1
|
Sengking J, Mahakkanukrauh P. The underlying mechanism of calcium toxicity-induced autophagic cell death and lysosomal degradation in early stage of cerebral ischemia. Anat Cell Biol 2024; 57:155-162. [PMID: 38680098 PMCID: PMC11184419 DOI: 10.5115/acb.24.003] [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: 01/05/2024] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 05/01/2024] Open
Abstract
Cerebral ischemia is the important cause of worldwide disability and mortality, that is one of the obstruction of blood vessels supplying to the brain. In early stage, glutamate excitotoxicity and high level of intracellular calcium (Ca2+) are the major processes which can promote many downstream signaling involving in neuronal death and brain tissue damaging. Moreover, autophagy, the reusing of damaged cell organelles, is affected in early ischemia. Under ischemic conditions, autophagy plays an important role to maintain energy of the brain and its function. In the other hand, over intracellular Ca2+ accumulation triggers excessive autophagic process and lysosomal degradation leading to autophagic process impairment which finally induce neuronal death. This article reviews the association between intracellular Ca2+ and autophagic process in acute stage of ischemic stroke.
Collapse
Affiliation(s)
- Jirakhamon Sengking
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pasuk Mahakkanukrauh
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Excellence in Osteology Research and Training Center (ORTC), Chaing Mai University, Chiang Mai, Thailand
| |
Collapse
|
2
|
Dhapola R, Medhi B, HariKrishnaReddy D. Insight into the pathophysiological advances and molecular mechanisms underlying cerebral stroke: current status. Mol Biol Rep 2024; 51:649. [PMID: 38733445 DOI: 10.1007/s11033-024-09597-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Molecular pathways involved in cerebral stroke are diverse. The major pathophysiological events that are observed in stroke comprises of excitotoxicity, oxidative stress, mitochondrial damage, endoplasmic reticulum stress, cellular acidosis, blood-brain barrier disruption, neuronal swelling and neuronal network mutilation. Various biomolecules are involved in these pathways and several major proteins are upregulated and/or suppressed following stroke. Different types of receptors, ion channels and transporters are activated. Fluctuations in levels of various ions and neurotransmitters have been observed. Cells involved in immune responses and various mediators involved in neuro-inflammation get upregulated progressing the pathogenesis of the disease. Despite of enormity of the problem, there is not a single therapy that can limit infarction and neurological disability due to stroke. This is because of poor understanding of the complex interplay between these pathophysiological processes. This review focuses upon the past to present research on pathophysiological events that are involved in stroke and various factors that are leading to neuronal death following cerebral stroke. This will pave a way to researchers for developing new potent therapeutics that can aid in the treatment of cerebral stroke.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
3
|
Norouzkhani N, Afshari S, Sadatmadani SF, Mollaqasem MM, Mosadeghi S, Ghadri H, Fazlizade S, Alizadeh K, Akbari Javar P, Amiri H, Foroughi E, Ansari A, Mousazadeh K, Davany BA, Akhtari kohnehshahri A, Alizadeh A, Dadkhah PA, Poudineh M. Therapeutic potential of berries in age-related neurological disorders. Front Pharmacol 2024; 15:1348127. [PMID: 38783949 PMCID: PMC11112503 DOI: 10.3389/fphar.2024.1348127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/10/2024] [Indexed: 05/25/2024] Open
Abstract
Aging significantly impacts several age-related neurological problems, such as stroke, brain tumors, oxidative stress, neurodegenerative diseases (Alzheimer's, Parkinson's, and dementia), neuroinflammation, and neurotoxicity. Current treatments for these conditions often come with side effects like hallucinations, dyskinesia, nausea, diarrhea, and gastrointestinal distress. Given the widespread availability and cultural acceptance of natural remedies, research is exploring the potential effectiveness of plants in common medicines. The ancient medical system used many botanical drugs and medicinal plants to treat a wide range of diseases, including age-related neurological problems. According to current clinical investigations, berries improve motor and cognitive functions and protect against age-related neurodegenerative diseases. Additionally, berries may influence signaling pathways critical to neurotransmission, cell survival, inflammation regulation, and neuroplasticity. The abundance of phytochemicals in berries is believed to contribute to these potentially neuroprotective effects. This review aimed to explore the potential benefits of berries as a source of natural neuroprotective agents for age-related neurological disorders.
Collapse
Affiliation(s)
- Narges Norouzkhani
- Department of Medical Informatics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shaghayegh Afshari
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | | | - Shakila Mosadeghi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hani Ghadri
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Safa Fazlizade
- Student Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Keyvan Alizadeh
- Student Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Pouyan Akbari Javar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Hamidreza Amiri
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Elaheh Foroughi
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Kourosh Mousazadeh
- School of Medicine, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | | | - Ata Akhtari kohnehshahri
- Student Research Committee, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Alaleh Alizadeh
- Student Research Committee, Faculty of Medicine, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Parisa Alsadat Dadkhah
- Student Research Committee, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
4
|
Yang Y, Hao T, Yao X, Che Y, Liu Y, Fang M, Wang Y, Zhou D, Chai H, Li N, Hou Y. Crebanine ameliorates ischemia-reperfusion brain damage by inhibiting oxidative stress and neuroinflammation mediated by NADPH oxidase 2 in microglia. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155044. [PMID: 37634486 DOI: 10.1016/j.phymed.2023.155044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/30/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND The urgent challenge for ischemic stroke treatment is the lack of effective neuroprotectants that target multiple pathological processes. Crebanine, an isoquinoline-like alkaloid with superior pharmacological activities, presents itself as a promising candidate for neuroprotection. However, its effects and mechanisms on ischemic stroke remain unknown. METHODS The effects of crebanine on brain damage following ischemic stroke were evaluated using the middle cerebral artery occlusion and reperfusion (MCAO/R) model. Mechanism of action was investigated using both MCAO/R rats and lipopolysaccharide (LPS)-activated BV-2 cells. RESULTS We initially demonstrated that crebanine effectively ameliorated the neurological deficits in MCAO/R rats, while also reducing brain edema and infarction. Treatment with crebanine resulted in the up-regulation of NeuN+ fluorescence density and down-regulation of FJB+ cell count, and mitigated synaptic damage. Crebanine attenuated the hyperactivation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) by downregulating NADP+ and NADPH levels, suppressing gp91phox and p47phox expressions, and reducing p47phox membrane translocation in Iba-1+ cells. Additionally, crebanine reduced the quantity of Iba-1+ cells and protein expression. Correlation analysis has demonstrated that the inhibition of NOX2 activation in microglia is beneficial for mitigating I/R brain injuries. Moreover, crebanine exhibited significant antioxidant properties by down-regulating the expression of superoxide anion and intracellular reactive oxygen species in vivo and in vitro, and reducing lipid and DNA peroxidation. Crebanine exerted anti-inflammatory effect, as evidenced by the reduction in the expressions of nitric oxide, interleukin 1β, tumor necrosis factor α, interleukin 6, and inducible nitric oxide synthase. The effect of crebanine was achieved through the suppression of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) signaling pathway. This is supported by evidence showing reduced NF-κB p65 promoter activity and nucleus translocation, as well as suppressed IκBα phosphorylation and degradation. Additionally, it inhibited the phosphorylation of ERK, JNK, and p38 MAPKs. Importantly, the anti-oxidative stress and neuroinflammation effects of crebanine were further enhanced after silencing gp91phox and p47phox. CONCLUSION Crebanine alleviated the brain damages of MCAO/R rats by inhibiting oxidative stress and neuroinflammation mediated by NOX2 in microglia, implying crebanine might be a potential natural drug for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Yanqiu Yang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China; National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Tingyu Hao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China; National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Xiaohu Yao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China; National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China
| | - Yue Che
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Mingxia Fang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yingjie Wang
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China
| | - Huifang Chai
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Key Laboratory of Innovative Traditional Chinese Medicine for Major Chronic Diseases of Liaoning province, Key Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang City, Shenyang Pharmaceutical University, Shenyang, China.
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, China; National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Key Laboratory of Data Analytics and Optimization for Smart Industry, Ministry of Education, Northeastern University, Shenyang, China.
| |
Collapse
|
5
|
He R, Zhang X, Pang C, Lin L, Li S, Jin L, Ding L, Wang W. Inhibition of NADPH oxidase 2 improves cognitive abilities by modulating aquaporin-4 after traumatic brain injury in mice. Heliyon 2023; 9:e22035. [PMID: 38053850 PMCID: PMC10694165 DOI: 10.1016/j.heliyon.2023.e22035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 09/02/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023] Open
Abstract
Traumatic brain injury (TBI) is caused by acquired damage that includes cerebral edema after a mechanical injury and may cause cognitive impairment. We explored the role of nicotinamide adenine dinucleotide phosphate oxidase 2 (NADPH oxidase 2; NOX2) and aquaporin-4 (AQP4) in the process of edema and cognitive abilities after TBI in NOX2-/- and AQP4-/- mice by using the Morris water maze test (MWM), step-down test (STD), novel object recognition test (NOR) and western blotting. Knockout of NOX2 in mice decreased the AQP4 and reduce edema in the hippocampus and cortex after TBI in mice. Moreover, inhibiting AQP4 by 2-(nicotinamide)-1,3,4-thiadiazole (TGN-020) or genetic deletion of AQP4 could attenuate neurological deficits without changing reactive oxygen species (ROS) levels after TBI in mice. Taken together, we suspected that inhibiting NOX2 could improve cognitive abilities by modulating ROS levels, then affecting AQP4 levels and brain edema after in TBI mice. Our study demonstrated that NOX2 play a key role in decreasing edema in brain and improving cognitive abilities by modulating AQP4 after TBI.
Collapse
Affiliation(s)
- Ruixing He
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Xiaotian Zhang
- Department of Neurosurgery, Hongze District People's Hospital of Huai'an City, Huai'an, Jiangsu, 223300, China
| | - Cong Pang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Lihui Lin
- Department of Pharmacy, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Shaoxun Li
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Luhao Jin
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Lianshu Ding
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| | - Weijie Wang
- Department of Neurosurgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, 223300, China
| |
Collapse
|
6
|
Kaur P, Khan H, Grewal AK, Dua K, Singh TG. Therapeutic potential of NOX inhibitors in neuropsychiatric disorders. Psychopharmacology (Berl) 2023; 240:1825-1840. [PMID: 37507462 DOI: 10.1007/s00213-023-06424-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
RATIONALE Neuropsychiatric disorders encompass a broad category of medical conditions that include both neurology as well as psychiatry such as major depressive disorder, autism spectrum disorder, bipolar disorder, schizophrenia as well as psychosis. OBJECTIVE NADPH-oxidase (NOX), which is the free radical generator, plays a substantial part in oxidative stress in neuropsychiatric disorders. It is thought that elevated oxidative stress as well as neuroinflammation plays a part in the emergence of neuropsychiatric disorders. Including two linked with membranes and four with subunits of cytosol, NOX is a complex of multiple subunits. NOX has been linked to a significant source of reactive oxygen species in the brain. NOX has been shown to control memory processing and neural signaling. However, excessive NOX production has been linked to cardiovascular disorders, CNS degeneration, and neurotoxicity. The increase in NOX leads to the progression of neuropsychiatric disorders. RESULT Our review mainly emphasized the characteristics of NOX and its various mechanisms, the modulation of NOX in various neuropsychiatric disorders, and various studies supporting the fact that NOX might be the potential therapeutic target for neuropsychiatric disorders. CONCLUSION Here, we summarizes various pharmacological studies involving NOX inhibitors in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Parneet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | | | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | | |
Collapse
|
7
|
Riggins TE, Whitsitt QA, Saxena A, Hunter E, Hunt B, Thompson CH, Moore MG, Purcell EK. Gene Expression Changes in Cultured Reactive Rat Astrocyte Models and Comparison to Device-Associated Effects in the Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.522870. [PMID: 36712012 PMCID: PMC9881929 DOI: 10.1101/2023.01.06.522870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Implanted microelectrode arrays hold immense therapeutic potential for many neurodegenerative diseases. However, a foreign body response limits long-term device performance. Recent literature supports the role of astrocytes in the response to damage to the central nervous system (CNS) and suggests that reactive astrocytes exist on a spectrum of phenotypes, from beneficial to neurotoxic. The goal of our study was to gain insight into the subtypes of reactive astrocytes responding to electrodes implanted in the brain. In this study, we tested the transcriptomic profile of two reactive astrocyte culture models (cytokine cocktail or lipopolysaccharide, LPS) utilizing RNA sequencing, which we then compared to differential gene expression surrounding devices inserted into rat motor cortex via spatial transcriptomics. We interpreted changes in the genetic expression of the culture models to that of 24 hour, 1 week and 6 week rat tissue samples at multiple distances radiating from the injury site. We found overlapping expression of up to ∼250 genes between in vitro models and in vivo effects, depending on duration of implantation. Cytokine-induced cells shared more genes in common with chronically implanted tissue (≥1 week) in comparison to LPS-exposed cells. We revealed localized expression of a subset of these intersecting genes (e.g., Serping1, Chi3l1, and Cyp7b1) in regions of device-encapsulating, glial fibrillary acidic protein (GFAP)-expressing astrocytes identified with immunohistochemistry. We applied a factorization approach to assess the strength of the relationship between reactivity markers and the spatial distribution of GFAP-expressing astrocytes in vivo . We also provide lists of hundreds of differentially expressed genes between reactive culture models and untreated controls, and we observed 311 shared genes between the cytokine induced model and the LPS-reaction induced control model. Our results show that comparisons of reactive astrocyte culture models with spatial transcriptomics data can reveal new biomarkers of the foreign body response to implantable neurotechnology. These comparisons also provide a strategy to assess the development of in vitro models of the tissue response to implanted electrodes.
