and Nogo-A upregulation were higher at 3 PLDs. Myelin impairment was maximum between 7 and 14 PLDs. Nogo-A expression was higher in the first week in comparison to control. The results add important histopathological features of ET-1 induced stroke in subacute and chronic survival times. In addition, the establishment of the temporal evolution of these neuropathological events is an important step for future studies seeking suitable neuroprotective drugs targeting neuroinflammation and white matter damage.
Collapse
|
28
|
Chacko BK, Wall SB, Kramer PA, Ravi S, Mitchell T, Johnson MS, Wilson L, Barnes S, Landar A, Darley-Usmar VM. Pleiotropic effects of 4-hydroxynonenal on oxidative burst and phagocytosis in neutrophils. Redox Biol 2016; 9:57-66. [PMID: 27393890 PMCID: PMC4939321 DOI: 10.1016/j.redox.2016.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 01/09/2023] Open
Abstract
Metabolic control of cellular function is significant in the context of inflammation-induced metabolic dysregulation in immune cells. Generation of reactive oxygen species (ROS) such as hydrogen peroxide and superoxide are one of the critical events that modulate the immune response in neutrophils. When activated, neutrophil NADPH oxidases consume large quantities of oxygen to rapidly generate ROS, a process that is referred to as the oxidative burst. These ROS are required for the efficient removal of phagocytized cellular debris and pathogens. In chronic inflammatory diseases, neutrophils are exposed to increased levels of oxidants and pro-inflammatory cytokines that can further prime oxidative burst responses and generate lipid oxidation products such as 4-hydroxynonenal (4-HNE). In this study we hypothesized that since 4-HNE can target glycolysis then this could modify the oxidative burst. To address this the oxidative burst was determined in freshly isolated healthy subject neutrophils using 13-phorbol myristate acetate (PMA) and the extracellular flux analyzer. Neutrophils pretreated with 4-HNE exhibited a significant decrease in the oxidative burst response and phagocytosis. Mass spectrometric analysis of alkyne-HNE treated neutrophils followed by click chemistry detected modification of a number of cytoskeletal, metabolic, redox and signaling proteins that are critical for the NADPH oxidase mediated oxidative burst. These modifications were confirmed using a candidate immunoblot approach for critical proteins of the active NADPH oxidase enzyme complex (Nox2 gp91phox subunit and Rac1 of the NADPH oxidase) and glyceraldehyde phosphate dehydrogenase, a critical enzyme in the metabolic regulation of oxidative burst. Taken together, these data suggest that 4-HNE-induces a pleiotropic mechanism to inhibit neutrophil function. These mechanisms may contribute to the immune dysregulation associated with chronic pathological conditions where 4-HNE is generated. Phagocytosis and glycolysis are inhibited in neutrophils by 4-hydroxynonenal. Click chemistry with alkyne-HNE identifies over 100 potential protein targets. Rac1, NOX2 and GAPDH are modified by 4-HNE. The 4-HNE-dependent inhibition of neutrophil function is mediated by a pleiotropic mechanism.
Collapse
Affiliation(s)
- Balu K Chacko
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Stephanie B Wall
- Department of Pathology, University of Alabama at Birmingham, United States
| | - Philip A Kramer
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Saranya Ravi
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Tanecia Mitchell
- Department of Urology, University of Alabama at Birmingham, United States
| | - Michelle S Johnson
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States
| | - Landon Wilson
- Department of Pharmacology and Toxicology, The Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, United States
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, The Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham, United States
| | - Aimee Landar
- Department of Pathology, University of Alabama at Birmingham, United States
| | - Victor M Darley-Usmar
- Mitochondrial Medicine Laboratory, University of Alabama at Birmingham, United States; Department of Pathology, University of Alabama at Birmingham, United States.
| |
Collapse
|
29
|
Fan F, Ge Y, Lv W, Elliott MR, Muroya Y, Hirata T, Booz GW, Roman RJ. Molecular mechanisms and cell signaling of 20-hydroxyeicosatetraenoic acid in vascular pathophysiology. Front Biosci (Landmark Ed) 2016; 21:1427-63. [PMID: 27100515 DOI: 10.2741/4465] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). 20-HETE is a vasoconstrictor that depolarizes vascular smooth muscle cells by blocking K+ channels. EETs serve as endothelial derived hyperpolarizing factors. Inhibition of the formation of 20-HETE impairs the myogenic response and autoregulation of renal and cerebral blood flow. Changes in the formation of EETs and 20-HETE have been reported in hypertension and drugs that target these pathways alter blood pressure in animal models. Sequence variants in CYP4A11 and CYP4F2 that produce 20-HETE, UDP-glucuronosyl transferase involved in the biotransformation of 20-HETE and soluble epoxide hydrolase that inactivates EETs are associated with hypertension in human studies. 20-HETE contributes to the regulation of vascular hypertrophy, restenosis, angiogenesis and inflammation. It also promotes endothelial dysfunction and contributes to cerebral vasospasm and ischemia-reperfusion injury in the brain, kidney and heart. This review will focus on the role of 20-HETE in vascular dysfunction, inflammation, ischemic and hemorrhagic stroke and cardiac and renal ischemia reperfusion injury.
