1
|
The Role of Copper Homeostasis in Brain Disease. Int J Mol Sci 2022; 23:ijms232213850. [PMID: 36430330 PMCID: PMC9698384 DOI: 10.3390/ijms232213850] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
In the human body, copper is an important trace element and is a cofactor for several important enzymes involved in energy production, iron metabolism, neuropeptide activation, connective tissue synthesis, and neurotransmitter synthesis. Copper is also necessary for cellular processes, such as the regulation of intracellular signal transduction, catecholamine balance, myelination of neurons, and efficient synaptic transmission in the central nervous system. Copper is naturally present in some foods and is available as a dietary supplement. Only small amounts of copper are typically stored in the body and a large amount of copper is excreted through bile and urine. Given the critical role of copper in a breadth of cellular processes, local concentrations of copper and the cellular distribution of copper transporter proteins in the brain are important to maintain the steady state of the internal environment. The dysfunction of copper metabolism or regulatory pathways results in an imbalance in copper homeostasis in the brain, which can lead to a myriad of acute and chronic pathological effects on neurological function. It suggests a unique mechanism linking copper homeostasis and neuronal activation within the central nervous system. This article explores the relationship between impaired copper homeostasis and neuropathophysiological progress in brain diseases.
Collapse
|
2
|
Physical-Exercise-Induced Antioxidant Effects on the Brain and Skeletal Muscle. Antioxidants (Basel) 2022; 11:antiox11050826. [PMID: 35624690 PMCID: PMC9138070 DOI: 10.3390/antiox11050826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
Erythroid-related nuclear factor 2 (NRF2) and the antioxidant-responsive-elements (ARE) signaling pathway are the master regulators of cell antioxidant defenses, playing a key role in maintaining cellular homeostasis, a scenario in which proper mitochondrial function is essential. Increasing evidence indicates that the regular practice of physical exercise increases cellular antioxidant defenses by activating NRF2 signaling. This manuscript reviewed classic and ongoing research on the beneficial effects of exercise on the antioxidant system in both the brain and skeletal muscle.
Collapse
|
3
|
Chen J, Jin J, Li K, Shi L, Wen X, Fang F. Progresses and Prospects of Neuroprotective Agents-Loaded Nanoparticles and Biomimetic Material in Ischemic Stroke. Front Cell Neurosci 2022; 16:868323. [PMID: 35480961 PMCID: PMC9035592 DOI: 10.3389/fncel.2022.868323] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/16/2022] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke remains the leading cause of death and disability, while the main mechanisms of dominant neurological damage in stroke contain excitotoxicity, oxidative stress, and inflammation. The clinical application of many neuroprotective agents is limited mainly due to their inability to cross the blood-brain barrier (BBB), short half-life and low bioavailability. These disadvantages can be better eliminated/reduced by nanoparticle as the carrier of these drugs. This review expounded the currently hot researched nanomedicines from the perspective of the mechanism of ischemic stroke. In addition, this review describes the bionic nanomedicine delivery strategies containing cells, cell membrane vesicles and exosomes that can effectively avoid the risk of clearance by the reticuloendothelial system. The potential challenges and application prospect for clinical translation of these delivery platforms were also discussed.
Collapse
Affiliation(s)
- Junfa Chen
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jing Jin
- Laboratory Medicine Center, Zhejiang Center for Clinical Laboratory, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Kaiqiang Li
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Lin Shi
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - Xuehua Wen
- Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Xuehua Wen,
| | - Fuquan Fang
- Department of Anesthesiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Fuquan Fang,
| |
Collapse
|
4
|
Wu SH, Yu JH, Liao YT, Liu KH, Chiang ER, Chang MC, Wang JP. Comparison of the Infant and Adult Adipose-Derived Mesenchymal Stem Cells in Proliferation, Senescence, Anti-oxidative Ability and Differentiation Potential. Tissue Eng Regen Med 2022; 19:589-601. [PMID: 35247199 PMCID: PMC9130449 DOI: 10.1007/s13770-022-00431-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/26/2021] [Accepted: 01/05/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Infant adipose-derived mesenchymal stem cells (ADSCs) collected from excised polydactyly fat tissue, which was surgical waste, could be cultured and expanded in vitro in this study. In addition, the collecting process would not cause pain in the host. In this study, the proliferation, reduction of senescence, anti-oxidative ability, and differentiation potential in the infant ADSCs were compared with those in the adult ADSCs harvested from thigh liposuction to determine the availability of infant ADSCs. METHODS Proliferation was determined by detecting the fold changes in cell numbers and doubling time periods. Senescence was analyzed by investigating the age-related gene expression levels and the replicative stress. The superoxide dismutase (SOD) gene expression, adipogenic, neurogenic, osteogenic, and tenogenic differentiation were compared by RT-qPCR. The chondrogenic differentiation efficiency was also determined using RT-qPCR and immunohistochemical staining. RESULTS The proliferation, SOD (SOD1, SOD2 and SOD3) gene expression, the stemness-related gene (c-MYC) and telomerase reverse transcriptase of the infant ADSCs at early passages were enhanced compared with those of the adults'. Cellular senescence related genes, including p16, p21 and p53, and replicative stress were reduced in the infant ADSCs. The adipogenic genes (PPARγ and LPL) and neurogenic genes (MAP2 and NEFH) of the infant ADSC differentiated cells were significantly higher than those of the adults' while the expression of the osteogenic genes (OCN and RUNX) and tenogenic genes (TNC and COL3A1) of both demonstrated opposite results. The chondrogenic markers (SOX9, COL2 and COL10) were enhanced in the infant ADSC differentiated chondrogenic pellets, and the expression levels of SODs were decreased during the differentiation process. CONCLUSION Cultured infant ADSCs demonstrate less cellular senescence and replicative stress, higher proliferation rates, better antioxidant defense activity, and higher potential of chondrogenic, adipogenic and neurogenic differentiation.
Collapse
Affiliation(s)
- Szu-Hsien Wu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, 112 Taiwan ,Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jin-Huei Yu
- Department of Orthopedic Surgery, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, 33004 Taiwan
| | - Yu-Ting Liao
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Kuo-Hao Liu
- Department of Orthopaedics, National Yang Ming Chiao Tung University Hospital, Yilan, 260 Taiwan
| | - En-Rung Chiang
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Ming-Chau Chang
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| | - Jung-pan Wang
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112 Taiwan ,Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, 112 Taiwan
| |
Collapse
|
5
|
Li C, Sun T, Jiang C. Recent advances in nanomedicines for the treatment of ischemic stroke. Acta Pharm Sin B 2021; 11:1767-1788. [PMID: 34386320 PMCID: PMC8343119 DOI: 10.1016/j.apsb.2020.11.019] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/27/2020] [Accepted: 09/13/2020] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke is a cerebrovascular disease normally caused by interrupted blood supply to the brain. Ischemia would initiate the cascade reaction consisted of multiple biochemical events in the damaged areas of the brain, where the ischemic cascade eventually leads to cell death and brain infarction. Extensive researches focusing on different stages of the cascade reaction have been conducted with the aim of curing ischemic stroke. However, traditional treatment methods based on antithrombotic therapy and neuroprotective therapy are greatly limited for their poor safety and treatment efficacy. Nanomedicine provides new possibilities for treating stroke as they could improve the pharmacokinetic behavior of drugs in vivo, achieve effective drug accumulation at the target site, enhance the therapeutic effect and meanwhile reduce the side effect. In this review, we comprehensively describe the pathophysiology of stroke, traditional treatment strategies and emerging nanomedicines, summarize the barriers and methods for transporting nanomedicine to the lesions, and illustrate the latest progress of nanomedicine in treating ischemic stroke, with a view to providing a new feasible path for the treatment of cerebral ischemia.
Collapse
Key Words
- AEPO, asialo-erythropoietin
- APOE, apolipoprotein E
- BBB, blood‒brain barrier
- BCECs, brain capillary endothelial cells
- Blood‒brain barrier
- CAT, catalase
- COX-1, cyclooxygenase-1
- CXCR-4, C-X-C chemokine receptor type 4
- Ce-NPs, ceria nanoparticles
- CsA, cyclosporine A
- DAMPs, damage-associated molecular patterns
- GFs, growth factors
- GPIIb/IIIa, glycoprotein IIb/IIIa
- HMGB1, high mobility group protein B1
- Hb, hemoglobin
- ICAM-1, intercellular adhesion molecule-1
- IL-1β, interleukin-1β
- IL-6, interleukin-6
- Ischemic cascade
- LFA-1, lymphocyte function-associated antigen-1
- LHb, liposomal Hb
- MCAO, middle cerebral artery occlusion
- MMPs, matrix metalloproteinases
- MSC, mesenchymal stem cell
- NF-κB, nuclear factor-κB
- NGF, nerve growth factor
- NMDAR, N-methyl-d-aspartate receptor
- NOS, nitric oxide synthase
- NPs, nanoparticles
- NSCs, neural stem cells
- Nanomedicine
- Neuroprotectant
- PBCA, poly-butylcyanoacrylate
- PCMS, poly (chloromethylstyrene)
- PEG, poly-ethylene-glycol
- PEG-PLA, poly (ethylene-glycol)-b-poly (lactide)
- PLGA NPs, poly (l-lactide-co-glycolide) nanoparticles
- PSD-95, postsynaptic density protein-95
- PSGL-1, P-selectin glycoprotein ligand-1
- RBCs, red blood cells
- RES, reticuloendothelial system
- RGD, Arg-Gly-Asp
- ROS, reactive oxygen species
- Reperfusion
- SDF-1, stromal cell-derived factor-1
- SHp, stroke homing peptide
- SOD, superoxide dismutase
- SUR1-TRPM4, sulfonylurea receptor 1-transient receptor potential melastatin-4
- Stroke
- TEMPO, 2,2,6,6-tetramethylpiperidine-1-oxyl
- TIA, transient ischemic attack
- TNF-α, tumor necrosis factor-α
- Thrombolytics
- cRGD, cyclic Arg-Gly-Asp
- e-PAM-R, arginine-poly-amidoamine ester
- iNOS, inducible nitric oxide synthase
- miRNAs, microRNAs
- nNOS, neuron nitric oxide synthase
- siRNA, small interfering RNA
Collapse
|
6
|
González-Nieto D, Fernández-Serra R, Pérez-Rigueiro J, Panetsos F, Martinez-Murillo R, Guinea GV. Biomaterials to Neuroprotect the Stroke Brain: A Large Opportunity for Narrow Time Windows. Cells 2020; 9:E1074. [PMID: 32357544 PMCID: PMC7291200 DOI: 10.3390/cells9051074] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke represents one of the most prevalent pathologies in humans and is a leading cause of death and disability. Anti-thrombolytic therapy with tissue plasminogen activator (t-PA) and surgical thrombectomy are the primary treatments to recanalize occluded vessels and normalize the blood flow in ischemic and peri-ischemic regions. A large majority of stroke patients are refractory to treatment or are not eligible due to the narrow time window of therapeutic efficacy. In recent decades, we have significantly increased our knowledge of the molecular and cellular mechanisms that inexorably lead to progressive damage in infarcted and peri-lesional brain areas. As a result, promising neuroprotective targets have been identified and exploited in several stroke models. However, these considerable advances have been unsuccessful in clinical contexts. This lack of clinical translatability and the emerging use of biomaterials in different biomedical disciplines have contributed to developing a new class of biomaterial-based systems for the better control of drug delivery in cerebral disorders. These systems are based on specific polymer formulations structured in nanoparticles and hydrogels that can be administered through different routes and, in general, bring the concentrations of drugs to therapeutic levels for prolonged times. In this review, we first provide the general context of the molecular and cellular mechanisms impaired by cerebral ischemia, highlighting the role of excitotoxicity, inflammation, oxidative stress, and depolarization waves as the main pathways and targets to promote neuroprotection avoiding neuronal dysfunction. In the second part, we discuss the versatile role played by distinct biomaterials and formats to support the sustained administration of particular compounds to neuroprotect the cerebral tissue at risk of damage.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - Rocío Fernández-Serra
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain;
- Brain Plasticity Group, Health Research Institute of the Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain; (R.F.-S.); (J.P.-R.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
7
|
Neurovascular protection by peroxisome proliferator-activated receptor α in ischemic stroke. Exp Neurol 2020; 331:113323. [PMID: 32320699 DOI: 10.1016/j.expneurol.2020.113323] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Currently, the only pharmacological therapy for ischemic stroke is thrombolysis with tissue plasminogen activator that has a narrow therapeutic window and increases the risk of intracerebral hemorrhage. New pharmacological treatments for ischemic stroke are desperately needed, but no neuroprotective drugs have successfully made it through clinical trials. Beneficial effects of peroxisome proliferator-activated receptor alpha (PPARα) activation on vascular integrity and function have been reported, and PPARα agonists have clinically been used for many years to manage cardiovascular disease. Thus, PPARα has gained interest in recent years as a target for neurovascular disease such as ischemic stroke. Accumulating preclinical evidence suggests that PPARα activation modulates several pathophysiological hallmarks of stroke such as oxidative stress, blood-brain barrier (BBB) dysfunction, and neuroinflammation to improve functional recovery. Therefore, this review summarizes the various actions PPARα exerts in neurovascular health and disease and the potential of employing exogenous PPARα agonists for future pharmacological treatment of ischemic stroke.