Collapse
|
8
|
Anwar MM, Sah R, Shrestha S, Ozaki A, Roy N, Fathah Z, Rodriguez-Morales AJ. Disengaging the COVID-19 Clutch as a Discerning Eye Over the Inflammatory Circuit During SARS-CoV-2 Infection. Inflammation 2022; 45:1875-1894. [PMID: 35639261 PMCID: PMC9153229 DOI: 10.1007/s10753-022-01674-5] [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] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the cytokine release syndrome (CRS) and leads to multiorgan dysfunction. Mitochondrial dynamics are fundamental to protect against environmental insults, but they are highly susceptible to viral infections. Defective mitochondria are potential sources of reactive oxygen species (ROS). Infection with SARS-CoV-2 damages mitochondria, alters autophagy, reduces nitric oxide (NO), and increases both nicotinamide adenine dinucleotide phosphate oxidases (NOX) and ROS. Patients with coronavirus disease 2019 (COVID-19) exhibited activated toll-like receptors (TLRs) and the Nucleotide-binding and oligomerization domain (NOD-), leucine-rich repeat (LRR-), pyrin domain-containing protein 3 (NLRP3) inflammasome. The activation of TLRs and NLRP3 by SARS-CoV-2 induces interleukin 6 (IL-6), IL-1β, IL-18, and lactate dehydrogenase (LDH). Herein, we outline the inflammatory circuit of COVID-19 and what occurs behind the scene, the interplay of NOX/ROS and their role in hypoxia and thrombosis, and the important role of ROS scavengers to reduce COVID-19-related inflammation.
Collapse
Affiliation(s)
- Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Ranjit Sah
- Tribhuvan University Institute of Medicine, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Pharmaceutical and Health Service Research, Nepal Health Research and Innovation Foundation, Lalitpur, Nepal
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Japan
- Medical Governance Research Institute, Tokyo, Japan
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Zareena Fathah
- Kings College London, London, UK
- College of Medicine and Health Sciences, United Arab University, Abu Dhabi, United Arab Emirates
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- Institución Universitaria Visión de Las Americas, Pereira, Risaralda, Colombia.
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| |
Collapse
|
9
|
Zhou Z, Wang M, Huang C, Li Y, Gao L, Zhu Y, Ying C, Zhou X. Treadmill exercise training alleviates diabetes-induced depressive-like behavior and cognitive impairment by improving hippocampal CA1 neurons injury in db/db mice. Brain Res Bull 2022; 190:84-96. [PMID: 36174874 DOI: 10.1016/j.brainresbull.2022.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/12/2022] [Accepted: 09/23/2022] [Indexed: 11/28/2022]
Abstract
Patients with diabetes mellitus (DM) have an increased risk of diabetic encephalopathy symptoms such as depressive-like behaviour and cognitive impairment. Exercise is an effective strategy for preventing and treating DM and diabetic complications. The aim of this study is to investigate the effects and potential mechanisms of treadmill exercise training on diabetes-induced depressive-like behavior and cognitive impairment in db/db mice. In this study, the mice were divided into three groups (n=10 per group) as follows: healthy-sedentary (db/m), diabetes-sedentary (db/db), and diabetes-treadmill exercise training (db/db-TET). The db/db-TET mice were performed five days per week at a speed of 8m/min for 60min/day for 8 weeks, following which body weight, fasting blood glucose, insulin resistance, behavioral, synaptic ultrastructure, oxidative stress, apoptotic signaling, and inflammatory responses were evaluated. As a result, treadmill exercise training significantly decreased body weight and fasting blood glucose levels, increased insulin sensitivity, protected synaptic ultrastructure, reduced depression-like behavior, and improved learning and memory deficits in db/db mice. In addition, treadmill exercise training significantly suppressed NOX2-mediated oxidative stress, resulting in a decrease in NOX2-dependent ROS generation in the db/db mouse hippocampus CA1 region. Reduced ROS generation prevented the apoptotic signaling pathway and NLRP3 inflammasome activation, thereby ameliorating hippocampus neuronal damage. In summary, the results indicated that treadmill exercise training significantly ameliorates hippocampus injury by suppressing oxidative stress-induced apoptosis and NLRP3 inflammasome activation, consequently ameliorating diabetes-induced depressive-like behavior and cognitive impairment in db/db mice.
Collapse
Affiliation(s)
- Zhongyuan Zhou
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China; Department of Pain, Lianyungang Maternal and Child Health Hospital, Lianyungang, 222000, P.R.China
| | - Meng Wang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China
| | - Chengyu Huang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China
| | - Yan Li
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China
| | - Lin Gao
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China
| | - Yandong Zhu
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China
| | - Changjiang Ying
- Department of Endocrinology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R.China.
| | - Xiaoyan Zhou
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R.China.
| |
Collapse
|
10
|
Wang W, Zhang X, Lin L, Ren J, He R, Sun K. Inhibition of NADPH oxidase 2 (NOX2) reverses cognitive deficits by modulating excitability and excitatory transmission in the hippocampus after traumatic brain injury. Biochem Biophys Res Commun 2022; 617:1-7. [DOI: 10.1016/j.bbrc.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 04/30/2022] [Accepted: 05/01/2022] [Indexed: 11/16/2022]
|
11
|
Marín-Prida J, Liberato JL, Llópiz-Arzuaga A, Stringhetta-Padovani K, Pavón-Fuentes N, Leopoldino AM, Cruz OG, González IH, Pérez ML, Espuny AC, Santos WFDSD, Uyemura SA, Pardo-Andreu GL, Pentón-Rol G. Novel Insights into the Molecular Mechanisms Involved in the Neuroprotective Effects of C-Phycocyanin Against Brain Ischemia in Rats. Curr Pharm Des 2022; 28:1187-1197. [PMID: 35524676 DOI: 10.2174/1381612828666220506145542] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ischemic stroke produces a large health impact worldwide, with scarce therapeutic options. OBJECTIVE This study aimed to reveal the role of NADPH oxidase and neuroinflammatory genes on the cerebral anti-ischemic effects of C-Phycocyanin (C-PC), the chief biliprotein of Spirulina platensis. METHODS Rats with either focal cerebral ischemia/reperfusion (I/R) or acute brain hypoperfusion, received C-PC at different doses, or a vehicle, for up to 6 h post-stroke. Neurological, behavioral and histochemical parameters were assessed in I/R rats at 24 h. Cerebral gene expression and hippocampal neuron viability were evaluated in hypoperfused rats at acute (24 h) or chronic phases (30 days), respectively. A molecular docking analysis between NOX2 and C-PC-derived Phycocyanobilin (PCB) was also performed. RESULTS C-PC, obtained with a purity of 4.342, significantly reduced the infarct volume and neurologic deficit in a dose-dependent manner, and improved the exploratory activity of the I/R rats. This biliprotein inhibited NOX2 expression, a crucial NAPDH oxidase isoform in the brain, and the superoxide increase produced by the ischemic event. Moreover, C-PC-derived PCB showed a high binding affinity in silico with NOX2. C-PC downregulated the expression of pro-inflammatory genes (IFN-γ, IL-6, IL-17A, CD74, CCL12) and upregulated immune suppressive genes (Foxp3, IL-4, TGF-β) in hypoperfused brain areas. This compound also decreased chronic neuronal death in the hippocampus of hypoperfused rats. CONCLUSION These results suggest that the inhibition of cerebral NADPH oxidase and the improvement of neuroinflammation are key mechanisms mediating the neuroprotective actions of C-PC against brain ischemia.
Collapse
Affiliation(s)
- Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana
| | - José Luiz Liberato
- Faculty of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Brazil
| | | | - Karina Stringhetta-Padovani
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | | | - Andréia Machado Leopoldino
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | - Mariela León Pérez
- Isotopes Center, Ave. Monumental Km 3.5, San José de Las Lajas, Mayabeque, Cuba
| | - Antoni Camins Espuny
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | | | - Sergio Akira Uyemura
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | - Gilberto L Pardo-Andreu
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana, Cuba
| | - Giselle Pentón-Rol
- Center for Genetic Engineering and Biotechnology, Havana, Cuba.,Latin American School of Medicine, Playa, Havana, Cuba
| |
Collapse
|
12
|
Hendricks KS, To EE, Luong R, Liong F, Erlich JR, Shah AM, Liong S, O’Leary JJ, Brooks DA, Vlahos R, Selemidis S. Endothelial NOX4 Oxidase Negatively Regulates Inflammation and Improves Morbidity During Influenza A Virus Lung Infection in Mice. Front Cell Infect Microbiol 2022; 12:883448. [PMID: 35601109 PMCID: PMC9115386 DOI: 10.3389/fcimb.2022.883448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/07/2022] [Indexed: 12/02/2022] Open
Abstract
Endosomal NOX2 oxidase-dependent ROS production promotes influenza pathogenicity, but the role of NOX4 oxidase, which is highly expressed in the lung endothelium, is largely unknown. The aim of this study was to determine if endothelial NOX4 expression can influence viral pathology in vivo, using a mouse model of influenza infection. WT and transgenic endothelial NOX4 overexpressing mice (NOX4 TG) were infected intranasally with the Hong Kong H3N2 X-31 influenza A virus (104 PFU; HK x-31) or PBS control. Mice were culled at either 3 or 7 days post-infection to analyse: airway inflammation by bronchoalveolar lavage fluid (BALF) cell counts; NOX4, as well as inflammatory cytokine and chemokine gene expression by QPCR; and ROS production by an L-012-enhanced chemiluminescence assay. Influenza A virus infection of WT mice resulted in a significant reduction in lung NOX4 mRNA at day 3, which persisted until day 7, when compared to uninfected mice. Influenza A virus infection of NOX4 TG mice resulted in significantly less weight loss than that of WT mice at 3-days post infection. Viral titres were decreased in infected NOX4 TG mice compared to the infected WT mice, at both 3- and 7-days post infection and there was significantly less lung alveolitis, peri-bronchial inflammation and neutrophil infiltration. The oxidative burst from BALF inflammatory cells extracted from infected NOX4 TG mice was significantly less than that in the WT mice. Expression of macrophage and neutrophil chemoattractants CXCL10, CCL3, CXCL1 and CXCL2 in the lung tissue were significantly lower in NOX4 TG mice compared to the WT mice at 3-days post infection. We conclude that endothelial NOX4 oxidase is protective against influenza morbidity and is a potential target for limiting influenza A virus-induced lung inflammation.
