1
|
Mohamud Yusuf A, Borbor M, Hussner T, Weghs C, Kaltwasser B, Pillath-Eilers M, Walkenfort B, Kolesnick R, Gulbins E, Hermann DM, Brockmeier U. Acid sphingomyelinase inhibition induces cerebral angiogenesis post-ischemia/reperfusion in an oxidative stress-dependent way and promotes endothelial survival by regulating mitochondrial metabolism. Cell Death Dis 2024; 15:650. [PMID: 39231943 PMCID: PMC11374893 DOI: 10.1038/s41419-024-06935-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Acid sphingomyelinase (ASM) inhibitors are widely used for the treatment of post-stroke depression. They promote neurological recovery in animal stroke models via neurorestorative effects. In a previous study, we found that antidepressants including amitriptyline, fluoxetine, and desipramine increase cerebral angiogenesis post-ischemia/reperfusion (I/R) in an ASM-dependent way. To elucidate the underlying mechanisms, we investigated the effects of the functional ASM inhibitor amitriptyline in two models of I/R injury, that is, in human cerebral microvascular endothelial hCMEC/D3 cells exposed to oxygen-glucose deprivation and in mice exposed to middle cerebral artery occlusion (MCAO). In addition to our earlier studies, we now show that amitriptyline increased mitochondrial reactive oxygen species (ROS) formation in hCMEC/D3 cells and increased ROS formation in the vascular compartment of MCAO mice. ROS formation was instrumental for amitriptyline's angiogenic effects. ROS formation did not result in excessive endothelial injury. Instead, amitriptyline induced a profound metabolic reprogramming of endothelial cells that comprised reduced endothelial proliferation, reduced mitochondrial energy metabolism, reduced endoplasmic reticulum stress, increased autophagy/mitophagy, stimulation of antioxidant responses and inhibition of apoptotic cell death. Specifically, the antioxidant heme oxygenase-1, which was upregulated by amitriptyline, mediated amitriptyline's angiogenic effects. Thus, heme oxygenase-1 knockdown severely compromised angiogenesis and abolished amitriptyline's angiogenic responses. Our data demonstrate that ASM inhibition reregulates a complex network of metabolic and mitochondrial responses post-I/R that contribute to cerebral angiogenesis without compromising endothelial survival.
Collapse
Affiliation(s)
- Ayan Mohamud Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mina Borbor
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tanja Hussner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carolin Weghs
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Britta Kaltwasser
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Matthias Pillath-Eilers
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Walkenfort
- Imaging Center Essen (Electron Microscopy), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | - Erich Gulbins
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Ulf Brockmeier
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2
|
Cheng C, Liu K, Shen F, Zhang J, Xie Y, Li S, Hou Y, Bai G. Astragaloside IV targets PRDX6, inhibits the activation of RAC subunit in NADPH oxidase 2 for oxidative damage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154795. [PMID: 37030053 DOI: 10.1016/j.phymed.2023.154795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Radix Astragali Mongolici, as a traditional Chinese medicine, is widely used in the treatment of qi deficiency, viral or bacterial infection, inflammation and cancer. Astragaloside IV (AST), a key active compound in Radix Astragali Mongolici, has been shown to reduce disease progression by inhibiting oxidative stress and inflammation. However, the specific target and mechanism of action of AST in improving oxidative stress are still unclear. PURPOSE This study aims to explore the target and mechanism of AST to improve oxidative stress, and to explain the biological process of oxidative stress. METHODS AST functional probes were designed to capture target proteins and combined with protein spectrum to analyze target proteins. Small molecule and protein interaction technologies were used to verify the mode of action, while computer dynamics simulation technology was used to analyze the site of interaction with the target protein. The pharmacological activity of AST in improving oxidative stress was evaluated in a mouse model of acute lung injury induced by LPS. Additionally, pharmacological and serial molecular biological approaches were used to explore the underlying mechanism of action. RESULTS AST inhibits PLA2 activity in PRDX6 by targeting the PLA2 catalytic triad pocket. This binding alters the conformation and structural stability of PRDX6 and interferes with the interaction between PRDX6 and RAC, hindering the activation of the RAC-GDI heterodimer. Inactivation of RAC prevents NOX2 maturation, attenuates superoxide anion production, and improves oxidative stress damage. CONCLUSION The findings of this research indicate that AST impedes PLA2 activity by acting on the catalytic triad of PRDX6. This, in turn, disrupts the interaction between PRDX6 and RAC, thereby hindering the maturation of NOX2 and diminishing the oxidative stress damage.
Collapse
Affiliation(s)
- Chuanjing Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Kaixin Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Fukui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Jinling Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China
| | - Yang Xie
- The Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Suyun Li
- The Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan province & Education Ministry of P.R., China, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| |
Collapse
|
3
|
Briyal S, Ranjan AK, Gulati A. Oxidative stress: A target to treat Alzheimer's disease and stroke. Neurochem Int 2023; 165:105509. [PMID: 36907516 DOI: 10.1016/j.neuint.2023.105509] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/01/2023] [Accepted: 03/05/2023] [Indexed: 03/13/2023]
Abstract
Oxidative stress has been established as a well-known pathological condition in several neurovascular diseases. It starts with increased production of highly oxidizing free-radicals (e.g. reactive oxygen species; ROS and reactive nitrogen species; RNS) and becomes too high for the endogenous antioxidant system to neutralize them, which results in a significantly disturbed balance between free-radicals and antioxidants levels and causes cellular damage. A number of studies have evidently shown that oxidative stress plays a critical role in activating multiple cell signaling pathways implicated in both progression as well as initiation of neurological diseases. Therefore, oxidative stress continues to remain a key therapeutic target for neurological diseases. This review discusses the mechanisms involved in reactive oxygen species (ROS) generation in the brain, oxidative stress, and pathogenesis of neurological disorders such as stroke and Alzheimer's disease (AD) and the scope of antioxidant therapies for these disorders.
Collapse
Affiliation(s)
- Seema Briyal
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA.
| | - Amaresh K Ranjan
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA
| | - Anil Gulati
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA; Pharmazz Inc. Research and Development, Willowbrook, IL, USA
| |
Collapse
|
4
|
Kadir RRA, Alwjwaj M, Rakkar K, Othman OA, Sprigg N, Bath PM, Bayraktutan U. Outgrowth Endothelial Cell Conditioned Medium Negates TNF-α-Evoked Cerebral Barrier Damage: A Reverse Translational Research to Explore Mechanisms. Stem Cell Rev Rep 2023; 19:503-515. [PMID: 36056287 PMCID: PMC9902316 DOI: 10.1007/s12015-022-10439-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2022] [Indexed: 02/07/2023]
Abstract
Improved understanding of the key mechanisms underlying cerebral ischemic injury is essential for the discovery of efficacious novel therapeutics for stroke. Through detailed analysis of plasma samples obtained from a large number of healthy volunteers (n = 90) and ischemic stroke patients (n = 81), the current study found significant elevations in the levels of TNF-α at baseline (within the first 48 h of stroke) and on days 7, 30, 90 after ischaemic stroke. It then assessed the impact of this inflammatory cytokine on an in vitro model of human blood-brain barrier (BBB) and revealed dramatic impairments in both barrier integrity and function, the main cause of early death after an ischemic stroke. Co-treatment of BBB models in similar experiments with outgrowth endothelial cell-derived conditioned media (OEC-CM) negated the deleterious effects of TNF-α on BBB. Effective suppression of anti-angiogenic factor endostatin, stress fiber formation, oxidative stress, and apoptosis along with concomitant improvements in extracellular matrix adhesive and tubulogenic properties of brain microvascular endothelial cells and OECs played an important role in OEC-CM-mediated benefits. Significant increases in pro-angiogenic endothelin-1 and monocyte chemoattractant protein-1 in OEC-CM compared to the secretomes of OEC and HBMEC, detected by proteome profiling assay, accentuate the beneficial effects of OEC-CM. In conclusion, this reverse translational study identifies TNF-α as an important mediator of post-ischemic cerebral barrier damage and proposes OEC-CM as a potential vasculoprotective therapeutic strategy by demonstrating its ability to regulate a wide range of mechanisms associated with BBB function. Clinical trial registration NCT02980354.
Collapse
Affiliation(s)
- Rais Reskiawan A Kadir
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Mansour Alwjwaj
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Kamini Rakkar
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Othman Ahmad Othman
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Nikola Sprigg
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Philip M Bath
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, Clinical Sciences Building, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
5
|
Kadir RRA, Alwjwaj M, Bayraktutan U. Treatment with outgrowth endothelial cells protects cerebral barrier against ischemic injury. Cytotherapy 2022; 24:489-499. [PMID: 35183443 DOI: 10.1016/j.jcyt.2021.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS We have previously reported that outgrowth endothelial cells (OECs) restore cerebral endothelial cell integrity through effective homing to the injury site. This study further investigates whether treatment with OECs can restore blood-brain barrier (BBB) function in settings of ischemia-reperfusion injury both in vitro and in vivo. METHODS An in vitro model of human BBB was established by co-culture of astrocytes, pericytes, and human brain microvascular endothelial cells (HBMECs) before exposure to oxygen-glucose deprivation alone or followed by reperfusion (OGD±R) in the absence or presence of exogenous OECs. Using a rodent model of middle cerebral artery occlusion (MCAO), we further assessed the therapeutic potential of OECs in vivo. RESULTS Owing to their prominent antioxidant, proliferative, and migratory properties, alongside their inherent capacity to incorporate into brain vasculature, treatments with OECs attenuated the extent of OGD±R injury on BBB integrity and function, as ascertained by increases in transendothelial electrical resistance and decreases in paracellular flux across the barrier. Similarly, intravenous delivery of OECs also led to better barrier protection in MCAO rats as evidenced by significant decreases in ipsilateral brain edema volumes on day 3 after treatment. Mechanistic studies subsequently showed that treatment with OECs substantially reduced oxidative stress and apoptosis in HBMECs subjected to ischemic damages. CONCLUSION This experimental study shows that OEC-based cell therapy restores BBB integrity in an effective manner by integrating into resident cerebral microvascular network, suppressing oxidative stress and cellular apoptosis.