Collapse
|
8
|
Intra- and extra-hospital improvement in ischemic stroke patients: influence of reperfusion therapy and molecular mechanisms. Sci Rep 2020; 10:3513. [PMID: 32103074 PMCID: PMC7044227 DOI: 10.1038/s41598-020-60216-x] [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: 11/29/2018] [Accepted: 01/31/2020] [Indexed: 12/31/2022] Open
Abstract
Neuroprotective treatments in ischemic stroke are focused to reduce the pernicious effect of excitotoxicity, oxidative stress and inflammation. However, those cellular and molecular mechanisms may also have beneficial effects, especially during the late stages of the ischemic stroke. The objective of this study was to investigate the relationship between the clinical improvement of ischemic stroke patients and the time-dependent excitotoxicity and inflammation. We included 4295 ischemic stroke patients in a retrospective study. The main outcomes were intra and extra-hospital improvement. High glutamate and IL-6 levels at 24 hours were associated with a worse intra-hospital improvement (OR:0.993, 95%CI: 0.990–0.996 and OR:0.990, 95%CI: 0.985–0.995). High glutamate and IL-6 levels at 24 hours were associated with better extra-hospital improvement (OR:1.13 95%CI, 1.07–1.12 and OR:1.14, 95%CI, 1.09–1.18). Effective reperfusion after recanalization showed the best clinical outcome. However, the long term recovery is less marked in patients with an effective reperfusion. The variations of glutamate and IL6 levels in the first 24 hours clearly showed a relationship between the molecular components of the ischemic cascade and the clinical outcome of patients. Our findings suggest that the rapid reperfusion after recanalization treatment blocks the molecular response to ischemia that is associated with restorative processes.
Collapse
|
9
|
Hausburg MA, Banton KL, Roman PE, Salgado F, Baek P, Waxman MJ, Tanner A, Yoder J, Bar-Or D. Effects of propofol on ischemia-reperfusion and traumatic brain injury. J Crit Care 2019; 56:281-287. [PMID: 32001426 DOI: 10.1016/j.jcrc.2019.12.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/07/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022]
Abstract
Oxidative stress exacerbates brain damage following ischemia-reperfusion and traumatic brain injury (TBI). Management of TBI and critically ill patients commonly involves use of propofol, a sedation medication that acts as a general anesthetic with inherent antioxidant properties. Here we review available evidence from animal model systems and clinical studies that propofol protects against ischemia-reperfusion injury. However, evidence of propofol toxicity in humans exists and manifests as a rare complication, "propofol infusion syndrome" (PRIS). Evidence in animal models suggests that brain injury induces expression of the p75 neurotrophin receptor (p75NTR), which is associated with proapoptotic signaling. p75NTR-mediated apoptosis of neurons is further exacerbated by propofol's superinduction of p75NTR and concomitant inhibition of neurotrophin processing. Propofol is toxic to neurons but not astrocytes, a type of glial cell. Evidence suggests that propofol protects astrocytes from oxidative stress and stimulates astroglial-mediated protection of neurons. One may speculate that in brain injury patients under sedation/anesthesia, propofol provides brain tissue protection or aids in recovery by enhancing astrocyte function. Nevertheless, our understanding of neurologic recovery versus long-term neurological sequelae leading to neurodegeneration is poor, and it is also conceivable that propofol plays a partial as yet unrecognized role in long-term impairment of the injured brain.
Collapse
Affiliation(s)
- Melissa A Hausburg
- Trauma Research Department, Swedish Medical Center, 501 E Hampden, Englewood, CO 80113, USA; Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA; Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075, USA; Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA; Trauma Research Department, Research Medical Center, 2316 E Meyer Blvd, Kansas City, MO 64132, USA; Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214, USA
| | - Kaysie L Banton
- Trauma Research Department, Swedish Medical Center, 501 E Hampden, Englewood, CO 80113, USA
| | - Phillip E Roman
- Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA; Department of Anesthesiology, St. Anthony Hospital, Lakewood, CO 80228, USA
| | - Fernando Salgado
- Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214, USA; Department of Anesthesiology, Wesley Medical Center, Wichita, KS 67214, USA
| | - Peter Baek
- Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075, USA; Department of Anesthesiology, Medical City Plano, Plano, TX 75075, USA
| | - Michael J Waxman
- Department of Critical Care, Research Medical Center, Kansas City, MO 64132, USA
| | - Allen Tanner
- Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA
| | - Jeffrey Yoder
- Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA; Department of Anesthesiology, St. Anthony Hospital, Lakewood, CO 80228, USA
| | - David Bar-Or
- Trauma Research Department, Swedish Medical Center, 501 E Hampden, Englewood, CO 80113, USA; Trauma Research Department, St. Anthony Hospital, 11600 W 2nd Pl, Lakewood, CO 80228, USA; Trauma Research Department, Medical City Plano, 3901 W 15th St, Plano, TX 75075, USA; Trauma Research Department, Penrose Hospital, 2222 N Nevada Ave, Colorado Springs, CO 80907, USA; Trauma Research Department, Research Medical Center, 2316 E Meyer Blvd, Kansas City, MO 64132, USA; Trauma Research Department, Wesley Medical Center, 550 N Hillside St, Wichita, KS 67214, USA; Department of Molecular Biology, Rocky Vista University, 8401 S Chambers Rd, Parker, CO 80134, USA.
| |
Collapse
|
10
|
Rasouli Vani J, Taghi Mohammadi M, Sarami Foroshani M, Rezazade E. Evaluation of the neuroprotective and antioxidant effects of Dorema aucheri extract on cerebral ischaemia-reperfusion injury in rats. PHARMACEUTICAL BIOLOGY 2019; 57:255-262. [PMID: 30957616 PMCID: PMC6461074 DOI: 10.1080/13880209.2019.1597132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
CONTEXT The hydroalcoholic extract of Dorema aucheri Bilhar (Umbelliferae) (DA) leaves, a medicinal plant, has powerful antioxidant properties. OBJECTIVE This study evaluates the neuroprotective effects of pre-treatment with DA leaves extract against cerebral ischaemia-induced brain injury through alteration of the antioxidant capacity. MATERIALS AND METHODS The study was conducted in three groups of Wistar rats (N = 47) as follows; sham, control ischaemic and pre-treated ischaemic groups. Rats were administered a fresh hydroalcoholic extract of DA leaves at a dosage of 200 mg/kg/day for 14 days. Then, the middle cerebral artery (MCA) of the right hemisphere was occluded for 90 min to achieve cerebral ischaemia. After 24 h reperfusion, cerebral infarction and superoxide dismutase (SOD) and catalase activities, as well as malondialdehyde (MDA), glutathione, and NOx contents were determined in the right hemispheres. RESULTS Occlusion of the right MCA caused noticeable cerebral infarction (298 ± 21 mm3) in control ischaemic group, but pre-treatment with DA extract considerably attenuated it (92 ± 14 mm3) in the pre-treated ischaemic group. DA extract significantly decreased the levels of MDA by 28% and NOx by 11% in pre-treated ischaemic group compared to the control ischaemic group. DA extract also enhanced glutathione content by 7%, SOD activity by 16% and catalase activity by 46% in pre-treated ischaemic rats compared to control ischaemic rats. DISCUSSION AND CONCLUSIONS DA is able to improve the antioxidant capacity and injuries of ischaemic brain. It is proposed as a neuroprotectant following cerebral ischaemia to decrease the injuries of ischaemic stroke.