Collapse
Affiliation(s)
- Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Ying Ge
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216 and Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Matthew R Elliott
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Yoshikazu Muroya
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216 and Department of General Medicine and Rehabilitation, Tohoku Medical and Pharmaceutical University School of Medicine, Sendai, Japan
| | - Takashi Hirata
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216 and Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216,
| |
Collapse
|
30
|
Gaman AM, Uzoni A, Popa-Wagner A, Andrei A, Petcu EB. The Role of Oxidative Stress in Etiopathogenesis of Chemotherapy Induced Cognitive Impairment (CICI)-"Chemobrain". Aging Dis 2016; 7:307-317. [PMID: 27330845 PMCID: PMC4898927 DOI: 10.14336/ad.2015.1022] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/22/2015] [Indexed: 11/29/2022] Open
Abstract
Chemobrain or chemotherapy induced cognitive impairment (CICI) represents a new clinical syndrome characterised by memory, learning and motor function impairment. As numerous patients with cancer are long-term survivors, CICI represent a significant factor which may interfere with their quality of life. However, this entity CICI must be distinguished from other cognitive syndromes and addressed accordingly. At the present time, experimental and clinical research suggests that CICI could be induced by numerous factors including oxidative stress. This type of CNS injury has been previously described in cancer patients treated with common anti-neoplastic drugs such as doxorubicine, carmustine, methotrexate and cyclophosphamide. It seems that all these pharmacological factors promote neuronal death through a final common pathway represented by TNF alpha (tumour necrosis factor). However, as cancer in general is diagnosed more commonly in the aging population, the elderly oncological patient must be treated with great care since aging per se is also impacted by oxidative stress and potentiually by TNF alpha deleterious action on brain parenchyma. In this context, some patients may develop cognitive dysfunction well before the appearance of CICI. In addition, chemotherapy may worsen their cognitive function. Therefore, at the present time, there is an acute need for development of effective therapeutic methods to prevent CICI as well as new methods of early CICI diagnosis.
Collapse
Affiliation(s)
- Amelia Maria Gaman
- Research Center of Experimental and Clinical Medicine, University of Medicine and Pharmacy of Craiova, 200349, Romania
- Filantropia City Hospital Craiova, Romania
| | - Adriana Uzoni
- Department of Psychiatry, University of Medicine Rostock, 18147 Rostock, Germany
| | - Aurel Popa-Wagner
- Research Center of Experimental and Clinical Medicine, University of Medicine and Pharmacy of Craiova, 200349, Romania
- Department of Psychiatry, University of Medicine Rostock, 18147 Rostock, Germany
| | - Anghel Andrei
- Biochemistry Department, University of Medicine and Pharmacy “Victor Babes” Timisoara
| | - Eugen-Bogdan Petcu
- Griffith University School of Medicine, Gold Coast Campus, Griffith University, QLD 4222, Australia
| |
Collapse
|
31
|
Stefano GB, Kream RM. Dysregulated mitochondrial and chloroplast bioenergetics from a translational medical perspective (Review). Int J Mol Med 2016; 37:547-55. [PMID: 26821064 PMCID: PMC4771107 DOI: 10.3892/ijmm.2016.2471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023] Open
Abstract
Mitochondria and chloroplasts represent endosymbiotic models of complex organelle development, driven by intense evolutionary pressure to provide exponentially enhanced ATP-dependent energy production functionally linked to cellular respiration and photosynthesis. Within the realm of translational medicine, it has become compellingly evident that mitochondrial dysfunction, resulting in compromised cellular bioenergetics, represents a key causative factor in the etiology and persistence of major diseases afflicting human populations. As a pathophysiological consequence of enhanced oxygen utilization that is functionally uncoupled from the oxidative phosphorylation of ADP, significant levels of reactive oxygen species (ROS) may be generated within mitochondria and chloroplasts, which may effectively compromise cellular energy production following prolonged stress/inflammatory conditions. Empirically determined homologies in biochemical pathways, and their respective encoding gene sequences between chloroplasts and mitochondria, suggest common origins via entrapped primordial bacterial ancestors. From evolutionary and developmental perspectives, the elucidation of multiple biochemical and molecular relationships responsible for errorless bioenergetics within mitochondrial and plastid complexes will most certainly enhance the depth of translational approaches to ameliorate or even prevent the destructive effects of multiple disease states. The selective choice of discussion points contained within the present review is designed to provide theoretical bases and translational insights into the pathophysiology of human diseases from a perspective of dysregulated mitochondrial bioenergetics with special reference to chloroplast biology.
Collapse
|
32
|
Son SW, Lee JS, Kim HG, Kim DW, Ahn YC, Son CG. Testosterone depletion increases the susceptibility of brain tissue to oxidative damage in a restraint stress mouse model. J Neurochem 2016; 136:106-117. [PMID: 26385432 DOI: 10.1111/jnc.13371] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/31/2022]
Abstract
Among sex hormones, estrogen is particularly well known to act as neuroprotective agent. Unlike estrogen, testosterone has not been well investigated in regard to its effects on the brain, especially under psychological stress. To investigate the role of testosterone in oxidative brain injuries under psychological stress, we adapted an orchiectomy and restraint stress model. BALB/c mice were subjected to either an orchiectomy or sham operation. After allowing 15 days for recovery, mice were re-divided into four groups according to exposure of restraint stress: sham, sham plus stress, orchiectomy, and orchiectomy plus stress. Serum testosterone was undetectable in orchiectomized groups and restraint-induced stress significantly reduced testosterone levels in sham plus stress group. The serum levels of corticosterone and adrenaline were notably elevated by restraint stress, and these elevated hormones were markedly augmented by orchiectomy. Two oxidative stressors and biomarkers for lipid and protein peroxidation were significantly increased in the cerebral cortex and hippocampus by restraint stress, while the reverse pattern was observed in antioxidant enzymes. These results were supported by histopathological findings, with 4-hydroxynonenal staining for oxidative injury and Fluoro-Jade B staining showing the degenerating neurons. The aforementioned patterns of oxidative injury were accelerated by orchiectomy. These findings strongly suggest the conclusion that testosterone exerts a protective effect against oxidative brain damage, especially under stressed conditions. Unlike estrogen, the effects of testosterone on the brain have not been thoroughly investigated. In order to investigate the role of testosterone in oxidative brain injuries under psychological stress, we adapted an orchiectomy and restraint stress model. Orchiectomy markedly augmented the restraint stress-induced elevation of serum corticosterone and adrenaline levels as well as oxidative alterations in brain tissues, especially in the hippocampus. These findings are the first evidence that testosterone depletion makes the brain prone to oxidative injury.