Collapse
Affiliation(s)
- Rais Reskiawan A Kadir
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Mansour Alwjwaj
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Nottingham, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Nottingham, UK.
| |
Collapse
|
6
|
Nakka VP, Gogada R, Simhadri PK, Qadeer MA, Phanithi PB. Post-treatment with apocynin at a lower dose regulates the UPR branch of eIF2α and XBP-1 pathways after stroke. Brain Res Bull 2022; 182:1-11. [PMID: 35143926 DOI: 10.1016/j.brainresbull.2022.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 11/18/2022]
Abstract
Stroke leads to disturbance in the physiology of the ER (Endoplasmic Reticulum) that triggers UPR (Unfolded Protein Response) pathways aimed to compensate neuronal cell damage. However, sustained UPR causes stressful conditions in the ER lumen forming abnormal protein aggregates. Stroke-induced oxidative stress also amalgamates with UPR to safeguard and ensure the proper functioning of brain cells. Thus we tested the effect of apocynin (a potent antioxidant) post-treatment in experimental stroke on the outcome of ER stress and UPR branch pathways. We administered a low dose of apocynin at 1 mg/kg (intraperitoneal) to adult Sprague-Dawley rats subjected to Middle Cerebral Artery Occlusion (MCAO) for two-time points. The first dose immediately after re-establishing the blood flow and another at 6 h of reperfusion. Apocynin post-treatment significantly reduced ROS (Reactive Oxygen Species) generation at an early reperfusion time point of 4 h. It preserved neuronal morphology, dendritic spine density, reduced protein aggregation, and brain damage after 24 h of reperfusion. Apocynin post-treatment regulates the two UPR branch pathways in our experimental paradigm. 1) Down-regulation of eIF2α (Eukaryotic Initiation Factor 2α) phosphorylation, and CHOP (C/EBP homologous protein) 2) by reducing the XBP-1 (X-Box binding Protein-1) mRNA splicing downstream to PERK (Protein Kinase RNA-Like ER Kinase) and IRE1α (Inositol Requiring Enzyme 1alpha) UPR pathways, respectively. Bioinformatics prediction showed that apocynin has binding sites for PERK (Protein Kinase RNA-Like ER Kinase) and IRE1α proteins. The amino acid residues interacting with apocynin were Cys891 and Gln889 (for PERK), and the amino acids Ser726, Arg722, and Ala719 (for IRE1α) lying within their activation loop. Overall, these studies indicate that apocynin post-treatment might regulate ER stress/UPR pathways and minimize stroke brain damage, thus having implications for developing newer strategies for stroke treatment.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India; Department of Biochemistry, Acharya Nagarjuna University, Andhra Pradesh 522510, India
| | - Raghu Gogada
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India; Department of Biochemistry and Plant Physiology, M.S. Swaminathan School of Agriculture, Centurion University of Technology and Management, Odisha 761211, India
| | - Praveen Kumar Simhadri
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India
| | | | - Prakash Babu Phanithi
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, 500046, India.
| |
Collapse
|
7
|
Kumar SP, Babu PP. NADPH Oxidase: a Possible Therapeutic Target for Cognitive Impairment in Experimental Cerebral Malaria. Mol Neurobiol 2021; 59:800-820. [PMID: 34782951 DOI: 10.1007/s12035-021-02598-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022]
Abstract
Long-term cognitive impairment associated with seizure-induced hippocampal damage is the key feature of cerebral malaria (CM) pathogenesis. One-fourth of child survivors of CM suffer from long-lasting neurological deficits and behavioral anomalies. However, mechanisms on hippocampal dysfunction are unclear. In this study, we elucidated whether gp91phox isoform of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) (a potent marker of oxidative stress) mediates hippocampal neuronal abnormalities and cognitive dysfunction in experimental CM (ECM). Mice symptomatic to CM were rescue treated with artemether monotherapy (ARM) and in combination with apocynin (ARM + APO) adjunctive based on scores of Rapid Murine Come behavior Scale (RMCBS). After a 30-day survivability period, we performed Barnes maze, T-maze, and novel object recognition cognitive tests to evaluate working and reference memory in all the experimental groups except CM. Sensorimotor tests were conducted in all the cohorts to assess motor coordination. We performed Golgi-Cox staining to illustrate cornu ammonis-1 (CA1) pyramidal neuronal morphology and study overall hippocampal neuronal density changes. Further, expression of NOX2, NeuN (neuronal marker) in hippocampal CA1 and dentate gyrus was determined using double immunofluorescence experiments in all the experimental groups. Mice administered with ARM monotherapy and APO adjunctive treatment exhibited similar survivability. The latter showed better locomotor and cognitive functions, reduced ROS levels, and hippocampal NOX2 immunoreactivity in ECM. Our results show a substantial increase in hippocampal NeuN immunoreactivity and dendritic arborization in ARM + APO cohorts compared to ARM-treated brain samples. Overall, our study suggests that overexpression of NOX2 could result in loss of hippocampal neuronal density and dendritic spines of CA1 neurons affecting the spatial working and reference memory during ECM. Notably, ARM + APO adjunctive therapy reversed the altered neuronal morphology and oxidative damage in hippocampal neurons restoring long-term cognitive functions after CM.
Collapse
Affiliation(s)
- Simhadri Praveen Kumar
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Phanithi Prakash Babu
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
8
|
Role of NADPH Oxidase-Induced Hypoxia-Induced Factor-1 α Increase in Blood-Brain Barrier Disruption after 2-Hour Focal Ischemic Stroke in Rat. Neural Plast 2021; 2021:9928232. [PMID: 34434231 PMCID: PMC8382561 DOI: 10.1155/2021/9928232] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/01/2021] [Indexed: 11/18/2022] Open
Abstract
We recently showed that inhibition of hypoxia-induced factor-1α (HIF-1α) decreased acute ischemic stroke-induced blood-brain barrier (BBB) damage. However, factors that induce the upregulation of HIF-1α expression remain unclear. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase played a critical role in reperfusion-induced BBB damage after stroke. However, the role of NADPH oxidase in BBB injury during the acute ischemia stage remains unclear. This study is aimed at investigating the role of NADPH oxidase in BBB injury and the expression of HIF-1α after acute ischemic stroke. A sutured middle cerebral artery occlusion (MCAO) model was used to mimic ischemic stroke in rats. Our results show that the inhibition of NADPH oxidase by apocynin can significantly reduce the BBB damage caused by 2 h ischemic stroke accompanied by reducing the degradation of tight junction protein occludin. In addition, treatment with apocynin significantly decreased the upregulation of HIF-1α induced by 2 h MCAO. More importantly, apocynin could also inhibit the MMP-2 upregulation. Of note, HIF-1α was not colocalized with a bigger blood vessel. Taken together, our results showed that inhibition of NADPH oxidase-mediated HIF-1α upregulation reduced BBB damage accompanied by downregulating MMP-2 expression and occludin degradation after 2 h ischemia stroke. These results explored the mechanism of BBB damage after acute ischemic stroke and may help reduce the associated cerebral hemorrhage transformation after thrombolysis and endovascular treatment after ischemic stroke.
Collapse
|
9
|
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: 15] [Impact Index Per Article: 3.8] [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
|
10
|
Hou K, Li G, Yu J, Xu K, Wu W. Receptors, Channel Proteins, and Enzymes Involved in Microglia-mediated Neuroinflammation and Treatments by Targeting Microglia in Ischemic Stroke. Neuroscience 2021; 460:167-180. [PMID: 33609636 DOI: 10.1016/j.neuroscience.2021.02.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022]
Abstract
Stroke is the largest contributor to global neurological disability-adjusted life-years, posing a huge economic and social burden to the world. Though pharmacological recanalization with recombinant tissue plasminogen activator and mechanical thrombectomy have greatly improved the prognosis of patients with ischemic stroke, clinically, there is still no effective treatment for the secondary injury caused by cerebral ischemia. In recent years, more and more evidences show that neuroinflammation plays a pivotal role in the pathogenesis and progression of ischemic cerebral injury. Microglia are brain resident innate immune cells and act the role peripheral macrophages. They play critical roles in mediating neuroinflammation after ischemic stroke. Microglia-mediated neuroinflammation is not an isolated process and has complex relationships with other pathophysiological processes as oxidative/nitrative stress, excitotoxicity, necrosis, apoptosis, pyroptosis, autophagy, and adaptive immune response. Upon activation, microglia differentially express various receptors, channel proteins, and enzymes involved in promoting or inhibiting the inflammatory processes, making them the targets of intervention for ischemic stroke. To inhibit microglia-related neuroinflammation and promote neurological recovery after ischemic stroke, numerous biochemical agents, cellular therapies, and physical methods have been demonstrated to have therapeutic potentials. Though accumulating experimental evidences have demonstrated that targeting microglia is a promising approach in the treatment of ischemic stroke, the clinical progress is slow. Till now, no clinical study could provide convincing evidence that any biochemical or physical therapies could exert neuroprotective effect by specifically targeting microglia following ischemic stroke.
Collapse
Affiliation(s)
- Kun Hou
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Guichen Li
- Department of Neurology, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Jinlu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Kan Xu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| | - Wei Wu
- Department of Neurosurgery, The First Hospital of Jilin University, 1 Xinmin Avenue, 130021 Changchun, China.
| |
Collapse
|
11
|
Zhang SR, Phan TG, Sobey CG. Targeting the Immune System for Ischemic Stroke. Trends Pharmacol Sci 2020; 42:96-105. [PMID: 33341247 DOI: 10.1016/j.tips.2020.11.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
Stroke is responsible for almost 6 million deaths and more than 10% of all mortalities each year, and two-thirds of stroke survivors remain disabled. With treatments for ischemic stroke still limited to clot lysis and/or mechanical removal, new therapeutic targets are desperately needed. In this review, we provide an overview of the complex mechanisms of innate and adaptive immune cell-mediated inflammatory injury, that exacerbates infarct development for several days after stroke. We also highlight the features of poststroke systemic immunodepression that commonly leads to infections and some mortalities, and argue that safe and effective therapies will need to balance pro- and anti-inflammatory mechanisms in a time-sensitive manner, to maximize the likelihood of an improved long-term outcome.