Collapse
Affiliation(s)
- Javad Rasouli Vani
- Neuroscience Research Center Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Neuroscience Research Center Baqiyatallah University of Medical Sciences, Tehran, Iran
- Department of Physiology and Biophysics School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
- CONTACT Mohammad Taghi Mohammadi ; Department of Physiology & Biophysics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Elham Rezazade
- Department of Physiology and Biophysics School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Lei H, Dirren E, Poitry-Yamate C, Schneider BL, Gruetter R, Aebischer P. Evolution of the neurochemical profiles in the G93A-SOD1 mouse model of amyotrophic lateral sclerosis. J Cereb Blood Flow Metab 2019; 39:1283-1298. [PMID: 29400109 PMCID: PMC6668519 DOI: 10.1177/0271678x18756499] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In vivo 1H magnetic resonance spectroscopy (1H-MRS) investigations of amyotrophic lateral sclerosis (ALS) mouse brain may provide neurochemical profiles and alterations in association with ALS disease progression. We aimed to longitudinally follow neurochemical evolutions of striatum, brainstem and motor cortex of mice transgenic for G93A mutant human superoxide dismutase type-1 (G93A-SOD1), an ALS model. Region-specific neurochemical alterations were detected in asymptomatic G93A-SOD1 mice, particularly in lactate (-19%) and glutamate (+8%) of brainstem, along with γ-amino-butyric acid (-30%), N-acetyl-aspartate (-5%) and ascorbate (+51%) of motor cortex. With disease progression towards the end-stage, increased numbers of metabolic changes of G93A-SOD1 mice were observed (e.g. glutamine levels increased in the brainstem (>+66%) and motor cortex (>+54%)). Through ALS disease progression, an overall increase of glutamine/glutamate in G93A-SOD1 mice was observed in the striatum (p < 0.01) and even more so in two motor neuron enriched regions, the brainstem and motor cortex (p < 0.0001). These 1H-MRS data underscore a pattern of neurochemical alterations that are specific to brain regions and to disease stages of the G93A-SOD1 mouse model. These neurochemical changes may contribute to early diagnosis and disease monitoring in ALS patients.
Collapse
Affiliation(s)
- Hongxia Lei
- 1 Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,2 Department of Radiology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Elisabeth Dirren
- 3 Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carole Poitry-Yamate
- 4 Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,5 Positron Emission Tomography Core, Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bernard L Schneider
- 3 Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Rolf Gruetter
- 1 Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,2 Department of Radiology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,4 Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,6 Department of Radiology, Faculty of Medicine, University of Lausanne, Lausanne, Switzerland
| | - Patrick Aebischer
- 3 Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
12
|
Gopalakrishnan P, Shrestha B, Kaskas AM, Green J, Alexander JS, Pattillo CB. Hydrogen sulfide: Therapeutic or injurious in ischemic stroke? PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2019; 26:1-10. [PMID: 30528175 DOI: 10.1016/j.pathophys.2018.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 07/10/2018] [Accepted: 10/22/2018] [Indexed: 01/09/2023]
Abstract
Hydrogen sulfide (H2S) has been identified as a vasodilatory, neuromodulatory, and anti-inflammatory gasotransmitter with antioxidant properties. Studies focused in cardiac tissue suggest H2S functions as a protective agent; however in the central nervous system (CNS) the effects of H2S during states of stress or injury, such as stroke, remain controversial. Currently, the application of H2S donors and modulators in stroke depends on the type of H2S donor and the timing of the therapy.
Collapse
Affiliation(s)
- Priya Gopalakrishnan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - B Shrestha
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - A M Kaskas
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - J Green
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - J S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - C B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA.
| |
Collapse
|
13
|
Antioxidant Properties of Fucoidan Alleviate Acceleration and Exacerbation of Hippocampal Neuronal Death Following Transient Global Cerebral Ischemia in High-Fat Diet-Induced Obese Gerbils. Int J Mol Sci 2019; 20:ijms20030554. [PMID: 30696078 PMCID: PMC6387260 DOI: 10.3390/ijms20030554] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/18/2019] [Accepted: 01/27/2019] [Indexed: 01/01/2023] Open
Abstract
Fucoidan, a natural sulfated polysaccharide, displays various biological activities including antioxidant properties. We examined the neuroprotective effect of fucoidan against transient global cerebral ischemia (tGCI) in high-fat diet (HFD)-induced obese gerbils and its related mechanisms. Gerbils received HFD for 12 weeks and fucoidan (50 mg/kg) daily for the last 5 days during HFD exposure, and they were subjected to 5-min tGCI. Pyramidal cell death was observed only in the CA 1 area (CA1) of the hippocampus in non-obese gerbils 5 days after tGCI. However, in obese gerbils, pyramidal cell death in the CA1 and CA2/3 occurred at 2 days and 5 days, respectively, after tGCI. In the obese gerbils, oxidative stress indicators (dihydroethidium, 8-hydroxyguanine and 4-hydroxy-2-nonenal) were significantly enhanced and antioxidant enzymes (SOD1 and SOD2) were significantly reduced in pre- and post-ischemic phases compared to the non-obese gerbils. Fucoidan treatment attenuated acceleration and exacerbation of tGCI-induced neuronal death in the CA1–3, showing that oxidative stress was significantly reduced, and antioxidant enzymes were significantly increased in pre- and post-ischemic phases. These findings indicate that pretreated fucoidan can relieve the acceleration and exacerbation of ischemic brain injury in an obese state via the attenuation of obesity-induced severe oxidative damage.
Collapse
|
14
|
Ismaeel A, Lavado R, Smith RS, Eidson JL, Sawicki I, Kirk JS, Bohannon WT, Koutakis P. Effects of Limb Revascularization Procedures on Oxidative Stress. J Surg Res 2018; 232:503-509. [DOI: 10.1016/j.jss.2018.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/27/2018] [Accepted: 07/11/2018] [Indexed: 12/20/2022]
|
15
|
Abedi M, Mesbah-Namin SA, Noori-Zadeh A, Tiraihi T, Taheri T. Human wild-type superoxide dismutase 1 gene delivery to rat bone marrow stromal cells: its importance and potential future trends. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:688-694. [PMID: 30140407 PMCID: PMC6098954 DOI: 10.22038/ijbms.2018.27721.6879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Objective(s): Human superoxide dismutase 1 (SOD1) is the cytosolic form of this enzyme it detoxifies superoxide anions and attenuates their toxicities and concomitant detrimental effects on the cells. It is believed that the amount of these enzymes present in the oxidative stress-induced diseases is crucial for preventing disease progression. Transfection of rat bone marrow stromal cells (BMSCs) by a constructed vector carrying the human wild-type SOD1 gene, a non-viral gene transfer method, was the main aim of this study. Materials and Methods: For this purpose, the rat BMSCs were transfected with the vector using Turbofect reagent and then stabilized. Western-blot and real-time PCR were also used for evaluation of SOD1 expression. Results: Data analysis from RT-PCR and Western-blot techniques revealed that the stable transfected cells could secrete human wild-type SOD1 in the supernatant. Also, the total activity of SOD1 was about 0.5±0.09 U/ml and 0.005±0.002 U/ml in the supernatants of the transfected and not-transfected of rat BMSCs, respectively. Conclusion: This study showed that expansion of the stable transfected rat BMSCs by a constructed vector carrying the human wild-type SOD1 gene is capable of secreting the active SOD1 enzyme under ex-vivo conditions. The recommendation of this study is that the same experiment would be applicable for expression of the other form of this enzyme, SOD3, as well. More valuable information could probably be provided about the variety of the diseases caused by superoxide anions toxicities by intervention and application of the non-viral method for expressions of SOD1 and SOD3 enzymes.
Collapse
Affiliation(s)
- Mohsen Abedi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Noori-Zadeh
- Department of Clinical Biochemistry, Faculty of Paramedicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Taher Taheri
- Shefa Neuroscience Research Center, Khatam-Alanbia Hospital, Tehran, Iran
| |
Collapse
|
16
|
Increased Superoxide Dismutase 2 by Allopregnanolone Ameliorates ROS-Mediated Neuronal Death in Mice with Pilocarpine-Induced Status Epilepticus. Neurochem Res 2018; 43:1464-1475. [PMID: 29855848 DOI: 10.1007/s11064-018-2561-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 05/23/2018] [Accepted: 05/26/2018] [Indexed: 12/19/2022]
Abstract
Excessive production of reactive oxygen species (ROS), along with dysfunction of the antioxidant defense system, such as that involving superoxide dismutase (SOD), may play a major role in neuronal death following status epilepticus (SE). Neurosteroids, which are allosteric modulators of the GABAA receptor in cerebral metabolism, have been suggested as being neuroprotective in various animal models; however, their effect to preventing ROS has not been examined. Herein, we investigate the neuroprotective role of allopregnanolone, the prototypical neurosteroid in the brain, in relation to the ROS-mediated neuronal injury. Adult male C57BL/6 mice were subjected to SE and treated with allopregnanolone. Hippocampal cell death was assessed by the terminal deoxynucleotidyl transferase dUTP nick end labeling assay, and ROS production was investigated by in situ detection of oxidized hydroethidine. SOD2 expression was analyzed by both western blot and immunofluorescent staining in the hippocampal subfields. In mice treated with allopregnanolone after SE, hippocampal cell death, DNA fragmentation, oxidative DNA damage, and ROS production were reduced significantly compared to mice subjected to vehicle treatment after SE. Hippocampal SOD2 expression was significantly increased by allopregnanolone. These finding suggest that allopregnanolone plays a neuroprotective role, with not only anticonvulsant but also antioxidant effects, by increasing SOD2 in pilocarpine-induced SE model.
Collapse
|
17
|
Carter JD, Madamanchi NR, Stouffer GA, Runge MS, Cascio WE, Tong H. Ultrafine particulate matter exposure impairs vasorelaxant response in superoxide dismutase 2-deficient murine aortic rings. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2017; 81:106-115. [PMID: 29279024 PMCID: PMC6136421 DOI: 10.1080/15287394.2017.1420504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/19/2017] [Indexed: 05/29/2023]
Abstract
Studies have linked exposure to ultrafine particulate matter (PM) and adverse cardiovascular events. PM-induced oxidative stress is believed to be a key mechanism underlying observed adverse vascular effects. Advanced age is one factor known to decrease antioxidant defenses and confer susceptibility to the detrimental vascular effects seen following PM exposure. The present study was designed to investigate the vasomotor responses following ultrafine PM exposure in wild type (WT) and superoxide dismutase 2-deficient (SOD2+/-) mice that possess decreased antioxidant defense. Thoracic aortic rings isolated from young and aged WT and SOD2+/- mice were exposed to ultrafine PM in a tissue bath system. Aortic rings were then constricted with increasing concentrations of phenylephrine, followed by relaxation with rising amounts of nitroglycerin (NTG). Data demonstrated that ultrafine PM decreased the relaxation response in both young WT and young SOD2+/- mouse aortas, and relaxation was significantly reduced in young SOD2+/- compared to WT mice. Ultrafine PM significantly diminished the NTG-induced relaxation response in aged compared to young mouse aortas. After ultrafine PM exposure, the relaxation response did not differ markedly between aged WT and aged SOD2+/- mice. Data demonstrated that the greater vascular effect in aortic rings in aged mice ex vivo after ultrafine PM exposure may be attributed to ultrafine PM-induced oxidative stress and loss of antioxidant defenses in aged vascular tissue. Consistent with this conclusion is the attenuation of NTG-induced relaxation response in young SOD2+/- mice. ABBREVIATIONS H2O2: hydrogen peroxide; NTG: nitroglycerin; PAH: polycyclic aromatic hydrocarbons; PE: l-phenylephrine; PM: particulate matter; ROS: reactive oxygen species; SOD2: superoxide dismutase 2 deficient; WT: wild type.