Collapse
Affiliation(s)
- Seung-Wan Son
- Department of Biomedical Engineering, College of Health Science, Korea University, Seongbuk-Gu, Seoul, Korea
| | - Jin-Seok Lee
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Oriental Medical Collage of Daejeon University, Jung-gu, Daejeon, South Korea
| | - Hyeong-Geug Kim
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Oriental Medical Collage of Daejeon University, Jung-gu, Daejeon, South Korea
| | - Dong-Woon Kim
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea
| | - Yo-Chan Ahn
- Department of Health Service Management, Daejeon University, Dong-gu, Daejeon, Korea
| | - Chang-Gue Son
- Department of Biomedical Engineering, College of Health Science, Korea University, Seongbuk-Gu, Seoul, Korea
- Liver and Immunology Research Center, Daejeon Oriental Hospital of Oriental Medical Collage of Daejeon University, Jung-gu, Daejeon, South Korea
| |
Collapse
|
33
|
L-carnitine enhances axonal plasticity and improves white-matter lesions after chronic hypoperfusion in rat brain. J Cereb Blood Flow Metab 2015; 35:382-91. [PMID: 25465043 PMCID: PMC4348379 DOI: 10.1038/jcbfm.2014.210] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 12/28/2022]
Abstract
Chronic cerebral hypoperfusion causes white-matter lesions (WMLs) with oxidative stress and cognitive impairment. However, the biologic mechanisms that regulate axonal plasticity under chronic cerebral hypoperfusion have not been fully investigated. Here, we investigated whether L-carnitine, an antioxidant agent, enhances axonal plasticity and oligodendrocyte expression, and explored the signaling pathways that mediate axonal plasticity in a rat chronic hypoperfusion model. Adult male Wistar rats subjected to ligation of the bilateral common carotid arteries (LBCCA) were treated with or without L-carnitine. L-carnitine-treated rats exhibited significantly reduced escape latency in the Morris water maze task at 28 days after chronic hypoperfusion. Western blot analysis indicated that L-carnitine increased levels of phosphorylated high-molecular weight neurofilament (pNFH), concurrent with a reduction in phosphorylated phosphatase tensin homolog deleted on chromosome 10 (PTEN), and increased phosphorylated Akt and mammalian target of rapamycin (mTOR) at 28 days after chronic hypoperfusion. L-carnitine reduced lipid peroxidation and oxidative DNA damage, and enhanced oligodendrocyte marker expression and myelin sheath thickness after chronic hypoperfusion. L-carnitine regulates the PTEN/Akt/mTOR signaling pathway, and enhances axonal plasticity while concurrently ameliorating oxidative stress and increasing oligodendrocyte myelination of axons, thereby improving WMLs and cognitive impairment in a rat chronic hypoperfusion model.
Collapse
|
34
|
Yoo DY, Kim W, Nam SM, Yoo M, Lee S, Yoon YS, Won MH, Hwang IK, Choi JH. Neuroprotective effects of Z-ajoene, an organosulfur compound derived from oil-macerated garlic, in the gerbil hippocampal CA1 region after transient forebrain ischemia. Food Chem Toxicol 2014; 72:1-7. [DOI: 10.1016/j.fct.2014.06.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 06/04/2014] [Accepted: 06/25/2014] [Indexed: 02/04/2023]
|
35
|
Lipoxin A4 methyl ester ameliorates cognitive deficits induced by chronic cerebral hypoperfusion through activating ERK/Nrf2 signaling pathway in rats. Pharmacol Biochem Behav 2014; 124:145-52. [DOI: 10.1016/j.pbb.2014.05.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/24/2014] [Accepted: 05/31/2014] [Indexed: 01/05/2023]
|
36
|
Oxidative Stress and the Use of Antioxidants in Stroke. Antioxidants (Basel) 2014; 3:472-501. [PMID: 26785066 PMCID: PMC4665418 DOI: 10.3390/antiox3030472] [Citation(s) in RCA: 184] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/08/2014] [Accepted: 05/14/2014] [Indexed: 12/12/2022] Open
Abstract
Transient or permanent interruption of cerebral blood flow by occlusion of a cerebral artery gives rise to an ischaemic stroke leading to irreversible damage or dysfunction to the cells within the affected tissue along with permanent or reversible neurological deficit. Extensive research has identified excitotoxicity, oxidative stress, inflammation and cell death as key contributory pathways underlying lesion progression. The cornerstone of treatment for acute ischaemic stroke remains reperfusion therapy with recombinant tissue plasminogen activator (rt-PA). The downstream sequelae of events resulting from spontaneous or pharmacological reperfusion lead to an imbalance in the production of harmful reactive oxygen species (ROS) over endogenous anti-oxidant protection strategies. As such, anti-oxidant therapy has long been investigated as a means to reduce the extent of injury resulting from ischaemic stroke with varying degrees of success. Here we discuss the production and source of these ROS and the various strategies employed to modulate levels. These strategies broadly attempt to inhibit ROS production or increase scavenging or degradation of ROS. While early clinical studies have failed to translate success from bench to bedside, the combination of anti-oxidants with existing thrombolytics or novel neuroprotectants may represent an avenue worthy of clinical investigation. Clearly, there is a pressing need to identify new therapeutic alternatives for the vast majority of patients who are not eligible to receive rt-PA for this debilitating and devastating disease.