Collapse
Affiliation(s)
- Shenpeng R Zhang
- Department of Physiology, Anatomy, and Microbiology, and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Thanh G Phan
- Clinical Trials, Imaging, and Informatics (CTI) Division, Stroke and Ageing Research (STARC), Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy, and Microbiology, and Centre for Cardiovascular Biology and Disease Research, School of Life Sciences, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
12
|
Vemuganti R, Arumugam TV. Molecular Mechanisms of Intermittent Fasting-induced Ischemic Tolerance. CONDITIONING MEDICINE 2020; 3:9-17. [PMID: 34278242 PMCID: PMC8281895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Diet is a significant factor in determining human well-being. Excessive eating and/or diets with higher than needed amounts of carbohydrates, salt, and fat are known to cause metabolic disorders and functional changes in the body. To compensate the ill effects, many designer diets including the Mediterranean diet, the Okinawa diet, vegetarian/vegan diets, keto diet, anti-inflammatory diet, and the anti-oxidant diet have been introduced in the past 2 decades. While these diets are either enriched or devoid of one or more specific components, a better way to control diet is to limit the amount of food consumed. Caloric restriction (CR), which involves limiting the amount of food consumed rather than eliminating any specific type of food, as well as intermittent fasting (IF), which entails limiting the time during which food can be consumed on a given day, have gained popularity because of their positive effects on human health. While the molecular mechanisms of these 2 dietary regimens have not been fully deciphered, they are known to prolong the life span, control blood pressure, and blood glucose levels. Furthermore, CR and IF were both shown to decrease the incidence of heart attack and stroke, as well as their ill effects. In particular, IF is thought to promote metabolic switching by altering gene expression profiles leading to reduced inflammation and oxidative stress, while increasing plasticity and regeneration.
Collapse
Affiliation(s)
- Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- William S. Middleton VA Hospital, Madison, WI, USA
| | | |
Collapse
|
13
|
Liao R, Wood TR, Nance E. Nanotherapeutic modulation of excitotoxicity and oxidative stress in acute brain injury. Nanobiomedicine (Rij) 2020; 7:1849543520970819. [PMID: 35186151 PMCID: PMC8855450 DOI: 10.1177/1849543520970819] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
Excitotoxicity is a primary pathological process that occurs during stroke, traumatic brain injury (TBI), and global brain ischemia such as perinatal asphyxia. Excitotoxicity is triggered by an overabundance of excitatory neurotransmitters within the synapse, causing a detrimental cascade of excessive sodium and calcium influx, generation of reactive oxygen species, mitochondrial damage, and ultimately cell death. There are multiple potential points of intervention to combat excitotoxicity and downstream oxidative stress, yet there are currently no therapeutics clinically approved for this specific purpose. For a therapeutic to be effective against excitotoxicity, the therapeutic must accumulate at the disease site at the appropriate concentration at the right time. Nanotechnology can provide benefits for therapeutic delivery, including overcoming physiological obstacles such as the blood-brain barrier, protect cargo from degradation, and provide controlled release of a drug. This review evaluates the use of nano-based therapeutics to combat excitotoxicity in stroke, TBI, and hypoxia-ischemia with an emphasis on mitigating oxidative stress, and consideration of the path forward toward clinical translation.
Collapse
Affiliation(s)
- Rick Liao
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Thomas R Wood
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA, USA
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
- Center on Human Development and Disability, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Klimova N, Fearnow A, Long A, Kristian T. NAD + precursor modulates post-ischemic mitochondrial fragmentation and reactive oxygen species generation via SIRT3 dependent mechanisms. Exp Neurol 2019; 325:113144. [PMID: 31837320 DOI: 10.1016/j.expneurol.2019.113144] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/23/2022]
Abstract
Global cerebral ischemia depletes brain tissue NAD+, an essential cofactor for mitochondrial and cellular metabolism, leading to bioenergetics failure and cell death. The post-ischemic NAD+ levels can be replenished by the administration of nicotinamide mononucleotide (NMN), which serves as a precursor for NAD+ synthesis. We have shown that NMN administration shows dramatic protection against ischemic brain damage and inhibits post-ischemic hippocampal mitochondrial fragmentation. To understand the mechanism of NMN-induced modulation of mitochondrial dynamics and neuroprotection we used our transgenic mouse models that express mitochondria targeted yellow fluorescent protein in neurons (mito-eYFP) and mice that carry knockout of mitochondrial NAD+-dependent deacetylase sirt3 gene (SIRT3KO). Following ischemic insult, the mitochondrial NAD+ levels were depleted leading to an increase in mitochondrial protein acetylation, high reactive oxygen species (ROS) production, and excessive mitochondrial fragmentation. Administration of a single dose of NMN normalized hippocampal mitochondria NAD+ pools, protein acetylation, and ROS levels. These changes were dependent on SIRT3 activity, which was confirmed using SIRT3KO mice. Ischemia induced increase in acetylation of the key mitochondrial antioxidant enzyme, superoxide dismutase 2 (SOD2) that resulted in inhibition of its activity. This was reversed after NMN treatment followed by reduction of ROS generation and suppression of mitochondrial fragmentation. Specifically, we found that the interaction of mitochondrial fission protein, pDrp1(S616), with neuronal mitochondria was inhibited in NMN treated ischemic mice. Our data thus provide a novel link between mitochondrial NAD+ metabolism, ROS production, and mitochondrial fragmentation. Using NMN to target these mechanisms could represent a new therapeutic approach for treatment of acute brain injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- Nina Klimova
- Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Adam Fearnow
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA
| | - Aaron Long
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA
| | - Tibor Kristian
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD 21201, USA; Department of Anesthesiology and the Center for Shock, Trauma, and Anesthesiology Research (S.T.A.R.), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
15
|
Tong Y, Elkin KB, Peng C, Shen J, Li F, Guan L, Ji Y, Wei W, Geng X, Ding Y. Reduced Apoptotic Injury by Phenothiazine in Ischemic Stroke through the NOX-Akt/PKC Pathway. Brain Sci 2019; 9:378. [PMID: 31847503 PMCID: PMC6955743 DOI: 10.3390/brainsci9120378] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023] Open
Abstract
Phenothiazine treatment has been shown to reduce post-stroke ischemic injury, though the underlying mechanism remains unclear. This study sought to confirm the neuroprotective effects of phenothiazines and to explore the role of the NOX (nicotinamide adenine dinucleotide phosphate oxidase)/Akt/PKC (protein kinase C) pathway in cerebral apoptosis. Sprague-Dawley rats underwent middle cerebral artery occlusion (MCAO) for 2 h and were randomly divided into 3 different cohorts: (1) saline, (2) 8 mg/kg chlorpromazine and promethazine (C+P), and (3) 8 mg/kg C+P as well as apocynin (NOX inhibitor). Brain infarct volumes were examined, and cell death/NOX activity was determined by assays. Western blotting was used to assess protein expression of kinase C-δ (PKC-δ), phosphorylated Akt (p-Akt), Bax, Bcl-XL, and uncleaved/cleaved caspase-3. Both C+P and C+P/NOX inhibitor administration yielded a significant reduction in infarct volumes and cell death, while the C+P/NOX inhibitor did not confer further reduction. In both treatment groups, anti-apoptotic Bcl-XL protein expression generally increased, while pro-apoptotic Bax and caspase-3 proteins generally decreased. PKC protein expression was decreased in both treatment groups, demonstrating a further decrease by C+P/NOX inhibitor at 6 and 24 h of reperfusion. The present study confirms C+P-mediated neuroprotection and suggests that the NOX/Akt/PKC pathway is a potential target for efficacious therapy following ischemic stroke.
Collapse
Affiliation(s)
- Yanna Tong
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
- Department of Neurology, Luhe Clinical Institute, Capital Medical University, Beijing 101100, China
| | - Kenneth B. Elkin
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
| | - Changya Peng
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
| | - Jiamei Shen
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
| | - Fengwu Li
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
| | - Longfei Guan
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
| | - Yu Ji
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
- Department of General Surgery, Luhe Clinical Institute, Capital Medical University, Beijing 101100, China
| | - Wenjing Wei
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
- China-America Institute of Neuroscience, Xuanwu Clinical Institute, Capital Medical University, Beijing 100053, China
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing 101100, China; (Y.T.); (J.S.); (F.L.); (L.G.)
- Department of Neurology, Luhe Clinical Institute, Capital Medical University, Beijing 101100, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA; (K.B.E.); (C.P.); (Y.D.)
- Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit, MI 4820, USA; (Y.J.); (W.W.)
| |
Collapse
|
16
|
Liu F, Lin C, Hong J, Cai C, Zhang W, Zhang J, Guo L. Apocynin protects retina cells from ultraviolet radiation damage via inducing sirtuin 1. J Drug Target 2019; 28:330-338. [PMID: 31479288 DOI: 10.1080/1061186x.2019.1663527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Direct exposure to Ultraviolet (UV) radiation causes progressive damages in retinal cells, which is one of the hypothetical mechanisms underlying age-related retinopathy or macular degeneration. The protective effects of Apocynin against UV damages were firstly tested in retinal pigment epithelium cells (RPEs) and retinal ganglion cells (RGCs). Subsequently the beneficial effect of Apocynin on mouse retinas against light damage were examined. Next, microarray profiling was used to identify the genes regulated by Apocynin in both RPEs and RGCs. A candidate gene was isolated for functional characterisation by knock-down study. Apocynin was shown to inhibit cell death, reduce oxidative stress and deoxyribonucleic acid damages in both RPEs and RGCs challenged with UV. Intravitreal application of Apocynin also improved retinal dysfunction caused by light damage. Sirtuin 1 (SIRT1) was identified as induced by Apocynin by microarray study. The induction was confirmed by realtime-PCR and western blotting. Knocking down SIRT1 antagonised the protective effect of Apocynin against UV damages in both RPEs and RGCs. Apocynin is a novel agent that shows both in vitro and in vivo efficacies against UV radiation induced retina damages. SIRT1 pathway is implicated in UV radiation protection of Apocynin in retinal cells.
Collapse
Affiliation(s)
- Feng Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Chen Lin
- Department of Ophthalmology, Shenzhen Aier Eye Hospital, Shenzhen, China
| | - Jinsheng Hong
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Chuanshu Cai
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Weijian Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Jianrong Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Lihong Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| |
Collapse
|
17
|
Barua S, Kim JY, Yenari MA, Lee JE. The role of NOX inhibitors in neurodegenerative diseases. IBRO Rep 2019; 7:59-69. [PMID: 31463415 PMCID: PMC6709343 DOI: 10.1016/j.ibror.2019.07.1721] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is a key player in both chronic and acute brain disease due to the higher metabolic demand of the brain. Among the producers of free radicals, NADPH-oxidase (NOX) is a major contributor to oxidative stress in neurological disorders. In the brain, the superoxide produced by NOX is mainly found in leukocytes. However, recent studies have reported that it can be found in several other cell types. NOX has been reported to regulate neuronal signaling, memory processing, and central cardiovascular homeostasis. However, overproduction of NOX can contribute to neurotoxicity, CNS degeneration, and cardiovascular disorders. Regarding the above functions, NOX has been shown to play a crucial role in chronic CNS diseases like Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), and in acute CNS disorders such as stroke, spinal cord injury, traumatic brain injury (TBI), and related cerebrovascular diseases. NOX is a multi-subunit complex consisting of two membrane-associated and four cytosolic subunits. Thus, in recent years, inhibition of NOX activity has drawn a great deal of attention from researchers in the field of treating chronic and acute CNS disorders and preventing secondary complications. Mounting evidence has shown that NOX inhibition is neuroprotective and that inhibiting NOX in circulating immune cells can improve neurological disease conditions. This review summarizes recent studies on the therapeutic effects and pharmacological strategies regarding NOX inhibitors in chronic and acute brain diseases and focuses on the hurdles that should be overcome before their clinical implementation.