Collapse
Affiliation(s)
- Jacqueline D. Carter
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, NC, 27514
| | | | - George A. Stouffer
- McAllister Heart Institute, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514
| | | | - Wayne E. Cascio
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, NC, 27514
| | - Haiyan Tong
- Environmental Public Health Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, Research Triangle Park, NC, 27514
| |
Collapse
|
18
|
The effects of Mucuna pruriens extract on histopathological and biochemical features in the rat model of ischemia. Neuroreport 2017; 28:1195-1201. [PMID: 28953092 DOI: 10.1097/wnr.0000000000000888] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Kim S, Jeong J, Jung HS, Kim B, Kim YE, Lim DS, Kim SD, Song YS. Anti-inflammatory Effect of Glucagon Like Peptide-1 Receptor Agonist, Exendin-4, through Modulation of IB1/JIP1 Expression and JNK Signaling in Stroke. Exp Neurobiol 2017; 26:227-239. [PMID: 28912645 PMCID: PMC5597553 DOI: 10.5607/en.2017.26.4.227] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 12/18/2022] Open
Abstract
Glucagon like peptide-1 (GLP-1) stimulates glucose-dependent insulin secretion. Dipeptidyl peptidase-4 (DPP-4) inhibitors, which block inactivation of GLP-1, are currently in clinical use for type 2 diabetes mellitus. Recently, GLP-1 has also been reported to have neuroprotective effects in cases of cerebral ischemia. We therefore investigated the neuroprotective effects of GLP-1 receptor (GLP-1R) agonist, exendin-4 (ex-4), after cerebral ischemia-reperfusion injury. Transient middle cerebral artery occlusion (tMCAO) was induced in rats by intracerebroventricular (i.c.v.) administration of ex-4 or ex9-39. Oxygen-glucose deprivation was also induced in primary neurons, bEnd.3 cells, and BV-2. Ischemia-reperfusion injury reduced expression of GLP-1R. Additionally, higher oxidative stress in SOD2 KO mice decreased expression of GLP-1R. Downregulation of GLP-1R by ischemic injury was 70% restored by GLP-1R agonist, ex-4, which resulted in significant reduction of infarct volume. Levels of intracellular cyclic AMP, a second messenger of GLP-1R, were also increased by 2.7-fold as a result of high GLP-1R expression. Moreover, our results showed that ex-4 attenuated pro-inflammatory cyclooxygenase-2 (COX-2) and prostaglandin E2 after MCAO. C-Jun NH2 terminal kinase (JNK) signaling, which stimulates activation of COX-2, was 36% inhibited by i.c.v. injection of ex-4 at 24 h. Islet-brain 1 (IB1), a scaffold regulator of JNK, was 1.7-fold increased by ex-4. GLP-1R activation by ex-4 resulted in reduction of COX-2 through increasing IB1 expression, resulting in anti-inflammatory neuroprotection during stroke. Our study suggests that the anti-inflammatory action of GLP-1 could be used as a new strategy for the treatment of neuroinflammation after stroke accompanied by hyperglycemia.
Collapse
Affiliation(s)
- Soojin Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Jaewon Jeong
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Hye-Seon Jung
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Bokyung Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Ye-Eun Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Da-Sol Lim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - So-Dam Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Yun Seon Song
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| |
Collapse
|
20
|
Hitsumoto T. Impact of Hemorheology Assessed by the Microchannel Method on Pulsatility Index of the Common Carotid Artery in Patients With Type 2 Diabetes Mellitus. J Clin Med Res 2017; 9:579-585. [PMID: 28611858 PMCID: PMC5458655 DOI: 10.14740/jocmr3031w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2017] [Indexed: 12/11/2022] Open
Abstract
Background Type 2 diabetes mellitus is known to be closely associated with the risk of ischemic stroke. Recent clinical studies have reported that a high pulsatility index (PI) of the cerebral or carotid artery, which is estimated by ultrasonography, also reflects a risk of ischemic stroke. This cross-sectional study aimed to clarify the impact of hemorheology assessed by the microchannel method on the PI of the common carotid artery (CCA) in patients with type 2 diabetes mellitus in terms of the primary prevention of ischemic stroke. Methods In total, 349 outpatients on treatment for type 2 diabetes mellitus (131 men and 218 women; mean age ± standard deviation: 65 ± 11 years) with no history of cardiovascular events, including ischemic stroke, were enrolled. The whole blood passage time (WBPT) as a marker of hemorheology and the PI of CCA were measured using commercial devices, and their relationships to various clinical parameters were examined. Results A significant positive correlation was observed between WBPT and the PI of CCA (r = 0.49, P < 0.001). Furthermore, multivariate analysis revealed that patients with high WBPT (≥70 s) had significantly higher risk (odds ratio: 5.2; 95% confidence interval: 2.4 - 9.2; P < 0.001) of being detected with a high PI of CCA (≥ 2) than those with low WBPT (≤ 52.0 s). Conclusion The results of this study indicated that WBPT was an important determination factor for the PI of CCA, suggesting that an increase in WBPT can potentially predict the incidence of ischemic stroke in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Takashi Hitsumoto
- Hitsumoto Medical Clinic, 2-7-7, Takezakicyou, Shimonoseki City, Yamaguchi 750-0025, Japan.
| |
Collapse
|
21
|
Tang N, Wu J, Zhu H, Yan H, Guo Y, Cai Y, Yan H, Shi Y, Shu S, Pei L, Lu Y. Genetic Mutation of GluN2B Protects Brain Cells Against Stroke Damages. Mol Neurobiol 2017; 55:2979-2990. [PMID: 28456939 DOI: 10.1007/s12035-017-0562-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/13/2017] [Indexed: 12/27/2022]
Abstract
Immediately following ischemia, glutamate accumulates in the extracellular space and results in extensive stimulation of its receptors including N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) receptors. A large amount of Ca2+ influx directly through the receptor-gated ion channels which leads to Ca2+ overload and triggers several downstream lethal reactions. As a result, cell dies via apoptosis or necrosis, or both. Death-associated protein kinase 1 (DAPK1) physically and functionally interacts with the NMDA receptor GluN2B subunit at extra-synaptic sites and this interaction acts as a central mediator for stroke damage. The goal of this study is to explore an effective strategy in the treatment of stroke with a molecular genetic manipulation to interrupt DAPK1-GluN2B interaction. We generated a mutant strain of mice with the conditional deletion of GluN2B C-terminal tail consisting of amino acids 886-1269 in the forebrain excitatory neurons (the GluN2B mutant mice) and tested the protective effects of this mutation in stroke damages. GluN2B mutation effectively disrupted the DAPK1-GluN2B interaction and inhibited extra-synaptic NMDA receptor currents without affecting synaptic NMDA receptor channel activity in the central neurons. GluN2B mutation protected against stroke damages both in vitro and in vivo and hence improved behavioral performance. Disruption of the DAPK1-GluN2B interaction is therapeutically effective against stroke damages.
Collapse
Affiliation(s)
- Na Tang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jianhua Wu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Houze Zhu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Honglin Yan
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Guo
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - You Cai
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huanhuan Yan
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Shi
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shu Shu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Pei
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China. .,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
22
|
Sheldon RA, Windsor C, Lee BS, Arteaga Cabeza O, Ferriero DM. Erythropoietin Treatment Exacerbates Moderate Injury after Hypoxia-Ischemia in Neonatal Superoxide Dismutase Transgenic Mice. Dev Neurosci 2017; 39:228-237. [PMID: 28445874 DOI: 10.1159/000472710] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/27/2017] [Indexed: 02/02/2023] Open
Abstract
The neonatal brain is highly susceptible to oxidative stress as developing endogenous antioxidant mechanisms are overwhelmed. In the neonate, superoxide dismutase (SOD) overexpression worsens hypoxic-ischemic injury due to H2O2 accumulation in the brain. Erythropoietin (EPO) is upregulated in 2 phases after HI, early (4 h) and late (7 days), and exogenous EPO has been effective in reducing the injury, possibly through reducing oxidative stress. We hypothesized that exogenous EPO would limit injury from excess H2O2 seen in SOD1-overexpressing mice, and thus enhance recovery after HI. We first wanted to confirm our previous findings in postnatal day 7 (P7) SOD-tg (CD1) mice using a P9 model of the Vannucci procedure of HI with SOD-tg mice from a different background strain (C57Bl/6), and then determine the efficacy of EPO treatment in this strain and their wild-type (WT) littermates. Thus, mice overexpressing copper/zinc SOD1 were subjected to HI, modified for the P9 mouse, and recombinant EPO (5 U/g) or vehicle (saline) was administered intraperitoneally 3 times: at 0 h, 24 h, and 5 days. Injury was assessed 7 days after HI. In addition, protein expression for EPO and EPO receptor was assessed in the cortex and hippocampus 24 h after HI. With the moderate insult, the SOD-tg mice had greater injury than the WT overall, confirming our previous results, as did the hippocampus and striatum when analyzed separately, but not the cortex or thalamus. EPO treatment worsened injury in SOD-tg overall and in the WT and SOD-tg hippocampus and striatum. With the more severe insult, all groups had greater injury than with the moderate insult, but differences between SOD-tg and WT were no longer observed and EPO treatment had no effect. Increased protein expression of EPO was observed in the cortex of SOD-tg mice given recombinant human EPO compared to SOD-tg given vehicle. This study confirms our previous results showing greater injury with SOD overexpression in the neonatal brain after HI at P7 in a different strain. These results also suggest that EPO treatment cannot ameliorate the damage seen in situations where there is excess H2O2 accumulation, and it may exacerbate injury in settings of extreme oxidative stress.