Collapse
|
37
|
Akyol S, Erdogan S, Idiz N, Celik S, Kaya M, Ucar F, Dane S, Akyol O. The role of reactive oxygen species and oxidative stress in carbon monoxide toxicity: an in-depth analysis. Redox Rep 2014; 19:180-9. [PMID: 24773392 DOI: 10.1179/1351000214y.0000000094] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The underlying mechanism of the central nervous system (CNS) injury after acute carbon monoxide (CO) poisoning is interlaced with multiple factors including apoptosis, abnormal inflammatory responses, hypoxia, and ischemia/reperfusion-like problems. One of the current hypotheses with regard to the molecular mechanism of CO poisoning is the oxidative injury induced by reactive oxygen species, free radicals, and neuronal nitric oxide. Up to now, the relevant mechanism of this injury remains poorly understood. The weakening of antioxidant systems and the increase of lipid peroxidation in the CNS have been implicated, however. Accordingly, in this review, we will highlight the relationship between oxidative stress and CO poisoning from the perspective of forensic toxicology and molecular toxicology.
Collapse
|
38
|
Miller SL, Yawno T, Alers NO, Castillo-Melendez M, Supramaniam VG, VanZyl N, Sabaretnam T, Loose JM, Drummond GR, Walker DW, Jenkin G, Wallace EM. Antenatal antioxidant treatment with melatonin to decrease newborn neurodevelopmental deficits and brain injury caused by fetal growth restriction. J Pineal Res 2014; 56:283-94. [PMID: 24456220 DOI: 10.1111/jpi.12121] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 01/17/2014] [Indexed: 12/12/2022]
Abstract
Fetal intrauterine growth restriction (IUGR) is a serious pregnancy complication associated with increased rates of perinatal morbidity and mortality, and ultimately with long-term neurodevelopmental impairments. No intervention currently exists that can improve the structure and function of the IUGR brain before birth. Here, we investigated whether maternal antenatal melatonin administration reduced brain injury in ovine IUGR. IUGR was induced in pregnant sheep at 0.7 gestation and a subset of ewes received melatonin via intravenous infusion until term. IUGR, IUGR + melatonin (IUGR + MLT) and control lambs were born naturally, neonatal behavioral assessment was used to examine neurological function and at 24 hr after birth the brain was collected for the examination of neuropathology. Compared to control lambs, IUGR lambs took significantly longer to achieve normal neonatal lamb behaviors, such as standing and suckling. IUGR brains showed widespread cellular and axonal lipid peroxidation, and white matter hypomyelination and axonal damage. Maternal melatonin administration ameliorated oxidative stress, normalized myelination and rescued axonopathy within IUGR lamb brains, and IUGR + MLT lambs demonstrated significant functional improvements including a reduced time taken to attach to and suckle at the udder after birth. Based on these observations, we began a pilot clinical trial of oral melatonin administration to women with an IUGR fetus. Maternal melatonin was not associated with adverse maternal or fetal effects and it significantly reduced oxidative stress, as evidenced by reduced malondialdehyde levels, in the IUGR + MLT placenta compared to IUGR alone. Melatonin should be considered for antenatal neuroprotective therapy in human IUGR.
Collapse
Affiliation(s)
- Suzanne L Miller
- The Ritchie Centre, Monash Institute of Medical Research, Monash University, Clayton, Vic., Australia; Department of Obstetrics & Gynaecology, Southern Clinical School, Monash University, Clayton, Vic., Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Narayanan SP, Xu Z, Putluri N, Sreekumar A, Lemtalsi T, Caldwell RW, Caldwell RB. Arginase 2 deficiency reduces hyperoxia-mediated retinal neurodegeneration through the regulation of polyamine metabolism. Cell Death Dis 2014; 5:e1075. [PMID: 24556690 PMCID: PMC3944241 DOI: 10.1038/cddis.2014.23] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 12/27/2013] [Accepted: 01/07/2014] [Indexed: 11/16/2022]
Abstract
Hyperoxia treatment has been known to induce neuronal and glial death in the developing central nervous system. Retinopathy of prematurity (ROP) is a devastating disease in premature infants and a major cause of childhood vision impairment. Studies indicate that, in addition to vascular injury, retinal neurons are also affected in ROP. Using an oxygen-induced retinopathy (OIR) mouse model for ROP, we have previously shown that deletion of the arginase 2 (A2) significantly reduced neuro-glial injury and improved retinal function. In the current study, we investigated the mechanism of A2 deficiency-mediated neuroprotection in the OIR retina. Hyperoxia treatment has been known to induce neuronal death in neonates. During the hyperoxia phase of OIR, a significant increase in the number of apoptotic cells was observed in the wild-type (WT) OIR retina compared with A2-deficient OIR. Mass spectrometric analysis showed alterations in polyamine metabolism in WT OIR retina. Further, increased expression level of spermine oxidase was observed in WT OIR retina, suggesting increased oxidation of polyamines in OIR retina. These changes were minimal in A2-deficient OIR retina. Treatment using the polyamine oxidase inhibitor, N, N'-bis (2, 3-butadienyl)-1, 4-butanediamine dihydrochloride, significantly improved neuronal survival during OIR treatment. Our data suggest that retinal arginase is involved in the hyperoxia-induced neuronal degeneration in the OIR model, through the regulation of polyamine metabolism.