Collapse
Affiliation(s)
- Sumit Barua
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea
| | - Midori A Yenari
- Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, 4150 Clement Street, MS 127, San Francisco, CA, 94121, United States
| | - Jong Eun Lee
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea.,Brain Korea 21, PLUS Project for Medical Science, College of Medicine, Yonsei University, Republic of Korea.,Brain Research Institute, College of Medicine, Yonsei University, Republic of Korea
| |
Collapse
|
18
|
Synergistic Role of Oxidative Stress and Blood-Brain Barrier Permeability as Injury Mechanisms in the Acute Pathophysiology of Blast-induced Neurotrauma. Sci Rep 2019; 9:7717. [PMID: 31118451 PMCID: PMC6531444 DOI: 10.1038/s41598-019-44147-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/07/2019] [Indexed: 12/24/2022] Open
Abstract
Blast-induced traumatic brain injury (bTBI) has been recognized as the common mode of neurotrauma amongst military and civilian personnel due to an increased insurgent activity domestically and abroad. Previous studies from our laboratory have identified enhanced blood-brain barrier (BBB) permeability as a significant, sub-acute (four hours post-blast) pathological change in bTBI. We also found that NADPH oxidase (NOX)-mediated oxidative stress occurs at the same time post-blast when the BBB permeability changes. We therefore hypothesized that oxidative stress is a major causative factor in the BBB breakdown in the sub-acute stages. This work therefore examined the role of NOX1 and its downstream effects on BBB permeability in the frontal cortex (a region previously shown to be the most vulnerable) immediately and four hours post-blast exposure. Rats were injured by primary blast waves in a compressed gas-driven shock tube at 180 kPa and the BBB integrity was assessed by extravasation of Evans blue and changes in tight junction proteins (TJPs) as well as translocation of macromolecules from blood to brain and vice versa. NOX1 abundance was also assessed in neurovascular endothelial cells. Blast injury resulted in increased extravasation and reduced levels of TJPs in tissues consistent with our previous observations. NOX1 levels were significantly increased in endothelial cells followed by increased superoxide production within 4 hours of blast. Blast injury also increased the levels/activation of matrix metalloproteinase 3 and 9. To test the role of oxidative stress, rats were administered apocynin, which is known to inhibit the assembly of NOX subunits and arrests its function. We found apocynin completely inhibited dye extravasation as well as restored TJP levels to that of controls and reduced matrix metalloproteinase activation in the sub-acute stages following blast. Together these data strongly suggest that NOX-mediated oxidative stress contributes to enhanced BBB permeability in bTBI through a pathway involving increased matrix metalloproteinase activation.
Collapse
|
19
|
Cananzi SG, Mayhan WG. In Utero Exposure to Alcohol Impairs Reactivity of Cerebral Arterioles and Increases Susceptibility of the Brain to Damage Following Ischemia/Reperfusion in Adulthood. Alcohol Clin Exp Res 2019; 43:607-616. [PMID: 30748017 DOI: 10.1111/acer.13979] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/01/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Maternal consumption of alcohol produces abnormalities in the developing fetus and can contribute to an increased incidence of many cardiovascular-related diseases. The first goal of this study was to determine whether in utero exposure to alcohol influences reactivity of cerebral arterioles in adult (12 to 15 weeks old) rats. The second goal of this study was to examine whether in utero exposure to alcohol increased the susceptibility of the brain to damage following an ischemic event in adult rats. METHODS We fed Sprague Dawley dams a liquid diet with or without alcohol (3% ethanol) for the duration of their pregnancy (21 to 23 days). In the first series of studies, we examined reactivity of cerebral arterioles to endothelial nitric oxide synthase (eNOS)- (adenosine diphosphate [ADP]) and neuronal nitric oxide synthase (nNOS)-dependent N-methyl-D-aspartate (NMDA, and NOS-independent agonists in adult rats before and during application of l-NMMA. In another series of studies, we examined infarct volume following middle cerebral artery occlusion in adult offspring exposed to alcohol in utero. In both series of studies, we also determined the role for an increase in oxidative stress by feeding dams apocynin for the duration of their pregnancy. RESULTS We found that in utero exposure to alcohol reduced responses of cerebral arterioles to ADP and NMDA, but not to nitroglycerin in adult rats. In addition, treatment of the dams with apocynin prevented this impairment in cerebral vascular function. We also found that in utero exposure to alcohol worsened brain damage following ischemia/reperfusion in adult rats and that treatment of dams with apocynin prevented this increase in brain damage following ischemia/reperfusion. CONCLUSIONS We suggest that our findings may have important implications for the pathogenesis of brain abnormalities associated with fetal alcohol exposure.
Collapse
Affiliation(s)
- Sergio G Cananzi
- Department of Molecular Biology , University of Texas-Southwestern, Dallas, Texas
| | - William G Mayhan
- Division of Basic Biomedical Sciences , Sanford School of Medicine, University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
20
|
Chandran R, Kim T, Mehta SL, Udho E, Chanana V, Cengiz P, Kim H, Kim C, Vemuganti R. A combination antioxidant therapy to inhibit NOX2 and activate Nrf2 decreases secondary brain damage and improves functional recovery after traumatic brain injury. J Cereb Blood Flow Metab 2018; 38:1818-1827. [PMID: 29083257 PMCID: PMC6168911 DOI: 10.1177/0271678x17738701] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Uncontrolled oxidative stress contributes to the secondary neuronal death that promotes long-term neurological dysfunction following traumatic brain injury (TBI). Surprisingly, both NADPH oxidase 2 (NOX2) that increases and transcription factor Nrf2 that decreases reactive oxygen species (ROS) are induced after TBI. As the post-injury functional outcome depends on the balance of these opposing molecular pathways, we evaluated the effect of TBI on the motor and cognitive deficits and cortical contusion volume in NOX2 and Nrf2 knockout mice. Genetic deletion of NOX2 improved, while Nrf2 worsened the post-TBI motor function recovery and lesion volume indicating that decreasing ROS levels might be beneficial after TBI. Treatment with either apocynin (NOX2 inhibitor) or TBHQ (Nrf2 activator) alone significantly improved the motor function after TBI, but had no effect on the lesion volume, compared to vehicle control. Whereas, the combo therapy (apocynin + TBHQ) given at either 5 min/24 h or 2 h/24 h improved motor and cognitive function and decreased cortical contusion volume compared to vehicle group. Thus, both the generation and disposal of ROS are important modulators of oxidative stress, and a combo therapy that prevents ROS formation and potentiates ROS disposal concurrently is efficacious after TBI.
Collapse
Affiliation(s)
| | - TaeHee Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Suresh L Mehta
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Eshwar Udho
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Vishal Chanana
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - Pelin Cengiz
- 2 Department of Pediatrics, University of Wisconsin, Madison, WI, USA
| | - HwuiWon Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Chanul Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,3 William S. Middleton Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
21
|
In-vivo evaluation of apocynin for prevention of Helicobacter pylori-induced gastric carcinogenesis. Eur J Cancer Prev 2018; 26:10-16. [PMID: 26938501 DOI: 10.1097/cej.0000000000000233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The emergence of antibiotic-resistant Helicobacter pylori strains impacts the efficacy of eradication therapy and promotes the development of alternative treatment strategies. Apocynin inhibits neutrophil NADPH oxidase and hence may decrease reactive oxygen species-mediated tissue damage in H. pylori-infected stomach tissue. Apocynin was tested in vitro for its cytotoxic and direct antibacterial effects. The therapeutic efficacy of orally administered apocynin (100 mg/kg/day through drinking water or 200 mg/kg/day through combined administration of drinking water and slow-release formulation) was assessed at 9 weeks after infection in the Mongolian gerbil model. Bacterial burdens were quantified by viable plate count and quantitative PCR. Histopathological evaluation of antrum and pylorus provided insight into mucosal inflammation and injury. Apocynin showed no cytotoxic or direct antibacterial effects in vitro or in vivo. Nine weeks of apocynin treatment at 200 mg/kg/day reduced active H. pylori gastritis as neutrophil infiltration in the mucous neck region and pit abscess formation decreased significantly. In our gerbil model, prolonged high-dose apocynin treatment significantly improved H. pylori-induced pit abscess formation without indications of drug toxicity and thus further investigation of the dosage regimen and formulation and the long-term impact on neoplastic development should be carried out.
Collapse
|
22
|
Li X, Guo H, Zhao L, Wang B, Liu H, Yue L, Bai H, Jiang H, Gao L, Feng D, Qu Y. Adiponectin attenuates NADPH oxidase-mediated oxidative stress and neuronal damage induced by cerebral ischemia-reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3265-3276. [DOI: 10.1016/j.bbadis.2017.08.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/12/2017] [Accepted: 08/09/2017] [Indexed: 10/19/2022]
|
23
|
Kim JY, Park J, Lee JE, Yenari MA. NOX Inhibitors - A Promising Avenue for Ischemic Stroke. Exp Neurobiol 2017; 26:195-205. [PMID: 28912642 PMCID: PMC5597550 DOI: 10.5607/en.2017.26.4.195] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/19/2022] Open
Abstract
NADPH-oxidase (NOX) mediated superoxide originally found on leukocytes, but now recognized in several types of cells in the brain. It has been shown to play an important role in the progression of stroke and related cerebrovascular disease. NOX is a multisubunit complex consisting of 2 membrane-associated and 4 cytosolic subunits. NOX activation occurs when cytosolic subunits translocate to the membrane, leading to transport electrons to oxygen, thus producing superoxide. Superoxide produced by NOX is thought to function in long-term potentiation and intercellular signaling, but excessive production is damaging and has been implicated to play an important role in the progression of ischemic brain. Thus, inhibition of NOX activity may prove to be a promising treatment for ischemic brain as well as an adjunctive agent to prevent its secondary complications. There is mounting evidence that NOX inhibition in the ischemic brain is neuroprotective, and targeting NOX in circulating immune cells will also improve outcome. This review will focus on therapeutic effects of NOX assembly inhibitors in brain ischemia and stroke. However, the lack of specificity and toxicities of existing inhibitors are clear hurdles that will need to be overcome before this class of compounds could be translated clinically.