Collapse
Affiliation(s)
- Keshia S. Hendricks
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Eunice E. To
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Raymond Luong
- Department of Pharmacology, Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Felicia Liong
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Jonathan R. Erlich
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Ajay M. Shah
- King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Stella Liong
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - John J. O’Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, St. James’s Hospital Dublin, Dublin, Ireland
| | - Doug A. Brooks
- Clinical and Health Sciences, Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, Science, Technology, Engineering and Mathematics (STEM) College, Royal Melbourne Institute of Technology (RMIT) University, Bundoora, VIC, Australia
| |
Collapse
|
13
|
Vesga-Jiménez DJ, Martin C, Barreto GE, Aristizábal-Pachón AF, Pinzón A, González J. Fatty Acids: An Insight into the Pathogenesis of Neurodegenerative Diseases and Therapeutic Potential. Int J Mol Sci 2022; 23:2577. [PMID: 35269720 PMCID: PMC8910658 DOI: 10.3390/ijms23052577] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
One of the most common lipids in the human body is palmitic acid (PA), a saturated fatty acid with essential functions in brain cells. PA is used by cells as an energy source, besides being a precursor of signaling molecules and protein tilting across the membrane. Although PA plays physiological functions in the brain, its excessive accumulation leads to detrimental effects on brain cells, causing lipotoxicity. This mechanism involves the activation of toll-like receptors (TLR) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, with the consequent release of pro-inflammatory cytokines, increased production of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy impairment. Importantly, some of the cellular changes induced by PA lead to an augmented susceptibility to the development of Alzheimer's and Parkinson´s diseases. Considering the complexity of the response to PA and the intrinsic differences of the brain, in this review, we provide an overview of the molecular and cellular effects of PA on different brain cells and their possible relationships with neurodegenerative diseases (NDs). Furthermore, we propose the use of other fatty acids, such as oleic acid or linoleic acid, as potential therapeutic approaches against NDs, as these fatty acids can counteract PA's negative effects on cells.
Collapse
Affiliation(s)
- Diego Julián Vesga-Jiménez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329, USA;
| | - Cynthia Martin
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329, USA;
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
- Health Research Institute, University of Limerick, V94 T9PX Limerick, Ireland
| | - Andrés Felipe Aristizábal-Pachón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogota 111321, Colombia;
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogota 110231, Colombia; (D.J.V.-J.); (A.F.A.-P.)
| |
Collapse
|
14
|
Kelmanson IV, Shokhina AG, Kotova DA, Pochechuev MS, Ivanova AD, Kostyuk AI, Panova AS, Borodinova AA, Solotenkov MA, Stepanov EA, Raevskii RI, Moshchenko AA, Pak VV, Ermakova YG, van Belle GJC, Tarabykin V, Balaban PM, Fedotov IV, Fedotov AB, Conrad M, Bogeski I, Katschinski DM, Doeppner TR, Bähr M, Zheltikov AM, Belousov VV, Bilan DS. In vivo dynamics of acidosis and oxidative stress in the acute phase of an ischemic stroke in a rodent model. Redox Biol 2021; 48:102178. [PMID: 34773835 PMCID: PMC8600061 DOI: 10.1016/j.redox.2021.102178] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023] Open
Abstract
Ischemic cerebral stroke is one of the leading causes of death and disability in humans. However, molecular processes underlying the development of this pathology remain poorly understood. There are major gaps in our understanding of metabolic changes that occur in the brain tissue during the early stages of ischemia and reperfusion. In particular, it is generally accepted that both ischemia (I) and reperfusion (R) generate reactive oxygen species (ROS) that cause oxidative stress which is one of the main drivers of the pathology, although ROS generation during I/R was never demonstrated in vivo due to the lack of suitable methods. In the present study, we record for the first time the dynamics of intracellular pH and H2O2 during I/R in cultured neurons and during experimental stroke in rats using the latest generation of genetically encoded biosensors SypHer3s and HyPer7. We detect a buildup of powerful acidosis in the brain tissue that overlaps with the ischemic core from the first seconds of pathogenesis. At the same time, no significant H2O2 generation was found in the acute phase of ischemia/reperfusion. HyPer7 oxidation in the brain was detected only 24 h later. Comparison of in vivo experiments with studies on cultured neurons under I/R demonstrates that the dynamics of metabolic processes in these models significantly differ, suggesting that a cell culture is a poor predictor of metabolic events in vivo.
Collapse
Affiliation(s)
- Ilya V Kelmanson
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Arina G Shokhina
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Daria A Kotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Matvei S Pochechuev
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Alexandra D Ivanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Biological Department, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Alexander I Kostyuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Anastasiya S Panova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Anastasia A Borodinova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Maxim A Solotenkov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Evgeny A Stepanov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia
| | - Roman I Raevskii
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Aleksandr A Moshchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117997, Russia
| | - Valeriy V Pak
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Yulia G Ermakova
- European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| | - Gijsbert J C van Belle
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Viktor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Ilya V Fedotov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia; Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, 420126, Russia; Department of Physics and Astronomy, Texas A&M University, College Station, TX, 77843, USA
| | - Andrei B Fedotov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia
| | - Marcus Conrad
- Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany
| | - Ivan Bogeski
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Dörthe M Katschinski
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, 37075, Germany; Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Istanbul, Turkey; Istanbul Medipol University, School of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - Aleksei M Zheltikov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia; Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, 420126, Russia; Department of Physics and Astronomy, Texas A&M University, College Station, TX, 77843, USA
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117997, Russia; Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany.
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| |
Collapse
|
15
|
Fang J, Sheng R, Qin ZH. NADPH Oxidases in the Central Nervous System: Regional and Cellular Localization and the Possible Link to Brain Diseases. Antioxid Redox Signal 2021; 35:951-973. [PMID: 34293949 DOI: 10.1089/ars.2021.0040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Significance: The significant role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) in signal transduction is mediated by the production of reactive oxygen species (ROS), especially in the central nervous system (CNS). The pathogenesis of some neurologic and psychiatric diseases is regulated by ROS, acting as a second messenger or pathogen. Recent Advances: In the CNS, the involvement of Nox-derived ROS has been implicated in the regulation of multiple signals, including cell survival/apoptosis, neuroinflammation, migration, differentiation, proliferation, and synaptic plasticity, as well as the integrity of the blood/brain barrier. In these processes, the intracellular signals mediated by the members of the Nox family vary among different tissues. The present review illuminates the regions and cellular, subcellular localization of Nox isoforms in the brain, the signal transduction, and the role of NOX enzymes in pathophysiology, respectively. Critical Issues: Different signal transduction cascades are coupled to ROS derived from various Nox homologues with varying degrees. Therefore, a critical issue worth noting is the varied role of the homologues of NOX enzymes in different signaling pathways and also they mediate different phenotypes in the diverse pathophysiological condition. This substantiates the effectiveness of selective Nox inhibitors in the CNS. Future Directions: Further investigation to elucidate the role of various homologues of NOX enzymes in acute and chronic brain diseases and signaling mechanisms, and the development of more specific NOX inhibitors for the treatment of CNS disease are urgently needed. Antioxid. Redox Signal. 35, 951-973.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
16
|
Hakami NY, Dusting GJ, Chan EC, Shah MH, Peshavariya HM. Wound Healing After Alkali Burn Injury of the Cornea Involves Nox4-Type NADPH Oxidase. Invest Ophthalmol Vis Sci 2021; 61:20. [PMID: 33079994 PMCID: PMC7585390 DOI: 10.1167/iovs.61.12.20] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Corneal injury that occurs after burning with alkali initiates wound-healing processes, including inflammation, neovascularization, and fibrosis. Excessive reactions to injury can reduce corneal transparency and thereby compromise vision. The NADPH oxidase (Nox) enzyme complex is known to be involved in cell signaling for wound-healing angiogenesis, but its role in corneal neovascularization has been little studied. Methods The center corneas of wild-type and Nox4 knockout (KO) mice were injured with 3 µL 1 M NaOH, while the contralateral corneas remained untouched. On day 7, mRNA expression levels of NADPH oxidase isoforms, the proangiogenic factors VEGF-A and TGFβ1, and proinflammatory genes ICAM-1 and VCAM-1 were determined. Corneal neovascularization and fibrosis were visualized using PECAM-1 antibody and picrosirius red staining, respectively, on the same day. Results Expressions of both Nox2 and Nox4 gene isoforms as well as the above genes were markedly increased in the injured corneas at 7 days. Injured corneas showed neovascularization and fibrosis as well as an increase in clinical opacity score. All responses stimulated by alkali burn were abrogated in Nox4 KO mice. Conclusions Nox4 could be a new target to treat pathologic corneal wound-healing responses and such targeting might prevent blindness caused by burn injuries.
Collapse
Affiliation(s)
- Nora Y Hakami
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia.,Faculty of Applied Medical Sciences, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Gregory J Dusting
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Elsa C Chan
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Manisha H Shah
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Hitesh M Peshavariya
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| |
Collapse
|
17
|
The Role of NADPH Oxidase in Neuronal Death and Neurogenesis after Acute Neurological Disorders. Antioxidants (Basel) 2021; 10:antiox10050739. [PMID: 34067012 PMCID: PMC8151966 DOI: 10.3390/antiox10050739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 01/22/2023] Open
Abstract
Oxidative stress is a well-known common pathological process involved in mediating acute neurological injuries, such as stroke, traumatic brain injury, epilepsy, and hypoglycemia-related neuronal injury. However, effective therapeutic measures aimed at scavenging free reactive oxygen species have shown little success in clinical trials. Recent studies have revealed that NADPH oxidase, a membrane-bound enzyme complex that catalyzes the production of a superoxide free radical, is one of the major sources of cellular reactive oxygen species in acute neurological disorders. Furthermore, several studies, including our previous ones, have shown that the inhibition of NADPH oxidase can reduce subsequent neuronal injury in neurological disease. Moreover, maintaining appropriate levels of NADPH oxidase has also been shown to be associated with proper neurogenesis after neuronal injury. This review aims to present a comprehensive overview of the role of NADPH oxidase in neuronal death and neurogenesis in multiple acute neurological disorders and to explore potential pharmacological strategies targeting the NADPH-related oxidative stress pathways.
Collapse
|
18
|
Pathophysiology and Therapeutic Potential of NADPH Oxidases in Ischemic Stroke-Induced Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6631805. [PMID: 33777315 PMCID: PMC7969100 DOI: 10.1155/2021/6631805] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/22/2021] [Accepted: 02/02/2021] [Indexed: 12/23/2022]
Abstract
Stroke is a leading cause of death and disability in humans. The excessive production of reactive oxygen species (ROS) is an important contributor to oxidative stress and secondary brain damage after stroke. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, an enzyme complex consisting of membrane subunits and cytoplasmic subunits, regulates neuronal maturation and cerebrovascular homeostasis. However, NADPH oxidase overproduction contributes to neurotoxicity and cerebrovascular disease. NADPH oxidase has been implicated as the principal source of ROS in the brain, and numerous studies have shown that the knockout of NADPH exerts a protective effect in the model of ischemic stroke. In this review, we summarize the mechanism of activation of the NADPH oxidase family members, the pathophysiological effects of NADPH oxidase isoforms in ischemic stroke, and the studies of NADPH oxidase inhibitors to explore potential clinical applications.