Collapse
Affiliation(s)
- R Ann Sheldon
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | |
Collapse
|
23
|
Park C, Ji HM, Kim SJ, Kil SH, Lee JN, Kwak S, Choe SK, Park R. Fenofibrate exerts protective effects against gentamicin-induced toxicity in cochlear hair cells by activating antioxidant enzymes. Int J Mol Med 2017; 39:960-968. [PMID: 28290603 PMCID: PMC5360428 DOI: 10.3892/ijmm.2017.2916] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 02/24/2017] [Indexed: 12/19/2022] Open
Abstract
Fenofibrate, an activator of peroxisome proliferator-activated receptors (PPARs), has been shown to protect the kidneys and brain cells from oxidative stress; however, its role in preventing hearing loss has not been reported to date, at least to the best of our knowledge. In this study, we demonstrated the protective effects of fenofibrate against gentamicin (GM)-induced ototoxicity. We found that the auditory brainstem response threshold which was increased by GM was significantly reduced by pre-treatment with fenofibrate in rats. In cochlear explants, the disruption of hair cell layers by GM was also markedly attenuated by pre-treatment with fenofibrate. In addition, fenofibrate almost completely abolished GM-induced reactive oxygen species generation, which seemed to be mediated at least in part by the restoration of the expression of PPAR-α-dependent antioxidant enzymes, including catalase and superoxide dismutase (SOD)-1. Of note, fenofibrate markedly increased the expression of heme oxygenase-1 (HO-1) which was also induced to a certain degree by GM alone. The induced expression of HO-1 by fenofibrate appeared to be essential for mediating the protective effects of fenofibrate, as the inhibition of HO-1 activity significantly diminished the protective effects of fenofibrate against the GM-mediated death of sensory hair cells in cochlea explant culture, as well as in zebrafish neuromasts. These results suggest that fenofibrate protects sensory hair cells from GM-induced toxicity by upregulating PPAR-α-dependent antioxidant enzymes, including HO-1. Our results provide insight into the preventive therapy for hearing loss caused by aminoglycoside antibiotics.
Collapse
Affiliation(s)
- Channy Park
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hye-Min Ji
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Se-Jin Kim
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Sung-Hee Kil
- Division of Cell Biology and Genetics, House Research Institute, Los Angeles, CA 90057, USA
| | - Joon No Lee
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Seongae Kwak
- Zoonosis Research Center, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology and Center for Metabolic Function Regulation, Wonkwang University School of Medicine, Iksan, Jeonbuk 54538, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| |
Collapse
|
24
|
Grant Liska M, Crowley MG, Lippert T, Corey S, Borlongan CV. Delta Opioid Receptor and Peptide: A Dynamic Therapy for Stroke and Other Neurological Disorders. Handb Exp Pharmacol 2017; 247:277-299. [PMID: 28315071 DOI: 10.1007/164_2017_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Research of the opioid system and its composite receptors and ligands has revealed its promise as a potential therapy for neurodegenerative diseases such as stroke and Parkinson's Disease. In particular, delta opioid receptors (DORs) have been elucidated as a therapeutically distinguished subset of opioid receptors and a compelling target for novel intervention techniques. Research is progressively shedding light on the underlying mechanism of DORs and has revealed two mechanisms of DOR neuroprotection; DORs function to maintain ionic homeostasis and also to trigger endogenous neuroprotective pathways. Delta opioid agonists such as (D-Ala2, D-Leu5) enkephalin (DADLE) have been shown to promote neuronal survival and decrease apoptosis, resulting in a substantial amount of research for its application as a neurological therapeutic. Most notably, DADLE has demonstrated significant potential to reduce cell death following ischemic events. Current research is working to reveal the complex mechanisms of DADLE's neuroprotective properties. Ultimately, our knowledge of the DOR receptors and agonists has made the opioid system a promising target for therapeutic intervention in many neurological disorders.
Collapse
Affiliation(s)
- M Grant Liska
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Marci G Crowley
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Trenton Lippert
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Sydney Corey
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
25
|
Serum Compounds of Energy Metabolism Impairment Are Related to Disability, Disease Course and Neuroimaging in Multiple Sclerosis. Mol Neurobiol 2016; 54:7520-7533. [DOI: 10.1007/s12035-016-0257-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/24/2016] [Indexed: 12/15/2022]
|
26
|
Katayama Y, Inaba T, Nito C, Suda S, Ueda M. Neuroprotective effects of clarithromycin against neuronal damage in cerebral ischemia and in cultured neuronal cells after oxygen-glucose deprivation. Life Sci 2016; 168:7-15. [PMID: 27825902 DOI: 10.1016/j.lfs.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/27/2016] [Accepted: 11/04/2016] [Indexed: 12/31/2022]
Abstract
AIMS Rats subjected to transient focal ischemia and cultured neuronal cells subjected to oxygen-glucose deprivation (OGD) were treated with clarithromycin (CAM) to evaluate the effects of CAM in protecting against neuronal damage. MAIN METHODS Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 90min and then reperfused. Each animal was given an oral dose clarithromycin (CAM, 100mg/kg) or vehicle alone just after the ischemia was commenced. The infarct volume, edema index and neurological performance were assessed after 24 and 72h of reperfusion. The cerebral blood flow (CBF) was measured with an MRI system at 90min after MCAO. After 24 and 72h, oxidative stress (4-HNE, 8-OHdG) and inflammation (Iba-1, TNF-α) were assessed by immunohistochemical analyses and degenerative cells were assessed in the cortex by Fluoro-Jade C (FJC) labeling. The cultured neuronal cells were also used to examine the effects of CAM exposure on the viability of the cells after OGD. KEY FINDINGS CBF was unchanged between the two groups. Significant reductions of the infarct volume and edema index, an improved neurological deficit score, a significant suppression of 4-HNE and 8-OHdG expression, marked reductions of Iba-1 and TNF-α expression, and a significant reduction of FJC-positive cells were also observed in the CAM-treated animals at both time points. Treatment with 10μM and 100μM CAM in vitro significantly reduced cell death after OGD. SIGNIFICANCE CAM appears to provide antioxidant and anti-inflammatory effects and protect against neuronal damage after cerebral ischemia and OGD.
Collapse
Affiliation(s)
- Yasuo Katayama
- Department of Neurology and Stroke Center, Tokyo General Hospital, 3-15-2 Ekoda Nakano-ku, Tokyo 165-8906, Japan; Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8603, Japan.
| | - Toshiki Inaba
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8603, Japan
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8603, Japan
| | - Satoshi Suda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8603, Japan
| | - Masayuki Ueda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi Bunkyo-ku, Tokyo 113-8603, Japan
| |
Collapse
|
27
|
Abstract
Ischemic brain injury produced by stroke or cardiac arrest is a major cause of human neurological disability. Steady advances in the neurosciences have elucidated the pathophysiological mechanisms of brain ischemia and have suggested many therapeutic approaches to achieve neuroprotection of the acutely ischemic brain that are directed at specific injury mechanisms. In the second portion of this two-part review, the following potential therapeutic approaches to acute ischemic injury are considered: 1) modulation of nonglutamatergic neurotransmission, including monoaminergic systems (dopamine, norepinephrine, serotonin), γ-aminobutyric acid, and adenosine; 2) mild-to-moderate therapeutic hypothermia; 3) calcium channel antagonism; 4) an tagonism of oxygen free radicals; 5) modulation of the nitric oxide system; 6) antagonism of cytoskeletal proteolysis; 7) growth factor administration; 8) therapy directed at cellular mediators of injury; and 9) the rationale for combination pharmacotherapy. The Neuroscientist 1:164-175, 1995
Collapse
Affiliation(s)
- Myron D. Ginsberg
- Cerebral Vascular Disease Research Center Department
of Neurology University of Miami School of Medicine Miami, Florida
| |
Collapse
|
28
|
Abstract
Nitric oxide is a unique biological messenger molecule. It mediates, in part, the immune functions of mac rophages ; it is produced by endothelial cells to mediate blood vessel relaxation; and it also serves as a neurotransmitter in the central and peripheral nervous system. Endothelial nitric oxide synthase and neuronal nitric oxide synthase are thought to be primarily constitutive, with activation induced by calcium entry into cells in the absence of protein synthesis, whereas the macrophage nitric oxide synthase is inducible with large increases in new nitric oxide synthase protein synthesis after immune stimulation. The molecular targets of nitric oxide are expanding, as are its physiological and pathophysiological roles in the nervous system. Nitric oxide may regulate neurotransmitter release, and it may play a key role in nervous system morpho genesis and synaptic plasticity and regulate gene expression. Under conditions of excessive formation, nitric oxide is emerging as an important neurotoxin in a variety of disorders of the nervous system. The Neuro scientist 1:7-18, 1995
Collapse
Affiliation(s)
| | - Valina L. Dawson
- Department of Neurology, Department of Physiology Johns Hopkins University School
of Medicine Baltimore, Maryland
| |
Collapse
|
29
|
Abstract
Global and focal ischemias induce a variety of gene families, including immediate early genes, cytokines, neurotransmitter receptors, and heat-shock proteins. The Janus-like effects of several of these gene prod ucts promote neuronal survival and degeneration. Therefore, determining the molecular pathways respon sible for the differential regulation of these genes is of paramount importance. The discovery of apoptosis as a mediator of delayed neuronal death has led to the identification of a number of other genes involved in postischemic brain damage. Future neuroprotective therapies for cerebral ischemia may be directed at preventing alterations in gene expression. NEUROSCIENTIST 5:238-253, 1999
Collapse
Affiliation(s)
- Sean I. Savitz
- Department of Neurology, Neuroscience, Albert Einstein
College of Medicine Bronx, New York
| | - Daniel M. Rosenbaum
- Department of Neurology, Neuroscience and Ophthalmology
Albert Einstein College of Medicine Bronx, New York
| |
Collapse
|
30
|
Vani JR, Mohammadi MT, Foroshani MS, Jafari M. Polyhydroxylated fullerene nanoparticles attenuate brain infarction and oxidative stress in rat model of ischemic stroke. EXCLI JOURNAL 2016; 15:378-90. [PMID: 27540350 PMCID: PMC4983868 DOI: 10.17179/excli2016-309] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/04/2016] [Indexed: 12/16/2022]
Abstract
Oxidative stress is the common underlying mechanism of damage in ischemic stroke. Therefore, we aimed to evaluate the possible protective effects of polyhydroxylated fullerene derivatives on brain infarction and oxidative/nitrosative stress in a rat model of ischemic stroke. The experiment was performed by four groups of rats (each; n=12); Sham, Control ischemia, and ischemic treatment groups (Pretreatment and Posttreatment). Brain ischemia was induced by 90 min middle cerebral artery occlusion (MCAO) followed by 24 hours reperfusion. Rats received fullerene nanoparticles at dose of 1 mg/kg 30 min before MCAO and immediately after beginning of reperfusion. Infarct volume, contents of malondialdehyde (MDA), glutathione (GSH) and nitrate as well as superoxide dismutase (SOD) activity were assessed 24 hours after termination of MCAO. Brain infarct volume was 310 ± 21 mm3 in control group. Administration of fullerene nanoparticles before and after MCAO significantly decreased the infarct volume by 53 % (145 ± 45 mm3) and 81 % (59 ± 13 mm3), respectively. Ischemia also enhanced MDA and nitrate contents of ischemic hemispheres by 45 % and 25 % , respectively. Fullerene nanoparticles considerably reduced the MDA and nitrate contents of ischemic hemispheres before MCAO by 58 % and 17 % , respectively, and after MCAO by 38 % and 21 % , respectively. Induction of MCAO significantly decreased GSH content (19 % ) and SOD activity (52 % ) of ischemic hemispheres, whereas fullerene nanoparticles increased the GSH content and SOD activity of ischemic hemispheres by 19 % and 52 % before MCAO, respectively, and 21 % and 55 % after MCAO, respectively. Our findings indicate that fullerene nanoparticles, as a potent scavenger of free radicals, protect the brain cells against ischemia/reperfusion injury and inhibit brain oxidative/nitrosative damage.