Collapse
Affiliation(s)
- Jong Youl Kim
- Department of Anatomy, Yensei University College of Medicine, Seoul 03722, Korea
| | - Joohyun Park
- Department of Anatomy, Yensei University College of Medicine, Seoul 03722, Korea
- BK21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yensei University College of Medicine, Seoul 03722, Korea
- BK21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Midori A. Yenari
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California 94121, USA
| |
Collapse
|
24
|
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: 32] [Impact Index Per Article: 4.0] [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
|
25
|
Cai L, Stevenson J, Geng X, Peng C, Ji X, Xin R, Rastogi R, Sy C, Rafols JA, Ding Y. Combining Normobaric Oxygen with Ethanol or Hypothermia Prevents Brain Damage from Thromboembolic Stroke via PKC-Akt-NOX Modulation. Mol Neurobiol 2017; 54:1263-1277. [PMID: 26820681 DOI: 10.1007/s12035-016-9695-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/05/2016] [Indexed: 01/01/2023]
Abstract
In a thromboembolic stroke model after reperfusion by recombinant tissue plasminogen activator (rt-PA), we aimed to determine whether therapeutic hypothermia (TH) and ethanol (EtOH) in combination with low concentration (60 %) of normobaric oxygen (NBO) enhanced neuroprotection, as compared to using each of these agents alone. We further aimed to elucidate a potential role of the NADPH oxidase (NOX), phosphorylated protein kinase B (Akt), and protein kinase C-δ (PKC-δ) pathway in oxidative stress and neuroprotection. In Sprague-Dawley rats, a focal middle cerebral artery (MCA) occlusion was induced by an autologous embolus in the following experimental groups: rt-PA treatment alone, rt-PA + NBO treatment, rt-PA + TH at 33 °C, rt-PA + EtOH, rt-PA + NBO + EtOH, rt-PA + NBO + TH, rt-PA + NOX inhibitor, rt-PA + EtOH + NOX inhibitor, or rt-PA + EtOH + Akt inhibitor. Control groups included sham-operated without stroke or stroke without treatment. Infarct volume and neurological deficit were assessed at 24 h after rt-PA-induced reperfusion with or without treatments. ROS levels, NOX activity, and the protein expression of NOX subunits p22phox, p47phox, p67phox, gp91phox, as well as PKC-δ and phosphorylated Akt were measured at 3 and 24 h after rt-PA-induced reperfusion. Following rt-PA in thromboembolic stroke rats, NBO combined with TH or EtOH more effectively decreased infarct volume and neurological deficit, as well as reactive oxygen species (ROS) production than with any of the used monotherapies. NOX activity and subunit expressions were downregulated and temporally associated with reduced PKC-δ and increased p-Akt expression. The present study demonstrated that combining NBO with either TH or EtOH conferred similar neuroprotection via modulation of NOX activation. The results suggest a role of Akt in NOX activation and implicate an upstream PKC-δ pathway in the Akt regulation of NOX. It is possible to substitute EtOH for TH, thus circumventing the difficulties in clinical application of TH through the comparatively easier usage of EtOH as a potential stroke management.
Collapse
Affiliation(s)
- Lipeng Cai
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 101149, China
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - James Stevenson
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - Xiaokun Geng
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - Changya Peng
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - Xunming Ji
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 101149, China.
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China.
| | - Ruiqiang Xin
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
- Department of Radiology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Radhika Rastogi
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - Christopher Sy
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA
| | - Jose A Rafols
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuchuan Ding
- China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 101149, China.
- Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China.
- Department of Neurological Surgery, Wayne State University School of Medicine, 550 E Canfield, Detroit, MI, 48201, USA.
| |
Collapse
|
26
|
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: 312] [Impact Index Per Article: 39.0] [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
|
27
|
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: 153] [Impact Index Per Article: 19.1] [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
|
28
|
Li W, Yang S. Targeting oxidative stress for the treatment of ischemic stroke: Upstream and downstream therapeutic strategies. Brain Circ 2016; 2:153-163. [PMID: 30276293 PMCID: PMC6126224 DOI: 10.4103/2394-8108.195279] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/04/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022] Open
Abstract
Excessive oxygen and its chemical derivatives, namely reactive oxygen species (ROS), produce oxidative stress that has been known to lead to cell injury in ischemic stroke. ROS can damage macromolecules such as proteins and lipids and leads to cell autophagy, apoptosis, and necrosis to the cells. This review describes studies on the generation of ROS, its role in the pathogenesis of ischemic stroke, and recent development in therapeutic strategies in reducing oxidative stress after ischemic stroke.
Collapse
Affiliation(s)
- Wenjun Li
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shaohua Yang
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
29
|
de Oliveira JK, Ronik DFV, Ascari J, Mainardes RM, Khalil NM. A stability-indicating high performance liquid chromatography method to determine apocynin in nanoparticles. J Pharm Anal 2016; 7:129-133. [PMID: 29404028 PMCID: PMC5686858 DOI: 10.1016/j.jpha.2016.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/01/2016] [Accepted: 08/02/2016] [Indexed: 12/27/2022] Open
Abstract
In this study, we developed and validated a fast, specific, sensitive, precise and stability-indicating high performance liquid chromatography (HPLC) method to determine the drug apocynin in bovine serum albumin (BSA) nanoparticles. Chromatographic analyses were performed on an RP C18 column and using a photodiode array detector at a wavelength of 276 nm. Mobile phase consisted of a mixture of acetonitrile and 1% acetic acid (60:40, v/v), and it was eluted isocratically at a flow rate of 0.8 mL/min. The retention time of apocynin chromatographic peak was 1.65 min. The method was linear, precise, accurate and specific in the range of 5–100 μg/mL. The intra- and inter-day precisions presented relative standard deviation (RSD) values lower than 2%. The method was robust regarding changes in mobile phase proportion, but not for flow rate. Limits of detection and quantitation were 78 ng/mL and 238 ng/mL, respectively. Apocynin was exposed to acid and alkali hydrolysis, oxidation and visible light. The drug suffered mild degradation under acid and oxidation conditions and great degradation under alkali conditions. Light exposure did not degrade the drug. The method was successfully applied to determine the encapsulation efficiency of apocynin in BSA nanoparticles.
Collapse
Affiliation(s)
- Juliana Kovalczuk de Oliveira
- Department of Pharmacy, Laboratory of Pharmaceutical Nanotechnology, Universidade Estadual do Centro-Oeste, Rua Simeão Camargo Varela de Sá, 85040-080 Guarapuava, Brazil
| | - Débora Fernanda Veres Ronik
- Department of Pharmacy, Laboratory of Pharmaceutical Nanotechnology, Universidade Estadual do Centro-Oeste, Rua Simeão Camargo Varela de Sá, 85040-080 Guarapuava, Brazil
| | - Jociani Ascari
- Department of Biological Sciences, Universidade Tecnológica Federal do Paraná, Rua Cerejeiras S/N, 85892-000 Santa Helena, PR, Brazil
| | - Rubiana Mara Mainardes
- Department of Pharmacy, Laboratory of Pharmaceutical Nanotechnology, Universidade Estadual do Centro-Oeste, Rua Simeão Camargo Varela de Sá, 85040-080 Guarapuava, Brazil
| | - Najeh Maissar Khalil
- Department of Pharmacy, Laboratory of Pharmaceutical Nanotechnology, Universidade Estadual do Centro-Oeste, Rua Simeão Camargo Varela de Sá, 85040-080 Guarapuava, Brazil
| |
Collapse
|
30
|
De Silva TM, Miller AA. Cerebral Small Vessel Disease: Targeting Oxidative Stress as a Novel Therapeutic Strategy? Front Pharmacol 2016; 7:61. [PMID: 27014073 PMCID: PMC4794483 DOI: 10.3389/fphar.2016.00061] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/04/2016] [Indexed: 12/25/2022] Open
Abstract
Cerebral small vessel disease (SVD) is a major contributor to stroke, and a leading cause of cognitive impairment and dementia. Despite the devastating effects of cerebral SVD, the pathogenesis of cerebral SVD is still not completely understood. Moreover, there are no specific pharmacological strategies for its prevention or treatment. Cerebral SVD is characterized by marked functional and structural abnormalities of the cerebral microcirculation. The clinical manifestations of these pathological changes include lacunar infarcts, white matter hyperintensities, and cerebral microbleeds. The main purpose of this review is to discuss evidence implicating oxidative stress in the arteriopathy of both non-amyloid and amyloid (cerebral amyloid angiopathy) forms of cerebral SVD and its most important risk factors (hypertension and aging), as well as its contribution to cerebral SVD-related brain injury and cognitive impairment. We also highlight current evidence of the involvement of the NADPH oxidases in the development of oxidative stress, enzymes that are a major source of reactive oxygen species in the cerebral vasculature. Lastly, we discuss potential pharmacological strategies for oxidative stress in cerebral SVD, including some of the historical and emerging NADPH oxidase inhibitors.
Collapse
Affiliation(s)
- T. Michael De Silva
- Department of Pharmacology, Biomedicine Discovery Institute, Monash UniversityMelbourne, VIC, Australia
| | - Alyson A. Miller
- Cerebrovascular and Stroke Laboratory, School of Health and Biomedical Sciences, RMIT UniversityMelbourne, VIC, Australia
| |
Collapse
|
31
|
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: 13] [Impact Index Per Article: 1.4] [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
|
32
|
Chen H, Guan B, Shen J. Targeting ONOO -/HMGB1/MMP-9 Signaling Cascades: Potential for Drug Development from Chinese Medicine to Attenuate Ischemic Brain Injury and Hemorrhagic Transformation Induced by Thrombolytic Treatment. ACTA ACUST UNITED AC 2016. [DOI: 10.1159/000442468] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
33
|
NADPH Oxidase: A Potential Target for Treatment of Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5026984. [PMID: 26941888 PMCID: PMC4752995 DOI: 10.1155/2016/5026984] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/31/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022]
Abstract
Stroke is the third leading cause of death in industrialized nations. Oxidative stress is involved in the pathogenesis of stroke, and excessive generation of reactive oxygen species (ROS) by mitochondria is thought to be the main cause of oxidative stress. NADPH oxidase (NOX) enzymes have recently been identified and studied as important producers of ROS in brain tissues after stroke. Several reports have shown that knockout or deletion of NOX exerts a neuroprotective effect in three major experimental stroke models. Recent studies also confirmed that NOX inhibitors ameliorate brain injury and improve neurological outcome after stroke. However, the physiological and pathophysiological roles of NOX enzymes in the central nervous system (CNS) are not known well. In this review, we provide a comprehensive summary of our current understanding about expression and physiological function of NOX enzymes in the CNS and its pathophysiological roles in the three major types of stroke: ischemic stroke, intracerebral hemorrhage, and subarachnoid hemorrhage.