Collapse
Affiliation(s)
- S P Narayanan
- Vision Discovery Institute, Georgia Regents University, Augusta, GA, USA
- Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Z Xu
- Vision Discovery Institute, Georgia Regents University, Augusta, GA, USA
- Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - N Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - A Sreekumar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - T Lemtalsi
- Vision Discovery Institute, Georgia Regents University, Augusta, GA, USA
- Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
| | - R W Caldwell
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA, USA
| | - R B Caldwell
- Vision Discovery Institute, Georgia Regents University, Augusta, GA, USA
- Vascular Biology Center, Georgia Regents University, Augusta, GA, USA
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
- Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
40
|
Miljković D, Spasojević I. Multiple sclerosis: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal 2013; 19:2286-334. [PMID: 23473637 PMCID: PMC3869544 DOI: 10.1089/ars.2012.5068] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/09/2012] [Accepted: 03/09/2013] [Indexed: 12/15/2022]
Abstract
The pathophysiology of multiple sclerosis (MS) involves several components: redox, inflammatory/autoimmune, vascular, and neurodegenerative. All of them are supported by the intertwined lines of evidence, and none of them should be written off. However, the exact mechanisms of MS initiation, its development, and progression are still elusive, despite the impressive pace by which the data on MS are accumulating. In this review, we will try to integrate the current facts and concepts, focusing on the role of redox changes and various reactive species in MS. Knowing the schedule of initial changes in pathogenic factors and the key turning points, as well as understanding the redox processes involved in MS pathogenesis is the way to enable MS prevention, early treatment, and the development of therapies that target specific pathophysiological components of the heterogeneous mechanisms of MS, which could alleviate the symptoms and hopefully stop MS. Pertinent to this, we will outline (i) redox processes involved in MS initiation; (ii) the role of reactive species in inflammation; (iii) prooxidative changes responsible for neurodegeneration; and (iv) the potential of antioxidative therapy.
Collapse
Affiliation(s)
- Djordje Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković,” University of Belgrade, Belgrade, Serbia
| | - Ivan Spasojević
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
41
|
ROS and brain diseases: the good, the bad, and the ugly. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:963520. [PMID: 24381719 PMCID: PMC3871919 DOI: 10.1155/2013/963520] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 12/31/2022]
Abstract
The brain is a major metabolizer of oxygen and yet has relatively feeble protective antioxidant mechanisms. This paper reviews the Janus-faced properties of reactive oxygen species. It will describe the positive aspects of moderately induced ROS but it will also outline recent research findings concerning the impact of oxidative and nitrooxidative stress on neuronal structure and function in neuropsychiatric diseases, including major depression. A common denominator of all neuropsychiatric diseases including schizophrenia and ADHD is an increased inflammatory response of the brain caused either by an exposure to proinflammatory agents during development or an accumulation of degenerated neurons, oxidized proteins, glycated products, or lipid peroxidation in the adult brain. Therefore, modulation of the prooxidant-antioxidant balance provides a therapeutic option which can be used to improve neuroprotection in response to oxidative stress. We also discuss the neuroprotective role of the nuclear factor erythroid 2-related factor (Nrf2) in the aged brain in response to oxidative stressors and nanoparticle-mediated delivery of ROS-scavenging drugs. The antioxidant therapy is a novel therapeutic strategy. However, the available drugs have pleiotropic actions and are not fully characterized in the clinic. Additional clinical trials are needed to assess the risks and benefits of antioxidant therapies for neuropsychiatric disorders.
Collapse
|
42
|
Abstract
Acrylamide, a food contaminant, belongs to a large class of structurally similar toxic chemicals, 'type-2 alkenes', to which humans are widely exposed. Besides, occupational exposure to acrylamide has received wide attention through the last decades. It is classified as a neurotoxin and there are three important hypothesis considering acrylamide neurotoxicity: inhibition of kinesin-based fast axonal transport, alteration of neurotransmitter levels, and direct inhibition of neurotransmission. While many researchers believe that exposure of humans to relatively low levels of acrylamide in the diet will not result in clinical neuropathy, some neurotoxicologists are concerned about the potential for its cumulative neurotoxicity. It has been shown in several studies that the same neurotoxic effects can be observed at low and high doses of acrylamide, with the low doses simply requiring longer exposures. This review is focused on the neurotoxicity of acrylamide and its possible outcomes.