Collapse
Affiliation(s)
- Javad Rasouli Vani
- Department of Nanotechnology, School of New Sciences and Technology, Islamic Azad University of Pharmaceutical Sciences Branch, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Department of Physiology and Biophysics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahsa Sarami Foroshani
- Department of Nanotechnology, School of New Sciences and Technology, Islamic Azad University of Pharmaceutical Sciences Branch, Tehran, Iran
| | - Mahvash Jafari
- Department of Biochemistry, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Gu SX, Blokhin IO, Wilson KM, Dhanesha N, Doddapattar P, Grumbach IM, Chauhan AK, Lentz SR. Protein methionine oxidation augments reperfusion injury in acute ischemic stroke. JCI Insight 2016; 1:e86460. [PMID: 27294204 PMCID: PMC4902298 DOI: 10.1172/jci.insight.86460] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/19/2016] [Indexed: 12/20/2022] Open
Abstract
Reperfusion injury can exacerbate tissue damage in ischemic stroke, but little is known about the mechanisms linking ROS to stroke severity. Here, we tested the hypothesis that protein methionine oxidation potentiates NF-κB activation and contributes to cerebral ischemia/reperfusion injury. We found that overexpression of methionine sulfoxide reductase A (MsrA), an antioxidant enzyme that reverses protein methionine oxidation, attenuated ROS-augmented NF-κB activation in endothelial cells, in part, by protecting against the oxidation of methionine residues in the regulatory domain of calcium/calmodulin-dependent protein kinase II (CaMKII). In a murine model, MsrA deficiency resulted in increased NF-κB activation and neutrophil infiltration, larger infarct volumes, and more severe neurological impairment after transient cerebral ischemia/reperfusion injury. This phenotype was prevented by inhibition of NF-κB or CaMKII. MsrA-deficient mice also exhibited enhanced leukocyte rolling and upregulation of E-selectin, an endothelial NF-κB-dependent adhesion molecule known to contribute to neurovascular inflammation in ischemic stroke. Finally, bone marrow transplantation experiments demonstrated that the neuroprotective effect was mediated by MsrA expressed in nonhematopoietic cells. These findings suggest that protein methionine oxidation in nonmyeloid cells is a key mechanism of postischemic oxidative injury mediated by NF-κB activation, leading to neutrophil recruitment and neurovascular inflammation in acute ischemic stroke.
Collapse
|
32
|
Watanabe Y, Cohen RA, Matsui R. Redox Regulation of Ischemic Angiogenesis - Another Aspect of Reactive Oxygen Species. Circ J 2016; 80:1278-84. [PMID: 27151566 DOI: 10.1253/circj.cj-16-0317] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antioxidants are expected to improve cardiovascular disease (CVD) by eliminating oxidative stress, but clinical trials have not shown promising results in chronic CVD. Animal studies have revealed that reactive oxygen species (ROS) exacerbate acute CVDs in which high levels of ROS are observed. However, ROS are also necessary for angiogenesis after ischemia, because ROS not only damage cells but also stimulate the cell signaling required for angiogenesis. ROS affect signaling by protein modifications, especially of cysteine amino acid thiols. Although there are several cysteine modifications, S-glutathionylation (GSH adducts; -SSG), a reversible cysteine modification by glutathione (GSH), plays an important role in angiogenic signal transduction by ROS. Glutaredoxin-1 (Glrx) is an enzyme that specifically removes GSH adducts in vivo. Overexpression of Glrx inhibits, whereas deletion of Glrx improves revascularization after mouse hindlimb ischemia. These studies indicate that increased levels of GSH adducts in ischemic muscle are beneficial in promoting angiogenesis. The underlying mechanism can be explained by multiple targets of S-gluathionylation, which mediate the angiogenic effects in ischemia. Increments in the master angiogenic transcriptional factor, HIF-1α, reduction of the anti-angiogenic factor sFlt1, activation of the endoplasmic reticulum Ca(2+)pump, SERCA, and inhibition of phosphatases may occur as a consequence of enhanced S-glutathionylation in ischemic tissue. In summary, inducing S-glutathionylation by inhibiting Glrx may be a therapeutic strategy to improve ischemic angiogenesis in CVD. (Circ J 2016; 80: 1278-1284).
Collapse
Affiliation(s)
- Yosuke Watanabe
- Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine
| | | | | |
Collapse
|
33
|
Katayama Y, Inaba T, Nito C, Ueda M. Neuroprotective effects of erythromycin on ischemic injury following permanent focal cerebral ischemia in rats. Neurol Res 2016; 38:275-84. [PMID: 27078702 DOI: 10.1080/01616412.2016.1138662] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE This study aims to determine if erythromycin provides neuroprotective effects against ischemic injury following permanent focal cerebral ischemia. METHODS Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO). Each animal received a single subcutaneous injection of erythromycin lactobionate (EM, 50 mg/kg) or vehicle immediately after ischemia. The infarct volume, edema index and neurological performance were evaluated at 24 and 72 h after MCAO. The cerebral blood flow (CBF) was measured with an MRI system at 30 min after MCAO. TUNEL staining and immunohistochemical analyses for oxidative stress (4-HNE, 8-OHdG) and inflammation (Iba-1, TNF-α) in the cortex were conducted at 24 and 72 h after MCAO. RESULTS The CBF did not differ between the EM-treated and vehicle-treated groups. The EM treatment significantly reduced the infarct volume (p < 0.01) at 24 and 72 h after MCAO and significantly reduced the edema index (p < 0.01) at 24 h. The EM treatment significantly improved the neurological deficit scores (p < 0.05) at 24 and 72 h. EM also significantly suppressed the accumulation of 4-HNE (p < 0.01) and 8-OHdG (p < 0.01) and markedly reduced Iba-1 (p < 0.01) and TNF-α expression (p < 0.05) at both time points. The EM treatment significantly reduced TUNEL-positive cells (p < 0.01) at both time points. CONCLUSION These findings suggest that EM can protect against the neuronal damage caused by cerebral ischemia by alleviating inflammation and reducing oxidant stress.
Collapse
Affiliation(s)
- Yasuo Katayama
- a Department of Neurology and Stroke Center , Tokyo General Hospital , Tokyo , Japan.,b Graduate School of Medicine , Nippon Medical School , Tokyo , Japan
| | - Toshiki Inaba
- c Department of Neurological Science, Graduate School of Medicine , Nippon Medical School , Tokyo , Japan
| | - Chikako Nito
- c Department of Neurological Science, Graduate School of Medicine , Nippon Medical School , Tokyo , Japan
| | - Masayuki Ueda
- c Department of Neurological Science, Graduate School of Medicine , Nippon Medical School , Tokyo , Japan
| |
Collapse
|
34
|
Tanaka Y, Toyama T, Wada-Takahashi S, Sasaki H, Miyamoto C, Maehata Y, Yoshino F, Yoshida A, Takahashi SS, Watanabe K, Lee MCI, Todoki K, Hamada N. Protective effects of (6R)-5,6,7,8-tetrahydro-l-biopterin on local ischemia/reperfusion-induced suppression of reactive hyperemia in rat gingiva. J Clin Biochem Nutr 2015; 58:69-75. [PMID: 26798200 PMCID: PMC4706094 DOI: 10.3164/jcbn.15-69] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/10/2015] [Indexed: 12/20/2022] Open
Abstract
We herein investigated the regulatory mechanism in the circulation responsible for rat gingival reactive hyperemia (RH) associated with ischemia/reperfusion (I/R). RH was analyzed using a laser Doppler flowmeter. RH and I/R were elicited by gingival compression and release with a laser Doppler probe. RH increased in a time-dependent manner when the duration of compression was between 30 s and 20 min. This increase was significantly suppressed by Nω-nitro-l-arginine-methyl-ester (l-NAME), 7-nitroindazole (7-NI), and 2,4-diamino-6-hydroxypyrimidine (DAHP). However, RH was markedly inhibited following 60 min of compression. This inhibition was significantly decreased by treatments with superoxide dismutase (SOD), (6R)-5,6,7,8-tetrahydro-l-biopterin (BH4), and sepiapterin. The luminescent intensity of superoxide anion (O2•−)-induced 2-methyl-6-(4-methoxyphenyl)-3,7-dihydroimidazo-[1,2-a] pyrazine-3-one (MCLA) was markedly decreased by SOD and BH4, but only slightly by sepiapterin. BH4 significantly decreased O2•− scavenging activity in a time-dependent manner. These results suggested that nitric oxide (NO) secreted by the nitrergic nerve played a role in regulating local circulation in rat gingiva. This NO-related regulation of local circulation was temporarily inhibited in the gingiva by the I/R treatment. The decrease observed in the production of NO, which was caused by suppression of NO synthase (NOS) activity subsequent to depletion of the NOS co-factor BH4 by O2•−, played a partial role in this inhibition.