Collapse
|
34
|
Oxidative Stress in Intracerebral Hemorrhage: Sources, Mechanisms, and Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3215391. [PMID: 26843907 PMCID: PMC4710930 DOI: 10.1155/2016/3215391] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is associated with the highest mortality and morbidity despite only constituting approximately 10–15% of all strokes. Complex underlying mechanisms consisting of cytotoxic, excitotoxic, and inflammatory effects of intraparenchymal blood are responsible for its highly damaging effects. Oxidative stress (OS) also plays an important role in brain injury after ICH but attracts less attention than other factors. Increasing evidence has demonstrated that the metabolite axis of hemoglobin-heme-iron is the key contributor to oxidative brain damage after ICH, although other factors, such as neuroinflammation and prooxidases, are involved. This review will discuss the sources, possible molecular mechanisms, and potential therapeutic targets of OS in ICH.
Collapse
|
35
|
Apocyanin, NADPH oxidase inhibitor prevents lipopolysaccharide induced α-synuclein aggregation and ameliorates motor function deficits in rats: Possible role of biochemical and inflammatory alterations. Behav Brain Res 2015; 296:177-190. [PMID: 26367469 DOI: 10.1016/j.bbr.2015.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/05/2015] [Accepted: 09/09/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), is an age-related, progressive neurodegenerative disorder that affects movement and is characterized by the loss of dopaminergic neurons in the nigrostriatal region. Although the clinical and pathological features of PD are complex, recent studies have indicated that microglial NADPH oxidase play a key role in its pathology. A little information is available regarding the role of apocyanin, an NADPH oxidase inhibitor, in ameliorating α-synuclein aggregation and neurobehavioral consequences of PD. Therefore, the present study evaluated its therapeutic potentials for the treatment of neurobehavioral consequences in lipolysaccharide (LPS) induced PD model. For the establishment of PD model LPS (5 μg/5 μl PBS) was injected into the Substantia nigra (SN) of rats. Apocyanin (10mg/kgb.wt) was injected intraperitoneal. Statistical analysis revealed that apocynin significantly ameliorated LPS induced inflammatory response characterized by NFkB, TNF-α and IL-1β upregulation as assessed by ELISA. It also prevented dopaminergic neurons from toxic insult of LPS as indicated by inhibition of apoptotic markers i.e., caspase 3 and caspase 9 as depicted from RT-PCR and ELISA studies. This was further supported by TUNEL assay for DNA fragmentation. Effectiveness of apocyanin in protecting dopaminergic neuronal degeneration was further confirmed by assessment of α-synuclein deposition as depicted by IHC analysis. Consequently, an improvement in the behavioral outcome was observed following apocyanin treatment as depicted from various behavioral tests performed. Hence the data suggests that specific NADPH oxidase inhibitors, such as apocynin, may provide a new therapeutic approach to the control of neurological disabilities induced by LPS induced PD.
Collapse
|
36
|
Diebold BA, Smith SM, Li Y, Lambeth JD. NOX2 As a Target for Drug Development: Indications, Possible Complications, and Progress. Antioxid Redox Signal 2015; 23:375-405. [PMID: 24512192 PMCID: PMC4545678 DOI: 10.1089/ars.2014.5862] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/08/2014] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE NOX2 is important for host defense, and yet is implicated in a large number of diseases in which inflammation plays a role in pathogenesis. These include acute and chronic lung inflammatory diseases, stroke, traumatic brain injury, and neurodegenerative diseases, including Alzheimer's and Parkinson's Diseases. RECENT ADVANCES Recent drug development programs have targeted several NOX isoforms that are implicated in a variety of diseases. The focus has been primarily on NOX4 and NOX1 rather than on NOX2, due, in part, to concerns about possible immunosuppressive side effects. Nevertheless, NOX2 clearly contributes to the pathogenesis of many inflammatory diseases, and its inhibition is predicted to provide a novel therapeutic approach. CRITICAL ISSUES Possible side effects that might arise from targeting NOX2 are discussed, including the possibility that such inhibition will contribute to increased infections and/or autoimmune disorders. The state of the field with regard to existing NOX2 inhibitors and targeted development of novel inhibitors is also summarized. FUTURE DIRECTIONS NOX2 inhibitors show particular promise for the treatment of inflammatory diseases, both acute and chronic. Theoretical side effects include pro-inflammatory and autoimmune complications and should be considered in any therapeutic program, but in our opinion, available data do not indicate that they are sufficiently likely to eliminate NOX2 as a drug target, particularly when weighed against the seriousness of many NOX2-related indications. Model studies demonstrating efficacy with minimal side effects are needed to encourage future development of NOX2 inhibitors as therapeutic agents.
Collapse
Affiliation(s)
- Becky A. Diebold
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Susan M.E. Smith
- Department of Biology and Physics, Kennesaw State University, Kennesaw, Georgia
| | - Yang Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - J. David Lambeth
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
37
|
Carbone F, Teixeira PC, Braunersreuther V, Mach F, Vuilleumier N, Montecucco F. Pathophysiology and Treatments of Oxidative Injury in Ischemic Stroke: Focus on the Phagocytic NADPH Oxidase 2. Antioxid Redox Signal 2015; 23:460-489. [PMID: 24635113 PMCID: PMC4545676 DOI: 10.1089/ars.2013.5778] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 03/05/2014] [Accepted: 03/16/2014] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Phagocytes play a key role in promoting the oxidative stress after ischemic stroke occurrence. The phagocytic NADPH oxidase (NOX) 2 is a membrane-bound enzyme complex involved in the antimicrobial respiratory burst and free radical production in these cells. RECENT ADVANCES Different oxidants have been shown to induce opposite effects on neuronal homeostasis after a stroke. However, several experimental models support the detrimental effects of NOX activity (especially the phagocytic isoform) on brain recovery after stroke. Therapeutic strategies selectively targeting the neurotoxic ROS and increasing neuroprotective oxidants have recently produced promising results. CRITICAL ISSUES NOX2 might promote carotid plaque rupture and stroke occurrence. In addition, NOX2-derived reactive oxygen species (ROS) released by resident and recruited phagocytes enhance cerebral ischemic injury, activating the inflammatory apoptotic pathways. The aim of this review is to update evidence on phagocyte-related oxidative stress, focusing on the role of NOX2 as a potential therapeutic target to reduce ROS-related cerebral injury after stroke. FUTURE DIRECTIONS Radical scavenger compounds (such as Ebselen and Edaravone) are under clinical investigation as a therapeutic approach against stroke. On the other hand, NOX inhibition might represent a promising strategy to prevent the stroke-related injury. Although selective NOX inhibitors are not yet available, nonselective compounds (such as apocynin and fasudil) provided encouraging results in preclinical studies. Whereas additional studies are needed to better evaluate this therapeutic potential in human beings, the development of specific NOX inhibitors (such as monoclonal antibodies, small-molecule inhibitors, or aptamers) might further improve brain recovery after stroke.
Collapse
Affiliation(s)
- Federico Carbone
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Priscila Camillo Teixeira
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Braunersreuther
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Fabrizio Montecucco
- Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
- Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino–IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
- Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
38
|
Hirano K, Chen WS, Chueng ALW, Dunne AA, Seredenina T, Filippova A, Ramachandran S, Bridges A, Chaudry L, Pettman G, Allan C, Duncan S, Lee KC, Lim J, Ma MT, Ong AB, Ye NY, Nasir S, Mulyanidewi S, Aw CC, Oon PP, Liao S, Li D, Johns DG, Miller ND, Davies CH, Browne ER, Matsuoka Y, Chen DW, Jaquet V, Rutter AR. Discovery of GSK2795039, a Novel Small Molecule NADPH Oxidase 2 Inhibitor. Antioxid Redox Signal 2015; 23:358-74. [PMID: 26135714 PMCID: PMC4545375 DOI: 10.1089/ars.2014.6202] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIMS The NADPH oxidase (NOX) family of enzymes catalyzes the formation of reactive oxygen species (ROS). NOX enzymes not only have a key role in a variety of physiological processes but also contribute to oxidative stress in certain disease states. To date, while numerous small molecule inhibitors have been reported (in particular for NOX2), none have demonstrated inhibitory activity in vivo. As such, there is a need for the identification of improved NOX inhibitors to enable further evaluation of the biological functions of NOX enzymes in vivo as well as the therapeutic potential of NOX inhibition. In this study, both the in vitro and in vivo pharmacological profiles of GSK2795039, a novel NOX2 inhibitor, were characterized in comparison with other published NOX inhibitors. RESULTS GSK2795039 inhibited both the formation of ROS and the utilization of the enzyme substrates, NADPH and oxygen, in a variety of semirecombinant cell-free and cell-based NOX2 assays. It inhibited NOX2 in an NADPH competitive manner and was selective over other NOX isoforms, xanthine oxidase, and endothelial nitric oxide synthase enzymes. Following systemic administration in mice, GSK2795039 abolished the production of ROS by activated NOX2 enzyme in a paw inflammation model. Furthermore, GSK2795039 showed activity in a murine model of acute pancreatitis, reducing the levels of serum amylase triggered by systemic injection of cerulein. INNOVATION AND CONCLUSIONS GSK2795039 is a novel NOX2 inhibitor that is the first small molecule to demonstrate inhibition of the NOX2 enzyme in vivo.