Collapse
|
43
|
Abstract
The pathogenesis of acute brain ischemia (ABI) is highly complex and involves multiple mechanisms including free radical generation. Imbalance between the cellular production of free radicals and the ability of cells to defend against them is referred to as oxidative stress. Oxidative stress is one of the mechanisms contributing to neuronal damage, potentially induced through the ABI. Through interactions with a large number of molecules, reactive oxygen species may irreversibly destroy or alter the function of the cellular lipids, proteins, and nucleic acids and initiate cell signaling pathways after cerebral ischemia. Future investigations should focus on the understanding of oxidative stress mechanisms and neuroprotection in order to discover new treatment targets.
Collapse
Affiliation(s)
- Djordje Radak
- Department of Vascular Surgery, Dedinje Cardiovascular Institute, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivana Resanovic
- Laboratory of Radiobiology and Molecular Genetics, Institute Vinca, University of Belgrade, Belgrade, Serbia
| | - Esma R Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Institute Vinca, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
44
|
Hyperglycemia accelerates apparent diffusion coefficient-defined lesion growth after focal cerebral ischemia in rats with and without features of metabolic syndrome. J Cereb Blood Flow Metab 2013; 33:1556-63. [PMID: 23838826 PMCID: PMC3790923 DOI: 10.1038/jcbfm.2013.107] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/31/2013] [Accepted: 06/04/2013] [Indexed: 01/04/2023]
Abstract
Poststroke hyperglycemia is associated with a poor outcome yet clinical management is inadequately informed. We sought to determine whether clinically relevant levels of hyperglycemia exert detrimental effects on the early evolution of focal ischemic brain damage, as determined by magnetic resonance imaging, in normal rats and in those modeling the 'metabolic syndrome'. Wistar Kyoto (WKY) or fructose-fed spontaneously hypertensive stroke-prone (ffSHRSP) rats were randomly allocated to groups for glucose or vehicle administration before permanent middle cerebral artery occlusion. Diffusion-weighted imaging was carried out over the first 4 hours after middle cerebral artery occlusion and lesion volume calculated from apparent diffusion coefficient maps. Infarct volume and immunostaining for markers of oxidative stress were measured in the fixed brain sections at 24 hours. Hyperglycemia rapidly exacerbated early ischemic damage in both WKY and ffSHRSP rats but increased infarct volume only in WKY rats. There was only limited evidence of oxidative stress in hyperglycemic animals. Acute hyperglycemia, at clinically relevant levels, exacerbates early ischemic damage in both normal and metabolic syndrome rats. Management of hyperglycemia may have greatest benefit when performed in the acute phase after stroke in the absence or presence of comorbidities.
Collapse
|
45
|
Narayanan SP, Rojas M, Suwanpradid J, Toque HA, Caldwell RW, Caldwell RB. Arginase in retinopathy. Prog Retin Eye Res 2013; 36:260-80. [PMID: 23830845 PMCID: PMC3759622 DOI: 10.1016/j.preteyeres.2013.06.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/14/2013] [Accepted: 06/25/2013] [Indexed: 12/12/2022]
Abstract
Ischemic retinopathies, such as diabetic retinopathy (DR), retinopathy of prematurity and retinal vein occlusion are a major cause of blindness in developed nations worldwide. Each of these conditions is associated with early neurovascular dysfunction. However, conventional therapies target clinically significant macula edema or neovascularization, which occur much later. Intra-ocular injections of anti-VEGF show promise in reducing retinal edema, but the effects are usually transient and the need for repeated injections increases the risk of intraocular infection. Laser photocoagulation can control pathological neovascularization, but may impair vision and in some patients the retinopathy continues to progress. Moreover, neither treatment targets early stage disease or promotes repair. This review examines the potential role of the ureahydrolase enzyme arginase as a therapeutic target for the treatment of ischemic retinopathy. Arginase metabolizes l-arginine to form proline, polyamines and glutamate. Excessive arginase activity reduces the l-arginine supply for nitric oxide synthase (NOS), causing it to become uncoupled and produce superoxide and less NO. Superoxide and NO react and form the toxic oxidant peroxynitrite. The catabolic products of polyamine oxidation and glutamate can induce more oxidative stress and DNA damage, both of which can cause cellular injury. Studies indicate that neurovascular injury during retinopathy is associated with increased arginase expression/activity, decreased NO, polyamine oxidation, formation of superoxide and peroxynitrite and dysfunction and injury of both vascular and neural cells. Furthermore, data indicate that the cytosolic isoform arginase I (AI) is involved in hyperglycemia-induced dysfunction and injury of vascular endothelial cells whereas the mitochondrial isoform arginase II (AII) is involved in neurovascular dysfunction and death following hyperoxia exposure. Thus, we postulate that activation of the arginase pathway causes neurovascular injury by uncoupling NOS and inducing polyamine oxidation and glutamate formation, thereby reducing NO and increasing oxidative stress, all of which contribute to the retinopathic process.