Collapse
|
19
|
Tan Y, Zhou F, Yang D, Zhang X, Zeng M, Wan L. MicroRNA-126a-5p Exerts Neuroprotective Effects on Ischemic Stroke via Targeting NADPH Oxidase 2. Neuropsychiatr Dis Treat 2021; 17:2089-2103. [PMID: 34234438 PMCID: PMC8242150 DOI: 10.2147/ndt.s293611] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/14/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Ischemic stroke is a destructive cerebrovascular disorder related to oxidative stress; NOX2 is a major source for ROS production; and miR-126a-5p is involved in several diseases, such as abdominal aortic aneurysm. We investigated the role of miR-126a-5p in regulating NOX2 in ischemic stroke. METHODS MiR-126a-5p and NOX2 were examined in the brains of rats subjected to cerebral ischemia/reperfusion (I/R) by RT-PCR and Western blot. MiR-126a-5p agomir was delivered to examine the effects of miR-126a-5p on I/R injury. The neurological deficit, infarct volume, and brain water content were evaluated. NOX activity, ROS production, and MDA and SOD levels were detected to assess oxidative stress. H&E staining was used to examine cell state. Apoptosis was evaluated by TUNEL, caspase-3 activity, and cleaved-caspase-3 protein level. The relationship between miR-126a-5p and NOX2 was analyzed by bioinformatics and luciferase reporter assay. MiR-126a-5p mimic, miR-126a-5p inhibitor, or pcDNA-NOX2 were transfected in SH-SY5Y cells to further assess the effects of miR-126a-5p on OGD/R-induced cells injury. RESULTS NOX2 was upregulated and miR-126a-5p was down-regulated in the brains of I/R rats. MiR-126a-5p agomir obviously reduced the neurological deficit, infarct volume, brain water content, oxidative stress, and apoptosis in I/R rats. MiR-126a-5p targeted NOX2 directly and regulated NOX2 negatively. Moreover, miR-126a-5p mimic elevated cell viability and inhibited oxidative stress and apoptosis in OGD/R-treated SH-SY5Y cells, while miR-126a-5p inhibitor had the opposite effects. NOX2 overexpression antagonized the protective effects of miR-126a-5p mimic on OGD/R-induced cell injury. CONCLUSION MiR-126a-5p is a novel potential target for ischemic stroke therapy due to its protection against cerebral I/R injury via directly targeting NOX2.
Collapse
Affiliation(s)
- Yu Tan
- Department of Neurology, The Third Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330008, People's Republic of China
| | - Feng Zhou
- Department of Neurology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai City, Guangdong Province, 519000, People's Republic of China
| | - Dejiang Yang
- Department of Neurology, The Third Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330008, People's Republic of China
| | - Xiaowei Zhang
- Department of Neurology, The Third Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330008, People's Republic of China
| | - Meihong Zeng
- Department of Neurology, The Third Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330008, People's Republic of China
| | - Lei Wan
- Department of Neurology, The Third Affiliated Hospital of Nanchang University, Nanchang City, Jiangxi Province, 330008, People's Republic of China
| |
Collapse
|
20
|
Hvozda Arana AG, Lasagni Vitar RM, Reides CG, Lerner SF, Ferreira SM. Glaucoma causes redox imbalance in the primary visual cortex by modulating NADPH oxidase-4, iNOS, and Nrf2 pathway in a rat experimental model. Exp Eye Res 2020; 200:108225. [DOI: 10.1016/j.exer.2020.108225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
|
21
|
Eastman CL, D'Ambrosio R, Ganesh T. Modulating neuroinflammation and oxidative stress to prevent epilepsy and improve outcomes after traumatic brain injury. Neuropharmacology 2020; 172:107907. [PMID: 31837825 PMCID: PMC7274911 DOI: 10.1016/j.neuropharm.2019.107907] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in young adults worldwide. TBI survival is associated with persistent neuropsychiatric and neurological impairments, including posttraumatic epilepsy (PTE). To date, no pharmaceutical treatment has been found to prevent PTE or ameliorate neurological/neuropsychiatric deficits after TBI. Brain trauma results in immediate mechanical damage to brain cells and blood vessels that may never be fully restored given the limited regenerative capacity of brain tissue. This primary insult unleashes cascades of events, prominently including neuroinflammation and massive oxidative stress that evolve over time, expanding the brain injury, but also clearing cellular debris and establishing homeostasis in the region of damage. Accumulating evidence suggests that oxidative stress and neuroinflammatory sequelae of TBI contribute to posttraumatic epileptogenesis. This review will focus on possible roles of reactive oxygen species (ROS), their interactions with neuroinflammation in posttraumatic epileptogenesis, and emerging therapeutic strategies after TBI. We propose that inhibitors of the professional ROS-generating enzymes, the NADPH oxygenases and myeloperoxidase alone, or combined with selective inhibition of cyclooxygenase mediated signaling may have promise for the treatment or prevention of PTE and other sequelae of TBI. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Clifford L Eastman
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA.
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA; Regional Epilepsy Center, University of Washington, 325 Ninth Ave., Seattle, WA, 98104, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, Georgia.
| |
Collapse
|
22
|
Dynamic Alterations of Brain Injury, Functional Recovery, and Metabolites Profile after Cerebral Ischemia/Reperfusion in Rats Contributes to Potential Biomarkers. J Mol Neurosci 2020; 70:667-676. [PMID: 31907865 DOI: 10.1007/s12031-019-01474-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/26/2019] [Indexed: 01/08/2023]
Abstract
Cerebral ischemia-reperfusion (I/R) is characterized by initial transient cerebral ischemia followed by reperfusion. Various pathophysiological processes are involved in brain injury and functional recovery during cerebral I/R. There are few studies on dynamic metabolic process after cerebral I/R. The present study was to observe dynamic alteration of brain injury, functional recovery, and metabolites after cerebral I/R in rats and discover potential metabolic markers. The cerebral I/R model was established by middle cerebral artery occlusion (MCAO) for 90 min, following reperfusion in rats. The results of cerebral infarction area, cerebral edema, and behavior test showed that there were dynamic changes in brain injury and functional recovery at different periods after cerebral I/R. Further analysis showed that the brain injury was severe on the first day of cerebral I/R, and there was a significant functional recovery from the 7th day of cerebral I/R, followed by an aggravation trend of brain injury from the days 7 to 28. Furthermore, Matrix-assisted laser desorption ionization mass spectrometry imaging analysis showed that the expression of ATP, glucose, and citric acid on 7th day was the highest during cerebral I/R, which indicated that energy metabolism and oxidative phosphorylation played important roles during cerebral I/R. In addition, the untargeted metabolomic results showed that the level of isocitric acid, the ratio of oxyglutaric acid/glutamic acid, and the level of pyruvic acid associated with the TCA cycle were also the highest on the 7th day during cerebral I/R, which indicated that the transient spontaneous recovery of ischemic brain on the 7th day after ischemia-reperfusion might be related to oxidative phosphorylation and energy metabolism in the brain in this period. In conclusion, the results suggest that some small molecule metabolites participate in the brain injury and functional recovery during cerebral I/R, which is of great significance to the development of therapeutic drugs and diagnostic markers.
Collapse
|
23
|
Wang J, Liu Y, Shen H, Li H, Wang Z, Chen G. Nox2 and Nox4 Participate in ROS-Induced Neuronal Apoptosis and Brain Injury During Ischemia-Reperfusion in Rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2020; 127:47-54. [PMID: 31407062 DOI: 10.1007/978-3-030-04615-6_8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previously studies have shown that Nox2 and Nox4, as members of nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase, Nox), participate in brain damage caused by ischemia-reperfusion (I/R). The aim of this study is to investigate the effects of specific chemical inhibitors of Nox2 and Nox4 on cerebral I/R-induced brain injury in rats. METHODS At 0.5 h before MCAO surgery, the rats were pretreated with vehicle, Nox2 inhibitor (gp91ds-tat), and Nox4 inhibitor (GKT137831), respectively. After reperfusion for 24 h, the infarct sizes of brain tissues in rats in various groups are determined. The penumbra (ischemic) tissues are collected to measure ROS levels, neuronal apoptosis, and degeneration, as well as the integrity of the blood-brain barrier (BBB) in brain tissues of rats. RESULTS gp91ds-tat and GKT137831 pretreatment significantly reduced the infarct sizes in brain tissues of rats, effectively suppressed I/R-induced increase in ROS levels, neuronal apoptosis and degeneration, and obviously alleviated BBB damage. CONCLUSION Under cerebral I/R conditions, Nox2 inhibitor (gp91ds-tat) and Nox4 inhibitor (GKT137831) can effectively play a protective role in the brain tissues of rats.
Collapse
Affiliation(s)
- Jinjin Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Neurosurgery, Jiangsu Shengze Hospital, Suzhou, China
| | - Yin Liu
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Neurosurgery, Suzhou Municipal Hospital, Suzhou, China
| | - Haitao Shen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haiying Li
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Gang Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Yang Q, Huang Q, Hu Z, Tang X. Potential Neuroprotective Treatment of Stroke: Targeting Excitotoxicity, Oxidative Stress, and Inflammation. Front Neurosci 2019; 13:1036. [PMID: 31611768 PMCID: PMC6777147 DOI: 10.3389/fnins.2019.01036] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 09/12/2019] [Indexed: 01/08/2023] Open
Abstract
Stroke is a major cause of death and adult disability. However, therapeutic options remain limited. Numerous pathways underlie acute responses of brain tissue to stroke. Early events following ischemic damage include reactive oxygen species (ROS)-mediated oxidative stress and glutamate-induced excitotoxicity, both of which contribute to rapid cell death within the infarct core. A subsequent cascade of inflammatory events escalates damage progression. This review explores potential neuroprotective strategies for targeting key steps in the cascade of ischemia–reperfusion (I/R) injury. NADPH oxidase (NOX) inhibitors and several drugs currently approved by the U.S. Food and Drug Administration including glucose-lowering agents, antibiotics, and immunomodulators, have shown promise in the treatment of stroke in both animal experiments and clinical trials. Ischemic conditioning, a phenomenon by which one or more cycles of a short period of sublethal ischemia to an organ or tissue protects against subsequent ischemic events in another organ, may be another potential neuroprotective strategy for the treatment of stroke by targeting key steps in the I/R injury cascade.
Collapse
Affiliation(s)
- Qianwen Yang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
25
|
Sabirzhanov B, Li Y, Coll-Miro M, Matyas JJ, He J, Kumar A, Ward N, Yu J, Faden AI, Wu J. Inhibition of NOX2 signaling limits pain-related behavior and improves motor function in male mice after spinal cord injury: Participation of IL-10/miR-155 pathways. Brain Behav Immun 2019; 80:73-87. [PMID: 30807841 PMCID: PMC6660361 DOI: 10.1016/j.bbi.2019.02.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/11/2019] [Accepted: 02/22/2019] [Indexed: 12/19/2022] Open
Abstract
NADPH oxidase (NOX2) is an enzyme that induces reactive oxygen species (ROS) and serves as a switch between the pro-inflammatory and neurorestorative microglial/macrophage phenotypes; such changes play an important role in neuropathic pain and motor dysfunction. Increased NOX2 expression after spinal cord injury (SCI) has been reported, and inhibition of NOX2 improves motor function. However, the underlying mechanisms of NOX2 in post-traumatic pain and motor deficit remain unexplored. In the present study, we report that depletion of NOX2 (NOX2-/-) or inhibition of NOX2 using NOX2ds-tat significantly reduced mechanical/thermal cutaneous hypersensitivity and motor dysfunction after moderate contusion SCI at T10 in male mice. Western blot (WB, 3 mm lesion area) and immunohistochemistry (IHC) showed that SCI elevates NOX2 expression predominantly in microglia/macrophages up to 8 weeks post-injury. Deletion of NOX2 significantly reduced CD11b+/CD45hiF4/80+ macrophage infiltration at 24 h post-injury detected by flow cytometry and 8-OHG+ ROS production at 8 weeks post-injury by IHC in both lesion area and lumbar enlargement. NOX2 deficiency also altered microglial/macrophage pro-inflammatory and anti-inflammatory balance towards the neurorestorative response. WB analysis showed robust increase of Arginase-1 and YM1 proteins in NOX2-/- mice. Furthermore, qPCR analysis showed significant up-regulation of anti-inflammatory cytokine IL-10 levels in NOX2-/- mice, associated with reduced microRNA-155 expression. These findings were confirmed in CD11b+ microglia/macrophages isolated from spinal cord at 3 days post-injury. Taken together, our data suggest an important role for IL-10/miR-155 pathway in regulating NOX2-mediated SCI-dysfunction. Thus, specific targeting of NOX2 may provide an effective strategy for treating neurological dysfunction in SCI patients.