Collapse
Affiliation(s)
- Kazufumi Hirano
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Woei Shin Chen
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Adeline L W Chueng
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Angela A Dunne
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Tamara Seredenina
- 2 Department of Pathology and Immunology, Medical School, Centre Médical Universitaire, University of Geneva , Geneva, Switzerland
| | - Aleksandra Filippova
- 2 Department of Pathology and Immunology, Medical School, Centre Médical Universitaire, University of Geneva , Geneva, Switzerland
| | - Sumitra Ramachandran
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Angela Bridges
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Laiq Chaudry
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Gary Pettman
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Craig Allan
- 3 Platform Technology & Sciences Department, GlaxoSmithKline , Stevenage, United Kingdom
| | - Sarah Duncan
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Kiew Ching Lee
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Jean Lim
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - May Thu Ma
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Agnes B Ong
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Nicole Y Ye
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Shabina Nasir
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Sri Mulyanidewi
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Chiu Cheong Aw
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Pamela P Oon
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Shihua Liao
- 4 Neuroimmunology Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Shanghai, China
| | - Dizheng Li
- 4 Neuroimmunology Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Shanghai, China
| | - Douglas G Johns
- 5 Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline , King of Prussia, Pennsylvania
| | - Neil D Miller
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Ceri H Davies
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Edward R Browne
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Yasuji Matsuoka
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Deborah W Chen
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| | - Vincent Jaquet
- 2 Department of Pathology and Immunology, Medical School, Centre Médical Universitaire, University of Geneva , Geneva, Switzerland
| | - A Richard Rutter
- 1 Neural Pathways Discovery Performance Unit, Neurosciences Therapeutic Area, GlaxoSmithKline , Biopolis, Singapore
| |
Collapse
|
39
|
Sharma N, Nehru B. Apocyanin, a Microglial NADPH Oxidase Inhibitor Prevents Dopaminergic Neuronal Degeneration in Lipopolysaccharide-Induced Parkinson's Disease Model. Mol Neurobiol 2015; 53:3326-3337. [PMID: 26081143 DOI: 10.1007/s12035-015-9267-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/28/2015] [Indexed: 02/05/2023]
Abstract
Microglia-associated inflammatory processes have been strongly implicated in the development and progression of Parkinson's disease (PD). Specifically, microglia are activated in response to lipopolysaccharide (LPS) and become chronic source of cytokines and reactive oxygen species (ROS) production. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex is responsible for extracellular as well as intracellular production of ROS by microglia and its expression is upregulated in PD. Therefore, targeting NADPH oxidase complex activation using an NADPH oxidase inhibitor, i.e., apocyanin seems to be an effective approach. The aim of present study was to investigate the neuroprotective effects of apocyanin in a LPS-induced PD model. LPS (5 μg) was injected intranigral and apocyanin was administered daily at a dose of 10 mg/kg b.wt (i.p.) during the experiment. LPS when injected into the substantia nigra (SN) reproduced the characteristic hallmark features of PD in rats. It elicited an inflammatory response characterized by glial cell activation (Iba-1, GFAP). Furthermore, LPS upregulated the gene expression of nuclear factor-κB (NFκB), iNOS, and gp91PHOX and resulted in an elevated total ROS production as well as NADPH oxidase activity. Subsequently, this resulted in dopaminergic loss as depicted by decreased tyrosine hydroxylase (TH) expression with substantial loss in neurotransmitter dopamine and its metabolites, whereas treatment with apocyanin significantly reduced the number of glial fibrillary acidic protein (GFAP) and Iba-1-positive cells in LPS-treated animals. It also mitigated microglial activation-induced inflammatory response and elevation in NADPH oxidase activity, thus reducing the extracellular as well as intracellular ROS production. The present study indicated that targeting NADPH oxidase can inhibit microglial activation and reduce a broad spectrum of toxic factors generation (i.e., cytokines, ROS, and reactive nitrogen species [RNS]), thus offering a hope in halting the progression of PD.
Collapse
Affiliation(s)
- Neha Sharma
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Bimla Nehru
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
40
|
Choi BY, Kim JH, Kho AR, Kim IY, Lee SH, Lee BE, Choi E, Sohn M, Stevenson M, Chung TN, Kauppinen TM, Suh SW. Inhibition of NADPH oxidase activation reduces EAE-induced white matter damage in mice. J Neuroinflammation 2015; 12:104. [PMID: 26017142 PMCID: PMC4449958 DOI: 10.1186/s12974-015-0325-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 05/15/2015] [Indexed: 11/10/2022] Open
Abstract
Background To evaluate the role of NADPH oxidase-mediated reactive oxygen species (ROS) production in multiple sclerosis pathogenesis, we examined the effects of apocynin, an NADPH oxidase assembly inhibitor, on experimental autoimmune encephalomyelitis (EAE). Methods EAE was induced by immunization with myelin oligodendrocyte glycoprotein (MOG (35-55)) in C57BL/6 female mice. Three weeks after initial immunization, the mice were analyzed for demyelination, immune cell infiltration, and ROS production. Apocynin (30 mg/kg) was given orally once daily for the entire experimental course or after the typical onset of clinical symptom (15 days after first MOG injection). Results Clinical signs of EAE first appeared on day 11 and reached a peak level on day 19 after the initial immunization. The daily clinical symptoms of EAE mice were profoundly reduced by apocynin. The apocynin-mediated inhibition of the clinical course of EAE was accompanied by suppression of demyelination, reduced infiltration by encephalitogenic immune cells including CD4, CD8, CD20, and F4/80-positive cells. Apocynin reduced MOG-induced pro-inflammatory cytokines in cultured microglia. Apocynin also remarkably inhibited EAE-associated ROS production and blood–brain barrier (BBB) disruption. Furthermore, the present study found that post-treatment with apocynin also reduced the clinical course of EAE and spinal cord demyelination. Conclusions These results demonstrate that apocynin inhibits the clinical features and neuropathological changes associated with EAE. Therefore, the present study suggests that inhibition of NADPH oxidase activation by apocynin may have a high therapeutic potential for treatment of multiple sclerosis pathogenesis.
Collapse
Affiliation(s)
- Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - Jin Hee Kim
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - A Ra Kho
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - In Yeol Kim
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - Song Hee Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - Bo Eun Lee
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - Eunhi Choi
- Chuncheon Sacred Heart Hospital, Department of Rehabilitation Medicine, College of Medicine, Hallym University, Chuncheon, South Korea.
| | - Min Sohn
- Department of Nursing, Inha University, Incheon, South Korea.
| | - Mackenzie Stevenson
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada.
| | - Tae Nyoung Chung
- CHA Bundang Medical Center, School of Medicine, CHA University, Kyunggi do, South Korea.
| | - Tiina M Kauppinen
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Canada.
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, South Korea.
| |
Collapse
|
41
|
Saleh TM, Connell BJ, Kucukkaya I, Abd-El-Aziz AS. Increasing the Biological Stability Profile of a New Chemical Entity, UPEI-104, and Potential Use as a Neuroprotectant Against Reperfusion-Injury. Brain Sci 2015; 5:130-43. [PMID: 25906324 PMCID: PMC4493460 DOI: 10.3390/brainsci5020130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/01/2015] [Accepted: 04/13/2015] [Indexed: 11/16/2022] Open
Abstract
Previous work in our laboratory demonstrated the utility of synthetic combinations of two naturally occurring, biologically active compounds. In particular, we combined two known anti-oxidant compounds, lipoic acid and apocynin, covalently linked via an ester bond (named UPEI-100). In an animal model of ischemia-reperfusion injury (tMCAO), UPEI-100 was shown to produce equivalent neuroprotection compared to each parent compound, but at a 100-fold lower dose. However, it was determined that UPEI-100 was undetectable in any tissue samples almost immediately following intravenous injection. Therefore, the present investigation was done to determine if biological stability of UPEI-100 could be improved by replacing the ester bond with a more bio cleavage-resistant bond, an ether bond (named UPEI-104). We then compared the stability of UPEI-104 to the original parent compound UPEI-100 in human plasma as well as liver microsomes. Our results demonstrated that both UPEI-100 and UPEI-104 could be detected in human plasma for over 120 min; however, only UPEI-104 was detectable for an average of 7 min following incubation with human liver microsomes. This increased stability did not affect the biological activity of UPEI-104 as measured using our tMCAO model. Our results suggest that combining compounds using an ether bond can improve stability while maintaining biological activity.
Collapse
Affiliation(s)
- Tarek M Saleh
- Department of Biomedical Sciences, Atlantic Veterinary College, Charlottetown, PE C1A 4P3, Canada.
| | - Barry J Connell
- Department of Biomedical Sciences, Atlantic Veterinary College, Charlottetown, PE C1A 4P3, Canada.
| | - Inan Kucukkaya
- Department of Chemistry, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada.
| | - Alaa S Abd-El-Aziz
- Department of Chemistry, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
42
|
Abstract
Microglia are considered the brain's resident immune cell involved in immune defense, immunocompetence, and phagocytosis. They maintain tissue homeostasis within the brain and spinal cord under normal condition and serves as its initial host defense system. However, when the central nervous system (CNS) faces injury, microglia respond through signaling molecules expressed or released by neighboring cells. Microglial responses are dual in nature. They induce a nonspecific immune response that may exacerbate CNS injury, especially in the acute stages, but are also essential to CNS recovery and repair. The full range of microglial mechanisms have yet to be clarified, but there is accumulating knowledge about microglial activation in acute CNS injury. Microglial responses require hours to days to fully develop, and may present a therapeutic target for intervention with a much longer window of opportunity compare to other neurological treatments. The challenge will be to find ways to selectively suppress the deleterious effects of microglial activation without compromising its beneficial functions. This review aims to provide an overview of the recent progress relating on the deleterious and beneficial effect of microglia in the setting of acute CNS injury and the potential therapeutic intervention against microglial activation to CNS injury.
Collapse
Affiliation(s)
- Masahito Kawabori
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA, 94121, USA
| | | |
Collapse
|
43
|
Chandasana H, Chhonker YS, Bala V, Prasad YD, Chaitanya TK, Sharma VL, Bhatta RS. Pharmacokinetic, bioavailability, metabolism and plasma protein binding evaluation of NADPH-oxidase inhibitor apocynin using LC–MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2015; 985:180-8. [DOI: 10.1016/j.jchromb.2015.01.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/13/2015] [Accepted: 01/17/2015] [Indexed: 10/24/2022]
|
44
|
Romanini CV, Ferreira EDF, Soares LM, Santiago AN, Milani H, de Oliveira RMW. 4-hydroxy-3-methoxy-acetophenone-mediated long-lasting memory recovery, hippocampal neuroprotection, and reduction of glial cell activation after transient global cerebral ischemia in rats. J Neurosci Res 2015; 93:1240-9. [DOI: 10.1002/jnr.23575] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 12/31/2014] [Accepted: 01/22/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Cássia Valério Romanini
- Department of Pharmacology and Therapeutics; State University of Maringá; Maringá Paraná Brazil
| | | | - Lígia Mendes Soares
- Department of Pharmacology and Therapeutics; State University of Maringá; Maringá Paraná Brazil
| | - Amanda Nunes Santiago
- Department of Pharmacology and Therapeutics; State University of Maringá; Maringá Paraná Brazil
| | - Humberto Milani
- Department of Pharmacology and Therapeutics; State University of Maringá; Maringá Paraná Brazil
| | | |
Collapse
|
45
|
Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and microhemorrhage in aged Tg2576 mice. Proc Natl Acad Sci U S A 2015; 112:E881-90. [PMID: 25675483 DOI: 10.1073/pnas.1414930112] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by deposition of amyloid β peptide (Aβ) within walls of cerebral arteries and is an important cause of intracerebral hemorrhage, ischemic stroke, and cognitive dysfunction in elderly patients with and without Alzheimer's Disease (AD). NADPH oxidase-derived oxidative stress plays a key role in soluble Aβ-induced vessel dysfunction, but the mechanisms by which insoluble Aβ in the form of CAA causes cerebrovascular (CV) dysfunction are not clear. Here, we demonstrate evidence that reactive oxygen species (ROS) and, in particular, NADPH oxidase-derived ROS are a key mediator of CAA-induced CV deficits. First, the NADPH oxidase inhibitor, apocynin, and the nonspecific ROS scavenger, tempol, are shown to reduce oxidative stress and improve CV reactivity in aged Tg2576 mice. Second, the observed improvement in CV function is attributed both to a reduction in CAA formation and a decrease in CAA-induced vasomotor impairment. Third, anti-ROS therapy attenuates CAA-related microhemorrhage. A potential mechanism by which ROS contribute to CAA pathogenesis is also identified because apocynin substantially reduces expression levels of ApoE-a factor known to promote CAA formation. In total, these data indicate that ROS are a key contributor to CAA formation, CAA-induced vessel dysfunction, and CAA-related microhemorrhage. Thus, ROS and, in particular, NADPH oxidase-derived ROS are a promising therapeutic target for patients with CAA and AD.