Collapse
Affiliation(s)
- S. Priya Narayanan
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Modesto Rojas
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Jutamas Suwanpradid
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology & Toxicology, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - R. William Caldwell
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Pharmacology & Toxicology, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Ruth B. Caldwell
- Vision Discovery Institute, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- VA Medical Center, One Freedom Way, Augusta, GA, USA
| |
Collapse
|
46
|
Zhang W, Sato K, Hayashi T, Omori N, Nagano I, Kato S, Horiuchi S, Abe K. Extension of ischemic therapeutic time window by a free radical scavenger, Edaravone, reperfused with tPA in rat brain. Neurol Res 2013; 26:342-8. [PMID: 15142331 DOI: 10.1179/016164104225014058] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
3-methyl-1-phenyl-2-pyrazolin-5-one (Edaravone) is a free radical scavenger. We tested the hypothesis that combination treatment of Edaravone and recombinant tissue plasminogen activator (tPA) extends the therapeutic time window. Male Wistar rats were subjected to 1.5-, 3.0- or 4.5-hour middle cerebral artery (MCA) occlusion (MCAO) by a nylon thread. Animals were randomly divided into four groups. The Sham group rats were operated without MCAO and drug injection. In the Vehicle-treated group the same volume of saline was given every 1.5 hours from just after MCAO to just before reperfusion. In the Vehicle + tPA-treated group saline injection was given as above and tPA (5 mg/kg, i.v.) was given once just after reperfusion. Edaravone+tPA-treated group: Edaravone (3 mg/kg, i.v.) was given every 1.5 hours instead of saline and tPA injection as above. Survival rate, infarct size and evidence of apoptosis and hemorrhage were examined in the animals. Combining administration of Edaravone+tPA significantly increased survival rate after 3 hours of transient MCAO, and reduced infarct volume after 1.5 hours of transient MCAO compared with the vehicle or vehicle+tPA groups. In Edaravone+tPA-treated group, the number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end labeling (TUNEL) and 4-hydroxynonenal (4-HNE) positive cells were reduced at 16 hours after 3 hours of transient MCAO, but not in advanced glycation end products (AGEs) and 8-hydroxy-2'-deoxyguanosine (8-OHdG). Hemorrhage rate and the area decreased in the Edaravone+tPA-treated group. The combination therapy of Edaravone+tPA increased survival rate, and reduced the infarct volume and hemorrhage with reduction of lipid peroxidation. Therefore, Edaravone combination is expected to extend the therapeutic time window of tPA in the clinical situation.
Collapse
Affiliation(s)
- Wenri Zhang
- Department of Neurology, Graduate School of Medicine and Dentistry, Okayama University, 2-5-1 Shikatacho, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Ghosh A, Sarkar S, Mandal AK, Das N. Neuroprotective role of nanoencapsulated quercetin in combating ischemia-reperfusion induced neuronal damage in young and aged rats. PLoS One 2013; 8:e57735. [PMID: 23620721 PMCID: PMC3631169 DOI: 10.1371/journal.pone.0057735] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/24/2013] [Indexed: 11/18/2022] Open
Abstract
Cerebral stroke is the leading cause of death and permanent disability among elderly people. In both humans and animals, cerebral ischemia damages the nerve cells in vulnerable regions of the brain, viz., hippocampus, cerebral cortex, cerebellum, and hypothalamus. The present study was conducted to evaluate the therapeutic efficacy of nanoencapsulated quercetin (QC) in combating ischemia-reperfusion-induced neuronal damage in young and aged Swiss Albino rats. Cerebral ischemia was induced by occlusion of the common carotid arteries of both young and aged rats followed by reperfusion. Nanoencapsulated quercetin (2.7 mg/kg b wt) was administered to both groups of animals via oral gavage two hours prior to ischemic insults as well as post-operation till day 3. Cerebral ischemia and 30 min consecutive reperfusion caused a substantial increase in lipid peroxidation, decreased antioxidant enzyme activities and tissue osmolality in different brain regions of both groups of animals. It also decreased mitochondrial membrane microviscosity and increased reactive oxygen species (ROS) generation in different brain regions of young and aged rats. Among the brain regions studied, the hippocampus appeared to be the worst affected region showing increased upregulation of iNOS and caspase-3 activity with decreased neuronal count in the CA1 and CA3 subfields of both young and aged rats. Furthermore, three days of continuous reperfusion after ischemia caused massive damage to neuronal cells. However, it was observed that oral treatment of nanoencapsulated quercetin (2.7 mg/kg b wt) resulted in downregulation of iNOS and caspase-3 activities and improved neuronal count in the hippocampal subfields even 3 days after reperfusion. Moreover, the nanoformulation imparted a significant level of protection in the antioxidant status in different brain regions, thus contributing to a better understanding of the given pathophysiological processes causing ischemic neuronal damage.
Collapse
Affiliation(s)
- Aparajita Ghosh
- Biomembrane Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| | - Sibani Sarkar
- Biomembrane Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Ardhendu K. Mandal
- Biomembrane Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Nirmalendu Das
- Biomembrane Division, Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| |
Collapse
|
48
|
Hamakawa M, Ishida A, Tamakoshi K, Shimada H, Nakashima H, Noguchi T, Toyokuni S, Ishida K. Repeated short-term daily exercise ameliorates oxidative cerebral damage and the resultant motor dysfunction after transient ischemia in rats. J Clin Biochem Nutr 2013; 53:8-14. [PMID: 23874064 PMCID: PMC3705152 DOI: 10.3164/jcbn.12-72] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 12/13/2012] [Indexed: 12/18/2022] Open
Abstract
Long-term exercise prior to brain ischemia enhances the activities of antioxidant enzymes and leads to a significant reduction in brain damage and neurological deficits in rats subjected to transient middle cerebral artery occlusion. However, it has not been established whether relatively short-term exercise generates similar results following middle cerebral artery occlusion. We aimed to determine whether short-term exercise could reduce oxidative damage and prevent sensori-motor dysfunction. Male Wistar rats were subjected to perform daily exercise on a treadmill for 30 min at a speed of 15 m/min for 3 weeks, followed by a 90-min middle cerebral artery occlusion. Animals were assessed after middle cerebral artery occlusion for neurological deficits and sensori-motor function. Brain tissues were processed to evaluate infarct volume and oxidative damage. Oxidative stress was assessed using immunohistochemistry for 4-hydroxy-2-nonenal-modified proteins and 8-hydroxy-2'-deoxyguanosine. Antioxidant enzymes were evaluated using immunohistochemistry for thioredoxin and activity assay for superoxide dismutase. Exercise for 3 weeks decreased the severity of paralysis and impairment in forelimb motor coordination. Furthermore, exercise had effect on superoxide dismutase and reduced the infarct volume and the number of cells immunopositive for 4-hydroxy-2-nonenal-modified proteins and 8-hydroxy-2'-deoxyguanosine. Our results suggest that pre-conditioning treadmill exercise for 3 weeks is useful for ameliorating ischemia-induced brain injury.