Collapse
Affiliation(s)
- Boris Sabirzhanov
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Marino Coll-Miro
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Jessica J. Matyas
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Alok Kumar
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Nicole Ward
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Jingwen Yu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201 USA,University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201 USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201 USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201 USA.
| |
Collapse
|
26
|
Liu W, Huang J, Doycheva D, Gamdzyk M, Tang J, Zhang JH. RvD1binding with FPR2 attenuates inflammation via Rac1/NOX2 pathway after neonatal hypoxic-ischemic injury in rats. Exp Neurol 2019; 320:112982. [PMID: 31247196 DOI: 10.1016/j.expneurol.2019.112982] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/29/2019] [Accepted: 06/22/2019] [Indexed: 12/19/2022]
Abstract
Neuroinflammation plays a crucial role in the pathological development after neonatal hypoxia-ischemia (HI). Resolvin D1 (RvD1), an agonist of formyl peptide receptor 2 (FPR2), has been shown to exert anti-inflammatory effects in many diseases. The objective of this study was to explore the protective role of RvD1 through reducing inflammation after HI and to study the contribution of Ras-related C3 botulinum toxin substrate 1 (Rac1)/nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) pathways in RvD1-mediated protection. Rat pups (10-day old) were subjected to HI or sham surgery. RvD1 was administrated by intraperitoneal injection 1 h after HI. FPR2 small interfering ribonucleic acid (siRNA) and Rac1 activation CRISPR were administered prior to RvD1 treatment to elucidate the possible mechanisms. Time course expression of FPR2 by Western blot and RvD1 by ELISA were conducted at 6 h, 12 h, 24 h, 48 h and 72 h post HI. Infarction area, short-term neurological deficits, immunofluorescent staining and Western blot were conducted at 24 h post HI. Long-term neurological behaviors were evaluated at 4 weeks post HI. Endogenous expression levels of RvD1 decreased in time dependent manner while the expression of FPR2 increased after HI, peaking at 24 h post HI. Activation of FPR2, with RvD1, reduced percent infarction area, and alleviated short- and long-term neurological deficits. Administration of RvD1 attenuated inflammation after HI, while, either inhibition of FPR2 with siRNA or activation of Rac1 with CRISPR reversed those effects. Our results showed that RvD1 attenuated neuroinflammation through FPR2, which then interacted with Rac1/NOX2 signaling pathway, thereby reducing infarction area and alleviating neurological deficits after HI in neonatal rat pups. RvD1 may be a potential therapeutic approach to reduce inflammation after HI.
Collapse
Affiliation(s)
- Wei Liu
- Department of Physiology, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda, CA 92354, USA
| | - Juan Huang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda, CA 92354, USA; Institute of Neuroscience, Chongqing Medical University, Chongqing 40016, China
| | - Desislava Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda, CA 92354, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda, CA 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda, CA 92354, USA.
| |
Collapse
|
27
|
Bu J, Shi S, Wang HQ, Niu XS, Zhao ZF, Wu WD, Zhang XL, Ma Z, Zhang YJ, Zhang H, Zhu Y. Acacetin protects against cerebral ischemia-reperfusion injury via the NLRP3 signaling pathway. Neural Regen Res 2019; 14:605-612. [PMID: 30632500 PMCID: PMC6352603 DOI: 10.4103/1673-5374.247465] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acacetin (5,7-dihydroxy-4'-methoxyflavone), a potential neuroprotective agent, has an inhibitory effect on lipopolysaccharide-induced neuroinflammatory reactions. However, whether acacetin has an effect on inflammatory corpuscle 3 (NLRP3) after cerebral ischemia-reperfusion injury has not been fully determined. This study used an improved suture method to establish a cerebral ischemia-reperfusion injury model in C57BL/6 mice. After ischemia with middle cerebral artery occlusion for 1 hour, reperfusion with intraperitoneal injection of 25 mg/kg of acacetin (acacetin group) or an equal volume of saline (0.1 mL/10 g, middle cerebral artery occlusion group) was used to investigate the effect of acacetin on cerebral ischemia-reperfusion injury. Infarct volume and neurological function scores were determined by 2,3,5-triphenyltetrazolium chloride staining and the Zea-Longa scoring method. Compared with the middle cerebral artery occlusion group, neurological function scores and cerebral infarction volumes were significantly reduced in the acacetin group. To understand the effect of acacetin on microglia-mediated inflammatory response after cerebral ischemia-reperfusion injury, immunohistochemistry for the microglia marker calcium adapter protein ionized calcium-binding adaptor molecule 1 (Iba1) was examined in the hippocampus of ischemic brain tissue. In addition, tumor necrosis factor-α, interleukin-1β, and interleukin-6 expression in ischemic brain tissue of mice was quantified by enzyme-linked immunosorbent assay. Expression of Iba1, tumor necrosis factor-α, interleukin-1β and interleukin-6 was significantly lower in the acacetin group compared with the middle cerebral artery occlusion group. Western blot assay results showed that expression of Toll-like receptor 4, nuclear factor kappa B, NLRP3, procaspase-1, caspase-1, pro-interleukin-1β, and interleukin-1β were significantly lower in the acacetin group compared with the middle cerebral artery occlusion group. Our findings indicate that acacetin has a protective effect on cerebral ischemia-reperfusion injury, and its mechanism of action is associated with inhibition of microglia-mediated inflammation and the NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Juan Bu
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Shen Shi
- Laboratory Animal Research Center, Center for Disease Control and Prevention, Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Hui-Qin Wang
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xiao-Shan Niu
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zong-Feng Zhao
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Wei-Dong Wu
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xiao-Ling Zhang
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Zhi Ma
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yan-Jun Zhang
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Hui Zhang
- Clinical Research Center, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Yi Zhu
- Department of Neurology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
28
|
Shen J, Rastogi R, Geng X, Ding Y. Nicotinamide adenine dinucleotide phosphate oxidase activation and neuronal death after ischemic stroke. Neural Regen Res 2019; 14:948-953. [PMID: 30761998 PMCID: PMC6404502 DOI: 10.4103/1673-5374.250568] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a multisubunit enzyme complex that utilizes nicotinamide adenine dinucleotide phosphate to produce superoxide anions and other reactive oxygen species. Under normal circumstances, reactive oxygen species mediate a number of important cellular functions, including the facilitation of adaptive immunity. In pathogenic circumstances, however, excess reactive oxygen species generated by NOX promotes apoptotic cell death. In ischemic stroke, in particular, it has been shown that both NOX activation and derangements in glucose metabolism result in increased apoptosis. Moreover, recent studies have established that glucose, as a NOX substrate, plays a vital role in the pathogenesis of reperfusion injury. Thus, NOX inhibition has the potential to mitigate the deleterious impact of hyperglycemia on stroke. In this paper, we provide an overview of this research, coupled with a discussion of its implications for the development of NOX inhibition as a strategy for the treatment of ischemic stroke. Both inhibition using apocynin, as well as the prospect of developing more specific inhibitors based on what is now understood of the biology of NOX assembly and activation, will be highlighted in the course of our discussion.
Collapse
Affiliation(s)
- Jiamei Shen
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Radhika Rastogi
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
29
|
Bennett C, Mohammed F, Álvarez-Ciara A, Nguyen MA, Dietrich WD, Rajguru SM, Streit WJ, Prasad A. Neuroinflammation, oxidative stress, and blood-brain barrier (BBB) disruption in acute Utah electrode array implants and the effect of deferoxamine as an iron chelator on acute foreign body response. Biomaterials 2019; 188:144-159. [PMID: 30343257 PMCID: PMC6300159 DOI: 10.1016/j.biomaterials.2018.09.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023]
Abstract
The use of intracortical microelectrode arrays has gained significant attention in being able to help restore function in paralysis patients and study the brain in various neurological disorders. Electrode implantation in the cortex causes vasculature or blood-brain barrier (BBB) disruption and thus elicits a foreign body response (FBR) that results in chronic inflammation and may lead to poor electrode performance. In this study, a comprehensive insight into the acute molecular mechanisms occurring at the Utah electrode array-tissue interface is provided to understand the oxidative stress, neuroinflammation, and neurovascular unit (astrocytes, pericytes, and endothelial cells) disruption that occurs following microelectrode implantation. Quantitative real time polymerase chain reaction (qRT-PCR) was used to quantify the gene expression at acute time-points of 48-hr, 72-hr, and 7-days for factors mediating oxidative stress, inflammation, and BBB disruption in rats implanted with a non-functional 4 × 4 Utah array in the somatosensory cortex. During vascular disruption, free iron released into the brain parenchyma can exacerbate the FBR, leading to oxidative stress and thus further contributing to BBB degradation. To reduce the free iron released into the brain tissue, the effects of an iron chelator, deferoxamine mesylate (DFX), was also evaluated.
Collapse
Affiliation(s)
- Cassie Bennett
- Department of Biomedical Engineering, University of Miami, FL, USA
| | - Farrah Mohammed
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | | | | | - Suhrud M Rajguru
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, FL, USA.
| |
Collapse
|
30
|
Shan BS, Mogi M, Iwanami J, Bai HY, Kan-No H, Higaki A, Min LJ, Horiuchi M. Attenuation of stroke damage by angiotensin II type 2 receptor stimulation via peroxisome proliferator-activated receptor-gamma activation. Hypertens Res 2018; 41:839-848. [PMID: 30089862 DOI: 10.1038/s41440-018-0082-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/01/2018] [Accepted: 07/04/2018] [Indexed: 11/09/2022]
Abstract
The brain renin-angiotensin system plays a crucial role in ischemic stroke. It is known that stimulation of the angiotensin II type 2 (AT2) receptor protects against ischemic brain injury. We recently demonstrated that AT2 receptor stimulation by compound 21 (C21), a direct AT2 receptor agonist, inhibited vascular intimal proliferation with activation of peroxisome proliferator-activated receptor-gamma (PPAR-γ). However, whether direct AT2 receptor stimulation protects against ischemic brain injury via PPAR-γ activation is still unknown. 8-week-old male C57BL/6 J mice were subjected to middle cerebral artery (MCA) occlusion. 2 weeks before MCA occlusion, they were administered C21 with or without GW9662, a PPAR-γ antagonist. Neurologic deficit, ischemic size, superoxide anion, superoxide dismutase (SOD) activity, expression of NADPH subunits and blood brain barrier (BBB) stabilization were assessed 24 h after MCA occlusion. Cerebral blood flow (CBF) was measured in the core and periphery of the MCA territory before, immediately after, 1 h and 24 h after MCA occlusion. Treatment with C21 markedly decreased the neurologic deficit and ischemic size with an increase in CBF, SOD activity and BBB stabilization genes compared with the non-treated group. Co-administration of GW9662 partially attenuated this protective effect of C21 on neurologic deficit and ischemic size via an increase in superoxide anion production and a decrease of SOD activity and BBB stabilization genes, while GW9662 treatment alone had no significant effect on neurologic deficit and ischemic size. These results suggest that direct AT2 receptor stimulation has a preventive effect on stroke-induced brain injury partly due to activation of PPAR-γ.
Collapse
Affiliation(s)
- Bao-Shuai Shan
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| | - Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan.