Collapse
|
46
|
Brennan-Minnella AM, Won SJ, Swanson RA. NADPH oxidase-2: linking glucose, acidosis, and excitotoxicity in stroke. Antioxid Redox Signal 2015; 22:161-74. [PMID: 24628477 PMCID: PMC4281853 DOI: 10.1089/ars.2013.5767] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE Neuronal superoxide production contributes to cell death in both glutamate excitotoxicity and brain ischemia (stroke). NADPH oxidase-2 (NOX2) is the major source of neuronal superoxide production in these settings, and regulation of NOX2 activity can thereby influence outcome in stroke. RECENT ADVANCES Reduced NOX2 activity can rescue cells from oxidative stress and cell death that otherwise occur in excitotoxicity and ischemia. NOX2 activity is regulated by several factors previously shown to affect outcome in stroke, including glucose availability, intracellular pH, protein kinase ζ/δ, casein kinase 2, phosphoinositide-3-kinase, Rac1/2, and phospholipase A2. The newly identified functions of these factors as regulators of NOX2 activity suggest alternative mechanisms for their effects on ischemic brain injury. CRITICAL ISSUES Key aspects of these regulatory influences remain unresolved, including the mechanisms by which rac1 and phospholipase activities are coupled to N-methyl-D-aspartate (NMDA) receptors, and whether superoxide production by NOX2 triggers subsequent superoxide production by mitochondria. FUTURE DIRECTIONS It will be important to establish whether interventions targeting the signaling pathways linking NMDA receptors to NOX2 in brain ischemia can provide a greater neuroprotective efficacy or a longer time window to treatment than provided by NMDA receptor blockade alone. It will likewise be important to determine whether dissociating superoxide production from the other signaling events initiated by NMDA receptors can mitigate the deleterious effects of NMDA receptor blockade.
Collapse
|
47
|
Kim JY, Kim N, Yenari MA. Mechanisms and potential therapeutic applications of microglial activation after brain injury. CNS Neurosci Ther 2014; 21:309-19. [PMID: 25475659 DOI: 10.1111/cns.12360] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 12/14/2022] Open
Abstract
As the resident immune cells of the central nervous system, microglia rapidly respond to brain insults, including stroke and traumatic brain injury. Microglial activation plays a major role in neuronal cell damage and death by releasing a variety of inflammatory and neurotoxic mediators. Their activation is an early response that may exacerbate brain injury and many other stressors, especially in the acute stages, but are also essential to brain recovery and repair. The full range of microglial activities is still not completely understood, but there is accumulating knowledge about their role following brain injury. We review recent progress related to the deleterious and beneficial effects of microglia in the setting of acute neurological insults and the current literature surrounding pharmacological interventions for intervention.
Collapse
Affiliation(s)
- Jong-Youl Kim
- Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | | | | |
Collapse
|
48
|
Schoknecht K, David Y, Heinemann U. The blood-brain barrier-gatekeeper to neuronal homeostasis: clinical implications in the setting of stroke. Semin Cell Dev Biol 2014; 38:35-42. [PMID: 25444848 DOI: 10.1016/j.semcdb.2014.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/24/2014] [Accepted: 10/31/2014] [Indexed: 12/23/2022]
Abstract
The blood-brain barrier is part of the neurovascular unit and serves as a functional and anatomical barrier between the blood and the extracellular space. It controls the flow of solutes in and out of the brain thereby providing an optimal environment for neuronal functioning. Paracellular transport between endothelial cells is restricted by tight junctions and transendothelial transport is reduced and more selective compared to capillaries of other organs. Further, the blood-brain barrier is involved in controlling blood flow and it is the site for signaling damage of the nervous system to the peripheral immune system. As an important player in brain homeostasis, blood-brain barrier dysfunction has been implicated in the pathophysiology of many brain diseases including stroke, traumatic brain injury, brain tumors, epilepsy and neurodegenerative disorders. In this article - highlighting recent advances in basic science - we review the features of the blood-brain barrier and their significance for neuronal homeostasis to discuss clinical implications for neurological complications following cerebral ischemia.
Collapse
Affiliation(s)
- Karl Schoknecht
- Institute for Neurophysiology, Charité - University Medicine Berlin, Germany
| | - Yaron David
- Departments of Physiology & Cell Biology, Cognitive & Brain Sciences, The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Uwe Heinemann
- Institute for Neurophysiology, Charité - University Medicine Berlin, Germany.
| |
Collapse
|
49
|
McCann SK, Dusting GJ, Roulston CL. Nox2 knockout delays infarct progression and increases vascular recovery through angiogenesis in mice following ischaemic stroke with reperfusion. PLoS One 2014; 9:e110602. [PMID: 25375101 PMCID: PMC4222846 DOI: 10.1371/journal.pone.0110602] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 09/23/2014] [Indexed: 12/14/2022] Open
Abstract
Evidence suggests the NADPH oxidases contribute to ischaemic stroke injury and Nox2 is the most widely studied subtype in the context of stroke. There is still conjecture however regarding the benefits of inhibiting Nox2 to improve stroke outcome. The current study aimed to examine the temporal effects of genetic Nox2 deletion on neuronal loss after ischaemic stroke using knockout (KO) mice with 6, 24 and 72 hour recovery. Transient cerebral ischaemia was induced via intraluminal filament occlusion and resulted in reduced infarct volumes in Nox2 KO mice at 24 h post-stroke compared to wild-type controls. No protection was evident at either 6 h or 72 h post-stroke, with both genotypes exhibiting similar volumes of damage. Reactive oxygen species were detected using dihydroethidium and were co-localised with neurons and microglia in both genotypes using immunofluorescent double-labelling. The effect of Nox2 deletion on vascular damage and recovery was also examined 24 h and 72 h post-stroke using an antibody against laminin. Blood vessel density was decreased in the ischaemic core of both genotypes 24 h post-stroke and returned to pre-stroke levels only in Nox2 KO mice by 72 h. Overall, these results are the first to show that genetic Nox2 deletion merely delays the progression of neuronal loss after stroke but does not prevent it. Additionally, we show for the first time that Nox2 deletion increases re-vascularisation of the damaged brain by 72 h, which may be important in promoting endogenous brain repair mechanisms that rely on re-vascularisation.
Collapse
Affiliation(s)
- Sarah K. McCann
- Stroke Injury and Repair Team, O'Brien Institute, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Surgery, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Gregory J. Dusting
- Cytoprotection Pharmacology Program, Centre for Eye Research, the Royal Eye and Ear Hospital, Melbourne, Victoria, Australia
- Department of Ophthalmology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Carli L. Roulston
- Stroke Injury and Repair Team, O'Brien Institute, St Vincent's Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Faculty of Medicine, Dentistry and Health Sciences, St Vincent's Campus, University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
50
|
Choi DH, Lee KH, Kim JH, Seo JH, Kim HY, Shin CY, Han JS, Han SH, Kim YS, Lee J. NADPH oxidase 1, a novel molecular source of ROS in hippocampal neuronal death in vascular dementia. Antioxid Redox Signal 2014; 21:533-50. [PMID: 24294978 PMCID: PMC4086030 DOI: 10.1089/ars.2012.5129] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
AIMS Chronic cerebral hypoperfusion (CCH) is a common pathological factor that contributes to neurodegenerative diseases such as vascular dementia (VaD). Although oxidative stress has been strongly implicated in the pathogenesis of VaD, the molecular mechanism underlying the selective vulnerability of hippocampal neurons to oxidative damage remains unknown. We assessed whether the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) complex, a specialized superoxide generation system, plays a role in VaD by permanent ligation of bilateral common carotid arteries in rats. RESULTS Male Wistar rats (10 weeks of age) were subjected to bilateral occlusion of the common carotid arteries (two-vessel occlusion [2VO]). Nox1 expression gradually increased in hippocampal neurons, starting at 1 week after 2VO and for approximately 15 weeks after 2VO. The levels of superoxide, DNA oxidation, and neuronal death in the CA1 subfield of the hippocampus, as well as consequential cognitive impairment, were increased in 2VO rats. Both inhibition of Nox by apocynin, a putative Nox inhibitor, and adeno-associated virus-mediated Nox1 knockdown significantly reduced 2VO-induced reactive oxygen species generation, oxidative DNA damage, hippocampal neuronal degeneration, and cognitive impairment. INNOVATION AND CONCLUSION We provided evidence that neuronal Nox1 is activated in the hippocampus under CCH, causing oxidative stress and consequential hippocampal neuronal death and cognitive impairment. This evidence implies that Nox1-mediated oxidative stress plays an important role in neuronal cell death and cognitive dysfunction in VaD. Nox1 may serve as a potential therapeutic target for VaD.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
- Department of Medical Science, School of Medicine, Konkuk University, Seoul, Korea
| | - Kyoung-Hee Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Ji-Hye Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Ju-Ha Seo
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Hahn Young Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Chan Young Shin
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Jung-Soo Han
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Seol-Heui Han
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Yoon-Seong Kim
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Jongmin Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
- Rehabilitation Medicine, School of Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|