Collapse
Affiliation(s)
- Michiru Hamakawa
- Department of Physical Therapy, Nagoya University Graduate School of Medicine, 1-1-20 Daiko-minami, Higashi-ku, Nagoya, Aichi 461-8673, Japan ; Okinawa Rehabilitation Center Hospital, 2-15-1 Hiyagon, Okinawa, Okinawa 904-2173, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kaur P, Liu F, Tan JR, Lim KY, Sepramaniam S, Karolina DS, Armugam A, Jeyaseelan K. Non-Coding RNAs as Potential Neuroprotectants against Ischemic Brain Injury. Brain Sci 2013; 3:360-95. [PMID: 24961318 PMCID: PMC4061830 DOI: 10.3390/brainsci3010360] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/19/2013] [Accepted: 03/06/2013] [Indexed: 01/24/2023] Open
Abstract
Over the past decade, scientific discoveries have highlighted new roles for a unique class of non-coding RNAs. Transcribed from the genome, these non-coding RNAs have been implicated in determining the biological complexity seen in mammals by acting as transcriptional and translational regulators. Non-coding RNAs, which can be sub-classified into long non-coding RNAs, microRNAs, PIWI-interacting RNAs and several others, are widely expressed in the nervous system with roles in neurogenesis, development and maintenance of the neuronal phenotype. Perturbations of these non-coding transcripts have been observed in ischemic preconditioning as well as ischemic brain injury with characterization of the mechanisms by which they confer toxicity. Their dysregulation may also confer pathogenic conditions in neurovascular diseases. A better understanding of their expression patterns and functions has uncovered the potential use of these riboregulators as neuroprotectants to antagonize the detrimental molecular events taking place upon ischemic-reperfusion injury. In this review, we discuss the various roles of non-coding RNAs in brain development and their mechanisms of gene regulation in relation to ischemic brain injury. We will also address the future directions and open questions for identifying promising non-coding RNAs that could eventually serve as potential neuroprotectants against ischemic brain injury.
Collapse
Affiliation(s)
- Prameet Kaur
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Fujia Liu
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Jun Rong Tan
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Kai Ying Lim
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Sugunavathi Sepramaniam
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Dwi Setyowati Karolina
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Arunmozhiarasi Armugam
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| | - Kandiah Jeyaseelan
- Department of Biochemistry and Neuroscience Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore.
| |
Collapse
|
50
|
Al Nimer F, Ström M, Lindblom R, Aeinehband S, Bellander BM, Nyengaard JR, Lidman O, Piehl F. Naturally occurring variation in the Glutathione-S-Transferase 4 gene determines neurodegeneration after traumatic brain injury. Antioxid Redox Signal 2013; 18:784-94. [PMID: 22881716 PMCID: PMC3555113 DOI: 10.1089/ars.2011.4440] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIM Genetic factors are important for outcome after traumatic brain injury (TBI), although exact knowledge of relevant genes/pathways is still lacking. We here used an unbiased approach to define differentially activated pathways between the inbred DA and PVG rat strains. The results prompted us to study further if a naturally occurring genetic variation in glutathione-S-transferase alpha 4 (Gsta4) affects the outcome after TBI. RESULTS Survival of neurons after experimental TBI is increased in PVG compared to the DA strain. Global expression profiling analysis shows the glutathione metabolism pathway to be the most regulated between the strains, with increased Gsta4 in PVG among top regulated transcripts. A congenic strain (R5) with a PVG genomic insert containing the Gsta4 gene on DA background displays a reversal of the strain pattern for Gsta4 expression and increased survival of neurons compared to DA. Gsta4 is known to effectively reduce 4-hydroxynonenal (4-HNE), a noxious by-product of lipid peroxidation. Immunostaining of 4-HNE was evident in both rat and human TBI. Intracerebral injection of 4-HNE resulted in neurodegeneration with increased levels of a marker for nerve injury in cerebrospinal fluid of DA compared to R5. INNOVATION These findings provide strong support for the notion that the inherent capability of coping with increased 4-HNE after TBI affects outcome in terms of nerve cell loss. CONCLUSION A naturally occurring variation in Gsta4 expression in rats affects neurodegeneration after TBI. Further studies are needed to explore if genetic variability in Gsta4 can be associated to outcome also in human TBI.
Collapse
Affiliation(s)
- Faiez Al Nimer
- Neuroimmunology Unit, Department of Clinical Neuroscience, Karolinska University Hospital, Stockholm, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|