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| | - Hui-Yu Bai
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| | - Harumi Kan-No
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| | - Akinori Higaki
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan.,Department of Cardiology, Pulmonology, Hypertension and Nephrology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| | - Li-Juan Min
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Matsuyama, Japan
| |
Collapse
|
31
|
Wang X, Jin C, Zhong Y, Li X, Han J, Xue W, Wu P, Xia X, Peng X. Glutathione Reduction of Patulin-Evoked Cytotoxicity in HEK293 Cells by the Prevention of Oxidative Damage and the Mitochondrial Apoptotic Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:7775-7785. [PMID: 29676913 DOI: 10.1021/acs.jafc.8b01212] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Patulin (PAT) is a mycotoxin frequently detected in moldy fruits and fruit products. This study investigated the protective role of glutathione (GSH), an antioxidant agent, against PAT-induced cytotoxicity and its potential mechanisms in HEK293 cells. The obtained results showed that the addition of GSH significantly increased cell viability and decreased apoptosis induced by PAT. Additionally, GSH decreased intracellular ROS and mitochondrial ROS overproduction, suppressed the decline of the mitochondrial membrane potential, and maintained cellular ATP contents. GSH prevented the impairment of mitochondrial oxidative-phosphorylation system and, especially, enhanced the mRNA and protein levels of electron-transport-chain complex III (UQCRC2) and complex V (ATP5, ATP6 and ATP8). Furthermore, GSH increased endogenous GSH contents; enhanced the antioxidant-enzyme activities of SOD, CAT, GR, and GPx; and modulated oxidative damage. These results suggest that GSH reduces PAT-induced cytotoxicity via inhibition of oxidative damage and the mitochondrial apoptotic pathway in HEK293 cells.
Collapse
Affiliation(s)
- Xiaorui Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University (BTBU) , Beijing 100048 , China
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Chengni Jin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University (BTBU) , Beijing 100048 , China
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Yujie Zhong
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Xuan Li
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Jiahui Han
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Wei Xue
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Peng Wu
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Xiaodong Xia
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| | - Xiaoli Peng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health , Beijing Technology and Business University (BTBU) , Beijing 100048 , China
- College of Food Science and Engineering , Northwest A&F University , 22 Xinong Road , Yangling , Shaanxi 712100 , China
| |
Collapse
|
32
|
Ma MW, Wang J, Dhandapani KM, Wang R, Brann DW. NADPH oxidases in traumatic brain injury - Promising therapeutic targets? Redox Biol 2018; 16:285-293. [PMID: 29571125 PMCID: PMC5952873 DOI: 10.1016/j.redox.2018.03.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Despite intense investigation, no neuroprotective agents for TBI have yet translated to the clinic. Recent efforts have focused on identifying potential therapeutic targets that underlie the secondary TBI pathology that evolves minutes to years following the initial injury. Oxidative stress is a key player in this complex cascade of secondary injury mechanisms and prominently contributes to neurodegeneration and neuroinflammation. NADPH oxidase (NOX) is a family of enzymes whose unique function is to produce reactive oxygen species (ROS). Human post-mortem and animal studies have identified elevated NOX2 and NOX4 levels in the injured brain, suggesting that these two NOXs are involved in the pathogenesis of TBI. In support of this, NOX2 and NOX4 deletion studies have collectively revealed that targeting NOX enzymes can reduce oxidative stress, attenuate neuroinflammation, promote neuronal survival, and improve functional outcomes following TBI. In addition, NOX inhibitor studies have confirmed these findings and demonstrated an extended critical window of efficacious TBI treatment. Finally, the translational potential, caveats, and future directions of the field are highlighted and discussed throughout the review.
Collapse
Affiliation(s)
- Merry W Ma
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ruimin Wang
- Department of Neurobiology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
33
|
Kahl A, Anderson CJ, Qian L, Voss H, Manfredi G, Iadecola C, Zhou P. Neuronal expression of the mitochondrial protein prohibitin confers profound neuroprotection in a mouse model of focal cerebral ischemia. J Cereb Blood Flow Metab 2018; 38:1010-1020. [PMID: 28714328 PMCID: PMC5999007 DOI: 10.1177/0271678x17720371] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The mitochondrial protein prohibitin (PHB) has emerged as an important modulator of neuronal survival in different injury modalities . We previously showed that viral gene transfer of PHB protects CA1 neurons from delayed neurodegeneration following transient forebrain ischemia through mitochondrial mechanisms. However, since PHB is present in all cell types, it is not known if its selective expression in neurons is protective, and if the protection occurs also in acute focal ischemic brain injury, the most common stroke type in humans. Therefore, we generated transgenic mice overexpressing human PHB1 specifically in neurons (PHB1 Tg). PHB1 Tg mice and littermate controls were subjected to transient middle cerebral artery occlusion (MCAo). Infarct volume and sensory-motor impairment were assessed three days later. Under the control of a neuronal promoter (CaMKIIα), PHB1 expression was increased by 50% in the forebrain and hippocampus in PHB1 Tg mice. The brain injury produced by MCAo was reduced by 63 ± 11% in PHB1 Tg mice compared to littermate controls. This reduction was associated with improved sensory-motor performance, suggesting that the salvaged brain remains functional. Approaches to enhance PHB expression may be useful to ameliorate the devastating impact of cerebral ischemia on the brain.
Collapse
Affiliation(s)
- Anja Kahl
- 1 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY, USA
| | - Corey J Anderson
- 1 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY, USA
| | - Liping Qian
- 1 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY, USA
| | - Henning Voss
- 2 Department of Radiology, Weill Cornell Medicine, NY, USA
| | - Giovanni Manfredi
- 1 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY, USA
| | - Costantino Iadecola
- 1 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY, USA
| | - Ping Zhou
- 1 Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY, USA
| |
Collapse
|
34
|
Abstract
Reactive oxygen species (ROS) are well known for their role in mediating both physiological and pathophysiological signal transduction. Enzymes and subcellular compartments that typically produce ROS are associated with metabolic regulation, and diseases associated with metabolic dysfunction may be influenced by changes in redox balance. In this review, we summarize the current literature surrounding ROS and their role in metabolic and inflammatory regulation, focusing on ROS signal transduction and its relationship to disease progression. In particular, we examine ROS production in compartments such as the cytoplasm, mitochondria, peroxisome, and endoplasmic reticulum and discuss how ROS influence metabolic processes such as proteasome function, autophagy, and general inflammatory signaling. We also summarize and highlight the role of ROS in the regulation metabolic/inflammatory diseases including atherosclerosis, diabetes mellitus, and stroke. In order to develop therapies that target oxidative signaling, it is vital to understand the balance ROS signaling plays in both physiology and pathophysiology, and how manipulation of this balance and the identity of the ROS may influence cellular and tissue homeostasis. An increased understanding of specific sources of ROS production and an appreciation for how ROS influence cellular metabolism may help guide us in the effort to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Daniel S Kikuchi
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Marina S Hernandes
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Qian Xu
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Kathy K Griendling
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA.
| |
Collapse
|
35
|
NADPH Oxidase-Related Pathophysiology in Experimental Models of Stroke. Int J Mol Sci 2017; 18:ijms18102123. [PMID: 29019942 PMCID: PMC5666805 DOI: 10.3390/ijms18102123] [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: 09/06/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/16/2022] Open
Abstract
Several experimental studies have indicated that nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox) exert detrimental effects on ischemic brain tissue; Nox-knockout mice generally exhibit resistance to damage due to experimental stroke following middle cerebral artery occlusion (MCAO). Furthermore, our previous MCAO study indicated that infarct size and blood-brain barrier breakdown are enhanced in mice with pericyte-specific overexpression of Nox4, relative to levels observed in controls. However, it remains unclear whether Nox affects the stroke outcome directly by increasing oxidative stress at the site of ischemia, or indirectly by modifying physiological variables such as blood pressure or cerebral blood flow (CBF). Because of technical problems in the measurement of physiological variables and CBF, it is often difficult to address this issue in mouse models due to their small body size; in our previous study, we examined the effects of Nox activity on focal ischemic injury in a novel congenic rat strain: stroke-prone spontaneously hypertensive rats with loss-of-function in Nox. In this review, we summarize the current literature regarding the role of Nox in focal ischemic injury and discuss critical issues that should be considered when investigating Nox-related pathophysiology in animal models of stroke.
Collapse
|
36
|
Chronic cerebral hypoperfusion alters amyloid-β peptide pools leading to cerebral amyloid angiopathy, microinfarcts and haemorrhages in Tg-SwDI mice. Clin Sci (Lond) 2017; 131:2109-2123. [PMID: 28667120 DOI: 10.1042/cs20170962] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/22/2017] [Accepted: 06/29/2017] [Indexed: 02/07/2023]
Abstract
Cerebral hypoperfusion is an early feature of Alzheimer's disease (AD) that influences the progression from mild cognitive impairment to dementia. Understanding the mechanism is of critical importance in the search for new effective therapies. We hypothesized that cerebral hypoperfusion promotes the accumulation of amyloid-β (Aβ) and degenerative changes in the brain and is a potential mechanism contributing to development of dementia. To address this, we studied the effects of chronic cerebral hypoperfusion induced by bilateral carotid artery stenosis on Aβ peptide pools in a transgenic mouse model of AD (transgenic mice with Swedish, Dutch and Iowa mutations in human amyloid precursor protein (APP) (Tg-SwDI)). Cerebrovascular integrity was characterized by quantifying the occurrence of microinfarcts and haemorrhages and compared with wild-type mice without Aβ. A significant increase in soluble Aβ peptides (Aβ40/42) was detected after 1 month of hypoperfusion in the parenchyma in parallel with elevated APP and APP proteolytic products. Following 3 months, a significant increase in insoluble Aβ40/42 was determined in the parenchyma and vasculature. Microinfarct load was significantly increased in the Tg-SwDI as compared with wild-type mice and further exacerbated by hypoperfusion at 1 and 3 months. In addition, the number of Tg-SwDI hypoperfused mice with haemorrhages was increased compared with hypoperfused wild-type mice. Soluble parenchymal Aβ was associated with elevated NADPH oxidase-2 (NOX2) which was exacerbated by 1-month hypoperfusion. We suggest that in response to hypoperfusion, increased Aβ production/deposition may contribute to degenerative processes by triggering oxidative stress promoting cerebrovascular disruption and the development of microinfarcts.
Collapse
|
37
|
Shanshan Y, Beibei J, Li T, Minna G, Shipeng L, Li P, Yong Z. Phospholipase A2 of Peroxiredoxin 6 Plays a Critical Role in Cerebral Ischemia/Reperfusion Inflammatory Injury. Front Cell Neurosci 2017; 11:99. [PMID: 28424593 PMCID: PMC5380807 DOI: 10.3389/fncel.2017.00099] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
Microglia-mediated inflammation is an important step in the progression of cerebral ischemia/reperfusion injury and the associated production of receptors of immunomoudulation, including Toll-like receptors (TLRs). Peroxiredoxin 6 (Prdx6) has been demonstrated as the endogenous antioxidant protein for its peroxidase properties. However, the role of the independent phospholipase A2 (iPLA2) activity of Prdx6 in stroke has not been well studied. In this study, we evaluated whether blocking the calcium-iPLA2 activity of Prdx6 using siRNA and inhibitors (1-hexadecyl-3-(trifluoroethgl)-sn-glycerol-2 phosphomethanol, MJ33) would have a critical effect on inflammatory brain damage. We conducted oxygen-glucose deprivation (OGD)/recovery (R) in vitro and middle cerebral artery occlusion (MCAO) in vivo in a microglia/neuron co-culture system and in rats. In vitro, we found that Prdx6-iPLA2 activity was associated with the secretion of neurotoxic inflammatory mediators interleukin1β (IL-1β), interleukin-17 (IL-17) and interleukin-23 (IL-23) and elevated expression of Toll-like receptor 2/4 (TLR2/4), leading to the formation of nuclear factor-kappa B (NF-κB), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in microglial cells. In vivo, combined treatment with Prdx6-iPLA2 activity inhibitor MJ33 showed a greater diminution in neurologic deficits, cerebral infarction, brain water content and inflammatory molecules than Prdx6-siRNA treatment alone. Our findings provide new insight into Prdx6-iPLA2 function in the brain. Inhibition of Prdx6-iPLA2 activity by gene therapy and/or pharmacology may constitute a promising new therapeutic approach to the treatment of stroke.