Collapse
Affiliation(s)
- Yusaku Tanaka
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Toshizo Toyama
- Division of Microbiology, Department of Infection Control, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Satoko Wada-Takahashi
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Haruka Sasaki
- Division of Microbiology, Department of Infection Control, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Chihiro Miyamoto
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Yojiro Maehata
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Fumihiko Yoshino
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Ayaka Yoshida
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Shun-Suke Takahashi
- Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Kiyoko Watanabe
- Division of Microbiology, Department of Infection Control, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Masaichi-Chang-Il Lee
- Yokosuka-Shonan Disaster Health Emergency Research Center & ESR Laboratories, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Kazuo Todoki
- Department of Health Science, School of Nursing, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| | - Nobushiro Hamada
- Division of Microbiology, Department of Infection Control, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan
| |
Collapse
|
35
|
Granger DN, Kvietys PR. Reperfusion injury and reactive oxygen species: The evolution of a concept. Redox Biol 2015; 6:524-551. [PMID: 26484802 PMCID: PMC4625011 DOI: 10.1016/j.redox.2015.08.020] [Citation(s) in RCA: 936] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022] Open
Abstract
Reperfusion injury, the paradoxical tissue response that is manifested by blood flow-deprived and oxygen-starved organs following the restoration of blood flow and tissue oxygenation, has been a focus of basic and clinical research for over 4-decades. While a variety of molecular mechanisms have been proposed to explain this phenomenon, excess production of reactive oxygen species (ROS) continues to receive much attention as a critical factor in the genesis of reperfusion injury. As a consequence, considerable effort has been devoted to identifying the dominant cellular and enzymatic sources of excess ROS production following ischemia-reperfusion (I/R). Of the potential ROS sources described to date, xanthine oxidase, NADPH oxidase (Nox), mitochondria, and uncoupled nitric oxide synthase have gained a status as the most likely contributors to reperfusion-induced oxidative stress and represent priority targets for therapeutic intervention against reperfusion-induced organ dysfunction and tissue damage. Although all four enzymatic sources are present in most tissues and are likely to play some role in reperfusion injury, priority and emphasis has been given to specific ROS sources that are enriched in certain tissues, such as xanthine oxidase in the gastrointestinal tract and mitochondria in the metabolically active heart and brain. The possibility that multiple ROS sources contribute to reperfusion injury in most tissues is supported by evidence demonstrating that redox-signaling enables ROS produced by one enzymatic source (e.g., Nox) to activate and enhance ROS production by a second source (e.g., mitochondria). This review provides a synopsis of the evidence implicating ROS in reperfusion injury, the clinical implications of this phenomenon, and summarizes current understanding of the four most frequently invoked enzymatic sources of ROS production in post-ischemic tissue. Reperfusion injury is implicated in a variety of human diseases and disorders. Evidence implicating ROS in reperfusion injury continues to grow. Several enzymes are candidate sources of ROS in post-ischemic tissue. Inter-enzymatic ROS-dependent signaling enhances the oxidative stress caused by I/R. .
Collapse
Affiliation(s)
- D Neil Granger
- Department of Molecular & Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, United States.
| | - Peter R Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
36
|
Dajas F, Abin-Carriquiry JA, Arredondo F, Blasina F, Echeverry C, Martínez M, Rivera F, Vaamonde L. Quercetin in brain diseases: Potential and limits. Neurochem Int 2015; 89:140-8. [DOI: 10.1016/j.neuint.2015.07.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/07/2015] [Accepted: 07/01/2015] [Indexed: 01/09/2023]
|
37
|
Kim KS, Lee D, Song CG, Kang PM. Reactive oxygen species-activated nanomaterials as theranostic agents. Nanomedicine (Lond) 2015; 10:2709-23. [PMID: 26328770 PMCID: PMC4612518 DOI: 10.2217/nnm.15.108] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) are generated from the endogenous oxidative metabolism or from exogenous pro-oxidant exposure. Oxidative stress occurs when there is excessive production of ROS, outweighing the antioxidant defense mechanisms which may lead to disease states. Hydrogen peroxide (H2O2) is one of the most abundant and stable forms of ROS, implicated in inflammation, cellular dysfunction and apoptosis, which ultimately lead to tissue and organ damage. This review is an overview of the role of ROS in different diseases. We will also examine ROS-activated nanomaterials with emphasis on hydrogen peroxide, and their potential medical implications. Further development of the biocompatible, stimuli-activated agent responding to disease causing oxidative stress, may lead to a promising clinical use.
Collapse
Affiliation(s)
- Kye S Kim
- Cardiovascular Institute, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, MA 02215, USA
| | - Dongwon Lee
- Department of Polymer Nano Science & Technology, Chonbuk National University, Jeonju, Chonbuk 561–756, South Korea
| | - Chul Gyu Song
- Department of Electronic Engineering, Chonbuk National University, Jeonju, Chonbuk 561–756, South Korea
| | - Peter M Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
38
|
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; 23:460-89. [PMID: 24635113 PMCID: PMC4545676 DOI: 10.1089/ars.2013.5778] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [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
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland .,2 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
- 3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| | - Vincent Braunersreuther
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland
| | - François Mach
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland
| | - Nicolas Vuilleumier
- 3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| | - Fabrizio Montecucco
- 1 Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva , Geneva, Switzerland .,2 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 .,3 Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals , Geneva, Switzerland
| |
Collapse
|
39
|
Lu Q, Harris VA, Rafikov R, Sun X, Kumar S, Black SM. Nitric oxide induces hypoxia ischemic injury in the neonatal brain via the disruption of neuronal iron metabolism. Redox Biol 2015. [PMID: 26209813 PMCID: PMC4804102 DOI: 10.1016/j.redox.2015.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We have recently shown that increased hydrogen peroxide (H2O2) generation is involved in hypoxia–ischemia (HI)-mediated neonatal brain injury. H2O2 can react with free iron to form the hydroxyl radical, through Fenton Chemistry. Thus, the objective of this study was to determine if there was a role for the hydroxyl radical in neonatal HI brain injury and to elucidate the underlying mechanisms. Our data demonstrate that HI increases the deposition of free iron and hydroxyl radical formation, in both P7 hippocampal slice cultures exposed to oxygen–glucose deprivation (OGD), and the neonatal rat exposed to HI. Both these processes were found to be nitric oxide (NO) dependent. Further analysis demonstrated that the NO-dependent increase in iron deposition was mediated through increased transferrin receptor expression and a decrease in ferritin expression. This was correlated with a reduction in aconitase activity. Both NO inhibition and iron scavenging, using deferoxamine administration, reduced hydroxyl radical levels and neuronal cell death. In conclusion, our results suggest that increased NO generation leads to neuronal cell death during neonatal HI, at least in part, by altering iron homeostasis and hydroxyl radical generation. HI increases the deposition of free iron and hydroxyl radical formation in the neonatal brain. Both these processes are NO dependent. Increased iron deposition is mediated via increased TfR and decreased ferritin expression. These processes are involved in the neuronal cell death associated with neonatal HI.
Collapse
Affiliation(s)
- Qing Lu
- Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - Valerie A Harris
- Vascular Biology Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Ruslan Rafikov
- Division of Translational and Regenerative Medicine, Department of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Xutong Sun
- Division of Translational and Regenerative Medicine, Department of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Sanjiv Kumar
- Vascular Biology Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, Department of Medicine, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
40
|
Hippophae salicifolia D.Don berries attenuate cerebral ischemia reperfusion injury in a rat model of middle cerebral artery occlusion. JOURNAL OF ACUTE DISEASE 2015. [DOI: 10.1016/s2221-6189(15)30021-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
41
|
Moris D, Georgopoulos S, Felekouras E, Patsouris E, Theocharis S. The effect of endocannabinoid system in ischemia-reperfusion injury: a friend or a foe? Expert Opin Ther Targets 2015; 19:1261-75. [PMID: 25936364 DOI: 10.1517/14728222.2015.1043268] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION In recent years, the endocannabinoid system has emerged as a new therapeutic target in variety of disorders associated with inflammation and tissue injury, including those of the neuronal, liver, renal and cardiovascular system. The aim of the present review is to elucidate the effect of endocannabinoid system on ischemia reperfusion injury (IRI) in different organs and systems. AREAS COVERED The MEDLINE/PubMed database was searched for publications with the medical subject heading Cannabinoids* (CBs), CB receptors*, organ*, ischemia/reperfusion injury*, endocannabinoid* and system*. The initial relevant studies retrieved from the literature were 91 from PubMed. This number was initially limited to 35, after excluding the reviews and studies reporting data for receptors other than cannabinoid. EXPERT OPINION CB2 receptors may play an important compensatory role in controlling tissue inflammation and injury in cells of the neuronal, cardiovascular, liver and renal systems, as well as in infiltrating monocytes/macrophages and leukocytes during various pathological conditions of the systems (atherosclerosis, restenosis, stroke, myocardial infarction, heart, liver and renal failure). These receptors limit inflammation and associated tissue injury. On the basis of preclinical results, pharmacological modulation of CB2 receptors may hold a unique therapeutic potential in stroke, myocardial infarction, atherosclerosis, IRI and liver disease.
Collapse
Affiliation(s)
- Demetrios Moris
- National and Kapodistrian University of Athens , Anastasiou Gennadiou 56, 11474, Athens , Greece +30 210 6440590 ;
| | | | | | | | | |
Collapse
|
42
|
Paspalj D, Nikic P, Savic M, Djuric D, Simanic I, Zivkovic V, Jeremic N, Srejovic I, Jakovljevic V. Redox status in acute ischemic stroke: correlation with clinical outcome. Mol Cell Biochem 2015; 406:75-81. [DOI: 10.1007/s11010-015-2425-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/22/2015] [Indexed: 01/26/2023]
|
43
|
Inaba T, Katayama Y, Ueda M, Nito C. Neuroprotective effects of pretreatment with macrolide antibiotics on cerebral ischemia reperfusion injury. Neurol Res 2015; 37:514-24. [PMID: 25591422 DOI: 10.1179/1743132815y.0000000005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVE This study aims to determine if macrolide antibiotics have neuroprotective effects against transient cerebral ischemia. METHODS Sprague-Dawley rats were subjected to cerebral ischemia for 90 minutes followed by 24 or 72 hours of reperfusion. An oral suspension of roxithromycin (RXM), clarithromycin (CAM), erythromycin (EM), azithromycin (AZM), or kitasamycin (INN) was given at 10 or 100 mg/kg for 7 days before ischemia. The infarct volume, edema volume, and neurological performance were evaluated after 24 and 72 hours of reperfusion. The cerebral blood flow (CBF) was measured with a magnetic resonance imaging (MRI) system after 90 minutes of ischemia. Another experiment was conducted to investigate how the ischemic injury was affected by the interval from the antibiotic pretreatment to the ischemia in rats pretreated with CAM. RESULTS Roxithromycin, CAM, AZM, and INN significantly reduced the infarct volume in the high-dose group after 24 and 72 hours of reperfusion. All of the agents significantly decreased the edema in the high-dose groups at 24 and 72 hours, while only CAM and AZM significantly reduced the edema volume in the low-dose groups at 24 hours. All of the macrolide antibiotics at the high dose significantly improved neurological deficit scores at 24 and 72 hours. There were no differences in the CBF between the vehicle and respective antibiotic groups. In the experiment examining the interval, the 24-hour interval group exhibited the strongest neuroprotective effect. DISCUSSION These results demonstrate that the macrolide antibiotics RXM, CAM, EM, AZM, and INN may confer neuroprotective effects against ischemic damage following cerebral ischemia without affecting the CBF.