Collapse
Affiliation(s)
- Yu Shanshan
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Jiang Beibei
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Tan Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Gao Minna
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Lei Shipeng
- Department of Respiratory Medicine, Jiangjin Center HospitalChongqing, China
| | - Peng Li
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| | - Zhao Yong
- Department of Pathology, Chongqing Medical UniversityChongqing, China.,Molecular Medical Laboratory, Chongqing Medical UniversityChongqing, China.,Institute of Neuroscience, Chongqing Medical UniversityChongqing, China.,Key Laboratory of Neurobiology, Chongqing Medical UniversityChongqing, China
| |
Collapse
|
38
|
Zhiqun T, Jian Z, Junli Y, Chunzi W, Danju Z. Allelopathic effects of volatile organic compounds from Eucalyptus grandis rhizosphere soil on Eisenia fetida assessed using avoidance bioassays, enzyme activity, and comet assays. CHEMOSPHERE 2017; 173:307-317. [PMID: 28113065 DOI: 10.1016/j.chemosphere.2017.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 01/01/2017] [Accepted: 01/02/2017] [Indexed: 06/06/2023]
Abstract
Allelopathy has been identified as an underlying mechanism of detrimental environmental impacts within commercial plantations. Eucalyptus spp. are known to generate huge amounts of volatile organic compounds (VOCs) that can function as phytotoxins and thus inhibit other plants. In the present study, biochemical markers, including activities of acetylcholinesterase (AChE) and oxidative stress enzymes, such as superoxide dismutase (SOD) and glutathione S-transferase (GST), were assayed to assess changes in Eisenia fetida at the physiological level induced by different doses of VOCs as part of an acute toxicity test over 7 and 14-day exposures. In addition, the toxicities of VOCs were investigated using a soil avoidance test and comet assay. The results revealed that E. fetida exhibited significant avoidance behavior towards the highest concentrations of undecane, decane, 2,4-dimethyl heptane, and 2,2,4,6,6-pentametyl heptane. The tail DNA percentages were significantly increased for all experimental treatments relative to control. However, under the treatments of VOCs, Olive tail moment content and comet tail length also display an obvious increase compared to control, except for that of octane, undecane and decane treatments. As VOC concentrations and durations increased in the soil, activities of AChE, SOD, and GST were either stimulated or inhibited. Among the VOCs, decane, 2,4-dimethyl heptane, 2,2,4,6,6-pentamethyl heptane, and 2,4-di tert buyl phenol exerted stronger effects on enzymatic activities. In summary, VOCs in rhizosphere soils of E. grandis might exert a toxic impact on E. fetida, among which 2,4-dimethyl heptane, 2,2,4,6,6-pentamethyl heptane, and 2,4-di tert buyl phenol have the strongest effects.
Collapse
Affiliation(s)
- Tang Zhiqun
- Institute of Ecological Forestry, Sichuan Provincial Key Laboratory of Ecological Forestry Engineering, College of Forestry, Sichuan Agricultural University, Wenjiang 611130, China
| | - Zhang Jian
- Institute of Ecological Forestry, Sichuan Provincial Key Laboratory of Ecological Forestry Engineering, College of Forestry, Sichuan Agricultural University, Wenjiang 611130, China
| | - Yu Junli
- Institute of Ecological Forestry, Sichuan Provincial Key Laboratory of Ecological Forestry Engineering, College of Forestry, Sichuan Agricultural University, Wenjiang 611130, China
| | - Wang Chunzi
- Institute of Ecological Forestry, Sichuan Provincial Key Laboratory of Ecological Forestry Engineering, College of Forestry, Sichuan Agricultural University, Wenjiang 611130, China
| | - Zhang Danju
- Institute of Ecological Forestry, Sichuan Provincial Key Laboratory of Ecological Forestry Engineering, College of Forestry, Sichuan Agricultural University, Wenjiang 611130, China.
| |
Collapse
|
39
|
Barrett JP, Henry RJ, Villapol S, Stoica BA, Kumar A, Burns MP, Faden AI, Loane DJ. NOX2 deficiency alters macrophage phenotype through an IL-10/STAT3 dependent mechanism: implications for traumatic brain injury. J Neuroinflammation 2017; 14:65. [PMID: 28340575 PMCID: PMC5366128 DOI: 10.1186/s12974-017-0843-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/16/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND NADPH oxidase (NOX2) is an enzyme system that generates reactive oxygen species (ROS) in microglia and macrophages. Excessive ROS production is linked with neuroinflammation and chronic neurodegeneration following traumatic brain injury (TBI). Redox signaling regulates macrophage/microglial phenotypic responses (pro-inflammatory versus anti-inflammatory), and NOX2 inhibition following moderate-to-severe TBI markedly reduces pro-inflammatory activation of macrophages/microglia resulting in concomitant increases in anti-inflammatory responses. Here, we report the signaling pathways that regulate NOX2-dependent macrophage/microglial phenotype switching in the TBI brain. METHODS Bone marrow-derived macrophages (BMDMs) prepared from wildtype (C57Bl/6) and NOX2 deficient (NOX2-/-) mice were treated with lipopolysaccharide (LPS; 10 ng/ml), interleukin-4 (IL-4; 10 ng/ml), or combined LPS/IL-4 to investigate signal transduction pathways associated with macrophage activation using western immunoblotting and qPCR analyses. Signaling pathways and activation markers were evaluated in ipsilateral cortical tissue obtained from adult male wildtype and NOX2-/- mice that received moderate-level controlled cortical impact (CCI). A neutralizing anti-IL-10 approach was used to determine the effects of IL-10 on NOX2-dependent transitions from pro- to anti-inflammatory activation states. RESULTS Using an LPS/IL-4-stimulated BMDM model that mimics the mixed pro- and anti-inflammatory responses observed in the injured cortex, we show that NOX2-/- significantly reduces STAT1 signaling and markers of pro-inflammatory activation. In addition, NOX2-/- BMDMs significantly increase anti-inflammatory marker expression; IL-10-mediated STAT3 signaling, but not STAT6 signaling, appears to be critical in regulating this anti-inflammatory response. Following moderate-level CCI, IL-10 is significantly increased in microglia/macrophages in the injured cortex of NOX2-/- mice. These changes are associated with increased STAT3 activation, but not STAT6 activation, and a robust anti-inflammatory response. Neutralization of IL-10 in NOX2-/- BMDMs or CCI mice blocks STAT3 activation and the anti-inflammatory response, thereby demonstrating a critical role for IL-10 in regulating NOX2-dependent transitions between pro- and anti-inflammatory activation states. CONCLUSIONS These studies indicate that following TBI NOX2 inhibition promotes a robust anti-inflammatory response in macrophages/microglia that is mediated by the IL-10/STAT3 signaling pathway. Thus, therapeutic interventions that inhibit macrophage/microglial NOX2 activity may improve TBI outcomes by not only limiting pro-inflammatory neurotoxic responses, but also enhancing IL-10-mediated anti-inflammatory responses that are neuroprotective.
Collapse
Affiliation(s)
- James P Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Sonia Villapol
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, DC, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Alok Kumar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Mark P Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, DC, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA.
| |
Collapse
|
40
|
[Salidroside protects PC12 cells from H 2O 2-induced apoptosis via suppressing NOX2-ROS-MAPKs signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37. [PMID: 28219860 PMCID: PMC6779671 DOI: 10.3969/j.issn.1673-4254.2017.02.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To investigate the molecular mechanism by which salidroside protects PC12 cells from H2O2-induced apoptosis. METHODS PC12 cells cultured in DMEM supplemented with 10% horse serum and 5% fetal bovine serum were pretreated with different doses of salidroside for 2 h and then stimulated with H2O2 for different lengths of time. The expression levels of PARP and caspase 3 and the phosphorylation of p38, ERK and JNK were determined with Western blotting. The cell nuclear morphology was observed after DAPI staining. The production of ROS was detected using a ROS detection kit, and the levels of gp91phox and p47phox in the membrane and cytoplasm were detected by membrane-cytoplasm separation experiment; the binding between gp91phox and p47phox was assayed by coimmunoprecipitation experiment. RESULTS Salidroside dose-dependently suppressed cell apoptosis, lowered phosphorylation levels of p38, ERK and JNK, inhibited the production of ROS, reduced the binding between gp91phox and p47phox, and inhibited the activity of NOX2 in PC12 cells exposed to H2O2. CONCLUSION Salidroside protects PC12 cells from H2O2-induced apoptosis at least partly by suppressing NOX2-ROS-MAPKs signaling pathway.
Collapse
|
41
|
Ren L, Tao J, Chen H, Bian Y, Yang X, Chen G, Zhang X, Liang G, Wu W, Song Z, Wang Y. Myeloid differentiation protein 2-dependent mechanisms in retinal ischemia-reperfusion injury. Toxicol Appl Pharmacol 2017; 317:1-11. [DOI: 10.1016/j.taap.2017.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/29/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022]
|
42
|
Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK, Brann DW. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener 2017; 12:7. [PMID: 28095923 PMCID: PMC5240251 DOI: 10.1186/s13024-017-0150-7] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is a common denominator in the pathology of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as in ischemic and traumatic brain injury. The brain is highly vulnerable to oxidative damage due to its high metabolic demand. However, therapies attempting to scavenge free radicals have shown little success. By shifting the focus to inhibit the generation of damaging free radicals, recent studies have identified NADPH oxidase as a major contributor to disease pathology. NADPH oxidase has the primary function to generate free radicals. In particular, there is growing evidence that the isoforms NOX1, NOX2, and NOX4 can be upregulated by a variety of neurodegenerative factors. The majority of recent studies have shown that genetic and pharmacological inhibition of NADPH oxidase enzymes are neuroprotective and able to reduce detrimental aspects of pathology following ischemic and traumatic brain injury, as well as in chronic neurodegenerative disorders. This review aims to summarize evidence supporting the role of NADPH oxidase in the pathology of these neurological disorders, explores pharmacological strategies of targeting this major oxidative stress pathway, and outlines obstacles that need to be overcome for successful translation of these therapies to the clinic.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Medical Drive, San Antonio, TX, 78229, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA.
| |
Collapse
|
43
|
Rastogi R, Geng X, Li F, Ding Y. NOX Activation by Subunit Interaction and Underlying Mechanisms in Disease. Front Cell Neurosci 2017; 10:301. [PMID: 28119569 PMCID: PMC5222855 DOI: 10.3389/fncel.2016.00301] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NAPDH) oxidase (NOX) is an enzyme complex with the sole function of producing superoxide anion and reactive oxygen species (ROS) at the expense of NADPH. Vital to the immune system as well as cellular signaling, NOX is also involved in the pathologies of a wide variety of disease states. Particularly, it is an integral player in many neurological diseases, including stroke, TBI, and neurodegenerative diseases. Pathologically, NOX produces an excessive amount of ROS that exceed the body’s antioxidant ability to neutralize them, leading to oxidative stress and aberrant signaling. This prevalence makes it an attractive therapeutic target and as such, NOX inhibitors have been studied and developed to counter NOX’s deleterious effects. However, recent studies of NOX have created a better understanding of the NOX complex. Comprised of independent cytosolic subunits, p47-phox, p67-phox, p40-phox and Rac, and membrane subunits, gp91-phox and p22-phox, the NOX complex requires a unique activation process through subunit interaction. Of these subunits, p47-phox plays the most important role in activation, binding and translocating the cytosolic subunits to the membrane and anchoring to p22-phox to organize the complex for NOX activation and function. Moreover, these interactions, particularly that between p47-phox and p22-phox, are dependent on phosphorylation initiated by upstream processes involving protein kinase C (PKC). This review will look at these interactions between subunits and with PKC. It will focus on the interaction involving p47-phox with p22-phox, key in bringing the cytosolic subunits to the membrane. Furthermore, the implication of these interactions as a target for NOX inhibitors such as apocynin will be discussed as a potential avenue for further investigation, in order to develop more specific NOX inhibitors based on the inhibition of NOX assembly and activation.
Collapse
Affiliation(s)
- Radhika Rastogi
- Department of Neurosurgery, Wayne State University School of Medicine Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| | - Fengwu Li
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
44
|
|
45
|
Kumar A, Barrett JP, Alvarez-Croda DM, Stoica BA, Faden AI, Loane DJ. NOX2 drives M1-like microglial/macrophage activation and neurodegeneration following experimental traumatic brain injury. Brain Behav Immun 2016; 58:291-309. [PMID: 27477920 PMCID: PMC5067217 DOI: 10.1016/j.bbi.2016.07.158] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/20/2016] [Accepted: 07/27/2016] [Indexed: 12/16/2022] Open
Abstract
Following traumatic brain injury (TBI), activation of microglia and peripherally derived inflammatory macrophages occurs in association with tissue damage. This neuroinflammatory response may have beneficial or detrimental effects on neuronal survival, depending on the functional polarization of these cells along a continuum from M1-like to M2-like activation states. The mechanisms that regulate M1-like and M2-like activation after TBI are not well understood, but appear in part to reflect the redox state of the lesion microenvironment. NADPH oxidase (NOX2) is a critical enzyme system that generates reactive oxygen species in microglia/macrophages. After TBI, NOX2 is strongly up-regulated in M1-like, but not in M2-like polarized cells. Therefore, we hypothesized that NOX2 drives M1-like neuroinflammation and contributes to neurodegeneration and loss of neurological function after TBI. In the present studies we inhibited NOX2 activity using NOX2-knockout mice or the selective peptide inhibitor gp91ds-tat. We show that NOX2 is highly up-regulated in infiltrating macrophages after injury, and that NOX2 deficiency reduces markers of M1-like activation, limits tissue loss and neurodegeneration, and improves motor recovery after moderate-level control cortical injury (CCI). NOX2 deficiency also promotes M2-like activation after CCI, through increased IL-4Rα signaling in infiltrating macrophages, suggesting that NOX2 acts as a critical switch between M1- and M2-like activation states after TBI. Administration of gp91ds-tat to wild-type CCI mice starting at 24h post-injury reduces deficits in cognitive function and increased M2-like activation in the hippocampus. Collectively, our data indicate that increased NOX2 activity after TBI drives M1-like activation that contributes to inflammatory-mediated neurodegeneration, and that inhibiting this pathway provides neuroprotection, in part by altering M1-/M2-like balance towards the M2-like neuroinflammatory response.
Collapse
Affiliation(s)
- Alok Kumar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James P. Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dulce-Mariely Alvarez-Croda
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA,Posgrado en Neuroetologia, Universidad Veracruzana, Xalapa, Mexico,Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Mexico
| | - Bogdan A. Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA,Correspondence: David J. Loane PhD, Department of Anesthesiology, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD 21201. Tel: 410-706-5188 Fax: 410-706-1639,
| |
Collapse
|
46
|
Coucha M, Abdelsaid M, Li W, Johnson MH, Orfi L, El-Remessy AB, Fagan SC, Ergul A. Nox4 contributes to the hypoxia-mediated regulation of actin cytoskeleton in cerebrovascular smooth muscle. Life Sci 2016; 163:46-54. [PMID: 27558234 DOI: 10.1016/j.lfs.2016.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/12/2016] [Accepted: 08/19/2016] [Indexed: 11/29/2022]
Abstract
UNLABELLED Ischemia/reperfusion and the resulting oxidative/nitrative stress impair cerebral myogenic tone via actin depolymerization. While it is known that NADPH oxidase (Nox) family is a major source of vascular oxidative stress; the extent and mechanisms by which Nox activation contributes to actin depolymerization, and equally important, the relative role of Nox isoforms in this response is not clear. AIM To determine the role of Nox4 in hypoxia-mediated actin depolymerization and myogenic-tone impairment in cerebral vascular smooth muscle. MAIN METHODS Control and Nox4 deficient (siRNA knock-down) human brain vascular smooth muscle cells (HBVSMC) were exposed to 30-min hypoxia/45-min reoxygenation. Nox2, Nox4, inducible and neuronal nitric oxide synthase (iNOS and nNOS) and nitrotyrosine levels as well as F:G actin were determined. Myogenic-tone was measured using pressurized arteriography in middle cerebral artery isolated from rats subjected to sham, 30-min ischemia/45-min reperfusion or ex-vivo oxygen glucose deprivation in the presence and absence of Nox inhibitors. RESULTS Nox4 and iNOS expression were significantly upregulated following hypoxia or ischemia/reperfusion. Hypoxia augmented nitrotyrosine levels while reducing F actin. These effects were nullified by inhibiting nitration with epicatechin or pharmacological or molecular inhibition of Nox4. Ischemia/reperfusion impaired myogenic-tone, which was restored by the selective inhibition of Nox4. CONCLUSION Nox4 activation in VSMCs contributes to actin depolymerization after hypoxia, which could be the underlying mechanism for myogenic-tone impairment following ischemia/reperfusion.
Collapse
Affiliation(s)
- Maha Coucha
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Mohammed Abdelsaid
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States
| | - Weiguo Li
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States
| | | | | | - Azza B El-Remessy
- Charlie Norwood VA Medical Center, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Susan C Fagan
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Neurology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta University, United States; Departments of Physiology, Augusta University, United States; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, United States.
| |
Collapse
|
47
|
Dexmedetomidine Protects against Transient Global Cerebral Ischemia/Reperfusion Induced Oxidative Stress and Inflammation in Diabetic Rats. PLoS One 2016; 11:e0151620. [PMID: 26982373 PMCID: PMC4794239 DOI: 10.1371/journal.pone.0151620] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/01/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Transient global cerebral ischemia/reperfusion (I/R) is a major perioperative complication, and diabetes increases the response of oxidative stress and inflammation induced by I/R. The objective of this study was to determine the protective effect of dexmedetomidine against transient global cerebral ischemia/reperfusion induced oxidative stress and inflammation in diabetic rats. METHODS Sixty-four rats were assigned into four experimental groups: normoglycemia, normoglycemia + dexmedetomidine, hyperglycemia, and hyperglycemia + dexmedetomidine and all subsequent neurological examinations were evaluated by a blinded observer. Damage to the brain was histologically assessed using the TUNEL staining method while western blotting was used to investigate changes in the expression levels of apoptosis-related proteins as well as the microglia marker, ionized calcium-binding adapter molecule 1 (Iba1). Water content in the brain was also analyzed. In addition, hippocampal concentrations of malondialdehyde (MDA) and Nox2 (a member of the Nox family of NADPH oxidases), and the activity of superoxide dismutase and catalase were analyzed. Finally, changes in serum concentrations of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 were detected. RESULTS Results showed that diabetes increased brain water content, the number of apoptotic neurons, early neurological deficit scores, oxidative stress (MDA and Nox2) and inflammation (pro-inflammatory cytokines including TNF-α and IL-6) levels following transient global I/R injury, but that these symptoms were attenuated following administration of dexmedetomidine. CONCLUSIONS These findings suggest that dexmedetomidine can significantly alleviate damage resulting from I/R, and this mechanism may be related to a reduction in both oxidative stress and inflammation which is normally associated with I/R.
Collapse
|
48
|
Ingberg E, Dock H, Theodorsson E, Theodorsson A, Ström JO. Method parameters' impact on mortality and variability in mouse stroke experiments: a meta-analysis. Sci Rep 2016; 6:21086. [PMID: 26876353 PMCID: PMC4753409 DOI: 10.1038/srep21086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Although hundreds of promising substances have been tested in clinical trials,
thrombolysis currently remains the only specific pharmacological treatment for
ischemic stroke. Poor quality, e.g. low statistical power, in the preclinical
studies has been suggested to play an important role in these failures. Therefore,
it would be attractive to use animal models optimized to minimize unnecessary
mortality and outcome variability, or at least to be able to power studies more
exactly by predicting variability and mortality given a certain experimental setup.
The possible combinations of methodological parameters are innumerous, and an
experimental comparison of them all is therefore not feasible. As an alternative
approach, we extracted data from 334 experimental mouse stroke articles and, using a
hypothesis-driven meta-analysis, investigated the method parameters’
impact on infarct size variability and mortality. The use of Swiss and C57BL6 mice
as well as permanent occlusion of the middle cerebral artery rendered the lowest
variability of the infarct size while the emboli methods increased variability. The
use of Swiss mice increased mortality. Our study offers guidance for researchers
striving to optimize mouse stroke models.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Hua Dock
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Sweden
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden.,School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
49
|
Kumar A, Alvarez-Croda DM, Stoica BA, Faden AI, Loane DJ. Microglial/Macrophage Polarization Dynamics following Traumatic Brain Injury. J Neurotrauma 2015; 33:1732-1750. [PMID: 26486881 DOI: 10.1089/neu.2015.4268] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Activated microglia and macrophages exert dual beneficial and detrimental roles after central nervous system injury, which are thought to be due to their polarization along a continuum from a classical pro-inflammatory M1-like state to an alternative anti-inflammatory M2-like state. The goal of the present study was to analyze the temporal dynamics of microglia/macrophage polarization within the lesion micro-environment following traumatic brain injury (TBI) using a moderate-level controlled cortical impact (CCI) model in mice. We performed a detailed phenotypic analysis of M1- and M2-like polarized microglia/macrophages, as well as nicotinamide adenine dinucleotide phosphate oxidase (NOX2) expression, through 7 days post-injury using real-time polymerase chain reaction (qPCR), flow cytometry and image analyses. We demonstrated that microglia/macrophages express both M1- and M2-like phenotypic markers early after TBI, but the transient up-regulation of the M2-like phenotype was replaced by a predominant M1- or mixed transitional (Mtran) phenotype that expressed high levels of NOX2 at 7 days post-injury. The shift towards the M1-like and Mtran phenotype was associated with increased cortical and hippocampal neurodegeneration. In a follow up study, we administered a selective NOX2 inhibitor, gp91ds-tat, to CCI mice starting at 24 h post-injury to investigate the relationship between NOX2 and M1-like/Mtran phenotypes. Delayed gp91ds-tat treatment altered M1-/M2-like balance in favor of the anti-inflammatory M2-like phenotype, and significantly reduced oxidative damage in neurons at 7 days post-injury. Therefore, our data suggest that despite M1-like and M2-like polarized microglia/macrophages being activated after TBI, the early M2-like response becomes dysfunctional over time, resulting in development of pathological M1-like and Mtran phenotypes driven by increased NOX2 activity.
Collapse
Affiliation(s)
- Alok Kumar
- 1 Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| | - Dulce-Mariely Alvarez-Croda
- 1 Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland.,2 Posgrado en Neuroetologia, Universidad Veracruzana , Xalapa, Mexico
| | - Bogdan A Stoica
- 1 Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| | - Alan I Faden
- 1 Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| | - David J Loane
- 1 Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
50
|
Zhang F, Lin YA, Kannan S, Kannan RM. Targeting specific cells in the brain with nanomedicines for CNS therapies. J Control Release 2015; 240:212-226. [PMID: 26686078 DOI: 10.1016/j.jconrel.2015.12.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 12/12/2022]
Abstract
Treatment of Central Nervous System (CNS) disorders still remains a major clinical challenge. The Blood-Brain Barrier (BBB), known as the major hindrance, greatly limits therapeutics penetration into the brain. Moreover, even though some therapeutics can cross BBB based on their intrinsic properties or via the use of proper nanoscale delivery vehicles, their therapeutic efficacy is still often limited without the specific uptake of drugs by the cancer or disease-associated cells. As more studies have started to elucidate the pathological roles of major cells in the CNS (for example, microglia, neurons, and astrocytes) for different disorders, nanomedicines that can enable targeting of specific cells in these diseases may provide great potential to boost efficacy. In this review, we aim to briefly cover the pathological roles of endothelial cells, microglia, tumor-associated microglia/macrophage, neurons, astrocytes, and glioma in CNS disorders and to highlight the recent advances in nanomedicines that can target specific disease-associated cells. Furthermore, we summarized some strategies employed in nanomedicine to achieve specific cell targeting or to enhance the drug neuroprotective effects in the CNS. The specific targeting at the cellular level by nanotherapy can be a more precise and effective means not only to enhance the drug availability but also to reduce side effects.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.,Department of Material Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yi-An Lin
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins Medical Institutions, MD, 21287 USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|