Collapse
|
44
|
Ouyang YB, Stary CM, White RE, Giffard RG. The use of microRNAs to modulate redox and immune response to stroke. Antioxid Redox Signal 2015; 22:187-202. [PMID: 24359188 PMCID: PMC4281877 DOI: 10.1089/ars.2013.5757] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Cerebral ischemia is a major cause of death and disability throughout the world, yet therapeutic options remain limited. The interplay between the cellular redox state and the immune response plays a critical role in determining the extent of neural cell injury after ischemia and reperfusion. Excessive amounts of reactive oxygen species (ROS) generated by mitochondria and other sources act both as triggers and effectors of inflammation. This review will focus on the interplay between these two mechanisms. RECENT ADVANCES MicroRNAs (miRNAs) are important post-transcriptional regulators that interact with multiple target messenger RNAs coordinately regulating target genes, including those involved in controlling mitochondrial function, redox state, and inflammatory pathways. This review will focus on the regulation of mitochondria, ROS, and inflammation by miRNAs in the chain of deleterious intra- and intercellular events that lead to brain cell death after cerebral ischemia. CRITICAL ISSUES Although pretreatment using miRNAs was effective in cerebral ischemia in rodents, testing treatment after the onset of ischemia is an essential next step in the development of acute stroke treatment. In addition, miRNA formulation and delivery into the CNS remain a challenge in the clinical translation of miRNA therapy. FUTURE DIRECTIONS Future research should focus on post-treatment and potential clinical use of miRNAs.
Collapse
Affiliation(s)
- Yi-Bing Ouyang
- Department of Anesthesia, Stanford University School of Medicine , Stanford, California
| | | | | | | |
Collapse
|
45
|
Coucha M, Li W, Hafez S, Abdelsaid M, Johnson MH, Fagan SC, Ergul A. SOD1 overexpression prevents acute hyperglycemia-induced cerebral myogenic dysfunction: relevance to contralateral hemisphere and stroke outcomes. Am J Physiol Heart Circ Physiol 2014; 308:H456-66. [PMID: 25552308 DOI: 10.1152/ajpheart.00321.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Admission hyperglycemia (HG) amplifies vascular injury and neurological deficits in acute ischemic stroke, but the mechanisms remain controversial. We recently reported that ischemia-reperfusion (I/R) injury impairs the myogenic response in both hemispheres via increased nitration. However, whether HG amplifies contralateral myogenic dysfunction and whether loss of tone in the contralateral hemisphere contributes to stroke outcomes remain to be determined. Our hypothesis was that contralateral myogenic dysfunction worsens stroke outcomes after acute hyperglycemic stroke in an oxidative stress-dependent manner. Male wild-type or SOD1 transgenic rats were injected with saline or 40% glucose solution 10 min before surgery and then subjected to 30 min of ischemia/45 min or 24 h of reperfusion. In another set of animals (n = 5), SOD1 was overexpressed only in the contralateral hemisphere by stereotaxic adenovirus injection 2-3 wk before I/R. Myogenic tone and neurovascular outcomes were determined. HG exacerbated myogenic dysfunction in contralateral side only, which was associated with infarct size expansion, increased edema, and more pronounced neurological deficit. Global and selective SOD1 overexpression restored myogenic reactivity in ipsilateral and contralateral sides, respectively, and enhanced neurovascular outcomes. In conclusion, our results show that SOD1 overexpression nullified the detrimental effects of HG on myogenic tone and stroke outcomes and that the contralateral hemisphere may be a novel target for the management of acute hyperglycemic stroke.
Collapse
Affiliation(s)
- Maha Coucha
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Weiguo Li
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Sherif Hafez
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia
| | - Mohammed Abdelsaid
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Maribeth H Johnson
- Department of Biostatistics, Georgia Regents University, Augusta, Georgia
| | - Susan C Fagan
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Neurology, Georgia Regents University, Augusta, Georgia; and Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia
| | - Adviye Ergul
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia; Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia
| |
Collapse
|
46
|
Bikis C, Moris D, Vasileiou I, Patsouris E, Theocharis S. FAK/Src family of kinases: protective or aggravating factor for ischemia reperfusion injury in nervous system? Expert Opin Ther Targets 2014; 19:539-49. [DOI: 10.1517/14728222.2014.990374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Ha JS, Dho SH, Youm TH, Kwon KS, Park SS. Astrocytic phospholipase A2 contributes to neuronal glutamate toxicity. Brain Res 2014; 1590:97-106. [DOI: 10.1016/j.brainres.2014.10.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/20/2014] [Accepted: 10/10/2014] [Indexed: 01/10/2023]
|
48
|
Katayama Y, Inaba T, Nito C, Ueda M, Katsura K. Neuroprotective effects of erythromycin on cerebral ischemia reperfusion-injury and cell viability after oxygen-glucose deprivation in cultured neuronal cells. Brain Res 2014; 1588:159-67. [PMID: 25264351 DOI: 10.1016/j.brainres.2014.09.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 09/04/2014] [Accepted: 09/05/2014] [Indexed: 11/16/2022]
Abstract
This study aims to determine if erythromycin has neuroprotective effects against transient ischemia and oxygen-glucose deprivation (OGD) in cultured neuronal cells. Sprague-Dawley rats were subjected to middle cerebral artery occlusion for 90 min, followed by reperfusion. The animals received a subcutaneous single injection of erythromycin lactobionate (EM, 50mg/kg) or vehicle immediately after ischemia. Infarct volume, edema index, and neurological performance were evaluated at 24 and 72 h after reperfusion. Immunohistochemical analyses for oxidative stress (4-HNE, 8-OHdG) and inflammation (Iba-1, TNF-α) were conducted in the cortex at 24h. Primary cortical neuronal cell cultures were prepared from the cerebral cortices of the animals and then subjected to OGD for 3h. Ten or 100 μM EM was added before OGD to determine the effect of EM on cell viability after OGD. EM significantly reduced infarct volume (p<0.01) and edema volume (p<0.05) and improved neurological deficit scores (p<0.05) at 24 and 72 h. EM significantly suppressed the accumulation of 4-HNE (p<0.01) and 8-OHdG (p<0.01) and markedly reduced Iba-1 (p<0.01) and TNF-α expression (p<0.01). Treatment with 100 μM EM in vitro significantly reduced cell death after OGD. EM reduces neuronal damage following cerebral ischemia and OGD and may have antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Yasuo Katayama
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan; Department of Neurology and Stroke Center, Tokyo General Hospital, 3-15-2 Ekoda, Nakano-ku, Tokyo 165-8906, Japan.
| | - Toshiki Inaba
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Chikako Nito
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Masayuki Ueda
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Kenichiro Katsura
- Department of Neurological Science, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| |
Collapse
|
49
|
Free radical scavenging activity and neuroprotective potentials of D138, one Cu(II)/Zn(II) Schiff-base complex derived from N,N'-bis(2-hydroxynaphthylmethylidene)-1,3-propanediamine. Neurochem Res 2014; 39:1834-44. [PMID: 25069642 DOI: 10.1007/s11064-014-1392-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/08/2014] [Accepted: 07/16/2014] [Indexed: 10/25/2022]
Abstract
There is increasing evidence that free radicals play an important role in neuronal damages induced by diabetes mellitus or cerebral ischemia insults. Antioxidants with free radical scavenging activities have been shown to be beneficial and neuroprotective for these pathological conditions. Here, we report free radical scavenging activity and neuroprotective potential of D138, one copper(II)/zinc(II) Schiff-base complex derived from N,N'-2(2-hydroxynaphthylmethylidene)-1,3-propanediamine. The data from three in vitro assays, 2,2-diphenyl-1-picrylhydrazyl assay, nitro blue tetrazolium assay and hydroxyl radical scavenging assay, indicated that D138 presented a potent free radical scavenging activity. The neuroprotective and antioxidative effects of D138 were further evaluated in vivo using bilateral common carotid artery occlusion (BCCAO) mouse model and streptozotocin (STZ) diabetic mouse model. Our results indicated that treatment of D138 significantly ameliorated the hippocampal neuronal damage and the oxidative stress levels in these animal models. Moreover, D138 also reversed the behavioral deficiencies induced by BCCAO or STZ, as assessed by Y-maze test and fear conditioning test. In conclusion, all these findings support that D138 exerts free radical scavenging and neuroprotective activities and has the potentials to be a potent therapeutic candidate for brain oxidative damage induced by cerebral ischemia or diabetes mellitus.
Collapse
|
50
|
Duong TTH, Chami B, McMahon AC, Fong GM, Dennis JM, Freedman SB, Witting PK. Pre-treatment with the synthetic antioxidant T-butyl bisphenol protects cerebral tissues from experimental ischemia reperfusion injury. J Neurochem 2014; 130:733-47. [PMID: 24766199 DOI: 10.1111/jnc.12747] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 04/16/2014] [Accepted: 04/20/2014] [Indexed: 12/30/2022]
Abstract
Treatments to inhibit or repair neuronal cell damage sustained during focal ischemia/reperfusion injury in stroke are largely unavailable. We demonstrate that dietary supplementation with the antioxidant di-tert-butyl-bisphenol (BP) before injury decreases infarction and vascular complications in experimental stroke in an animal model. We confirm that BP, a synthetic polyphenol with superior radical-scavenging activity than vitamin E, crosses the blood-brain barrier and accumulates in rat brain. Supplementation with BP did not affect blood pressure or endogenous vitamin E levels in plasma or cerebral tissue. Pre-treatment with BP significantly lowered lipid, protein and thiol oxidation and decreased infarct size in animals subjected to middle cerebral artery occlusion (2 h) and reperfusion (24 h) injury. This neuroprotective action was accompanied by down-regulation of hypoxia inducible factor-1α and glucose transporter-1 mRNA levels, maintenance of neuronal tissue ATP concentration and inhibition of pro-apoptotic factors that together enhanced cerebral tissue viability after injury. That pre-treatment with BP ameliorates oxidative damage and preserves cerebral tissue during focal ischemic insult indicates that oxidative stress plays at least some causal role in promoting tissue damage in experimental stroke. The data strongly suggest that inhibition of oxidative stress through BP scavenging free radicals in vivo contributes significantly to neuroprotection. We demonstrate that pre-treatment with ditert-butyl bisphenol(Di-t-Bu-BP) inhibits lipid, protein, and total thiol oxidation and decreases caspase activation and infarct size in rats subjected to middle cerebral artery occlusion (2 h) and reperfusion (24 h) injury. These data suggest that inhibition of oxidative stress contributes significantly to neuroprotection.
Collapse
Affiliation(s)
- Thi Thuy Hong Duong
- Vascular Biology Group, ANZAC Research Institute, Concord Hospital, Concord, NSW, Australia
| | | | | | | | | | | | | |
Collapse
|