1
|
Xie J, Wang L, Tian S, Li R, Zhang L, Shi H, Liu Z, Ma T, Hu H, She Z, Wang L. The Protective Role of Transcript-Induced in Spermiogenesis 40 in Cerebral Ischemia-Reperfusion Injury. Neurochem Res 2024; 49:2519-2534. [PMID: 38884889 DOI: 10.1007/s11064-024-04170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/18/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024]
Abstract
Prompt reperfusion after cerebral ischemia is important to maintain neuronal survival and reduce permanent disability and death. However, the resupply of blood can induce oxidative stress, inflammatory response and apoptosis, further leading to tissue damage. Here, we report the versatile biological roles of transcript-induced in spermiogenesis 40 (Tisp40) in ischemic stroke. We found that the expression of Tisp40 was upregulated in ischemia/reperfusion-induced brain tissues and oxygen glucose deprivation/returned -stimulated neurons. Tisp40 deficiency increased the infarct size and neurological deficit score, and promoted inflammation and apoptosis. Tisp40 overexpression played the opposite role. In vitro, the oxygen glucose deprivation/returned model was established in Tisp40 knockdown and overexpression primary cultured cortical neurons. Tisp40 knockdown can aggravate the process of inflammation and apoptosis, and Tisp40 overexpression ameliorated the aforementioned processes. Mechanistically, Tisp40 protected against ischemic stroke via activating the AKT signaling pathway. Tisp40 may be a new therapeutic target in brain ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jing Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Neurosurgery, Huanggang Central Hospital, Huanggang, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Ruyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Zhang
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Hongjie Shi
- Department of Neurosurgery, Huanggang Central Hospital, Huanggang, China
| | - Zhen Liu
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Tengfei Ma
- Department of Neurosurgery, Huanggang Central Hospital, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Heng Hu
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
- Institute of Model Animal of Wuhan University, Wuhan, China.
| | - Lang Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Zaharia AL, Oprea VD, Coadă CA, Tutunaru D, Romila A, Stan B, Croitoru A, Ionescu AM, Lungu M. Serum Caspase-3 Levels as a Predictive Molecular Biomarker for Acute Ischemic Stroke. Int J Mol Sci 2024; 25:6772. [PMID: 38928477 PMCID: PMC11204031 DOI: 10.3390/ijms25126772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Caspases are key players in the apoptotic process and have been found to contribute to the pathogenesis of a variety of diseases, including neurological disorders such as ischemic stroke. This study aimed to investigate the serum levels of Caspase-3 in patients with acute ischemic stroke (AIS) and in control patients without ischemic events. Moreover, we explored any potential associations with the clinical outcomes of AIS. We enrolled 69 consecutive patients with clinical signs and symptoms of AIS in the presence of a negative CT scan who presented themselves at the Clinical Neurological Department from the Emergency Clinical Hospital of Galati within the first 24 h of symptom onset. The control group comprised 68 patients without cerebral ischemic pathologies. A comparison of the two groups showed significantly higher levels of caspase-3 at 24 and 48 h after hospital admission. No significant associations between caspase-3 levels and clinical features of AIS were seen. However, in a subgroup analysis conducted on patients with moderate/severe and severe stroke, lower levels of caspase-3 were associated with early mortality. Caspase-3 levels did not directly correlate with AIS severity or prognosis when considering all AIS patients. In patients with moderate to severe National Institute of Health Stroke Scale (NIHSS) scores, caspase-3 might be a prognostic indicator of early death. Further studies are required to confirm these results and further explore the mechanisms behind these findings.
Collapse
Affiliation(s)
- Andrei-Lucian Zaharia
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| | - Violeta Diana Oprea
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| | - Camelia Alexandra Coadă
- Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Dana Tutunaru
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| | - Aurelia Romila
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| | - Bianca Stan
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| | - Ana Croitoru
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| | - Ana-Maria Ionescu
- Faculty of Medicine and Pharmacy, Ovidius University of Constanța, 900470 Constanța, Romania;
| | - Mihaiela Lungu
- Faculty of Medicine and Pharmacy, “Dunarea de Jos” University of Galati, 800216 Galati, Romania; (A.-L.Z.); (V.D.O.); (A.R.); (B.S.); (A.C.); (M.L.)
- “St. Apostle Andrei” Clinical Emergency County Hospital Galati, 800578 Galati, Romania
| |
Collapse
|
3
|
Han Q, Wang F. Electroacupuncture at GB20 improves cognitive ability and synaptic plasticity via the CaM-CaMKII-CREB signaling pathway following cerebral ischemia-reperfusion injury in rats. Acupunct Med 2024; 42:23-31. [PMID: 38126262 DOI: 10.1177/09645284231202805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
BACKGROUND This study aimed to investigate the effects of electroacupuncture (EA) on cognitive recovery and synaptic remodeling in a rat model of middle cerebral artery occlusion (MCAO) followed by reperfusion and explore the possible mechanism. METHOD Focal cerebral ischemia was modeled in healthy adult Sprague-Dawley rats by MCAO. The MCAO rats were classified into four groups: sham, MCAO, MCAO + GB20 (receiving EA at GB20) and MCAO + NA (receiving EA at a "non-acupoint" location not corresponding to any traditional acupuncture point location about 10 mm above the iliac crest). Neurological deficit scores and behavior were assessed before and during the treatment. After intervention for 7 days, the hippocampus was dissected to analyze growth-associated protein (GAP)-43, synaptophysin (SYN) and postsynaptic density protein (PSD)-95 expression levels by Western blotting. Bioinformatic analysis and primary hippocampal neurons with calcium-voltage gated channel subunit alpha 1B (CACNA1B) gene overexpression were used to screen the target genes for EA against MCAO. RESULTS Significant amelioration of neurological deficits and learning/memory were found in MCAO + GB20 rats compared with MCAO or MCAO + NA rats. Protein levels of GAP-43, SYN and PSD-95 were significantly improved in MCAO + GB20-treated rats together with an increase in the number of synapses in the hippocampal CA1 region. CACNA1B appeared to be a target gene of EA in MCAO. There were increased mRNA levels of CACNA1B, calmodulin (CaM), Ca2+/calmodulin-dependent protein kinase type II (CaMKII) and cyclic adenosine monophosphate response element binding (CREB) and increased phosphorylation of CaM, CaMKII and CREB in the hippocampal region in MCAO + GB20 versus MCAO and MCAO + NA groups. CACNA1B overexpression modulated expression of the CaM-CaMKII-CREB axis. CONCLUSION EA treatment at GB20 may ameliorate the negative effects of MCAO on cognitive function in rats by enhancing synaptic plasticity. EA treatment at GB20 may exert this neuroprotective effect by regulating the CACNA1B-CaM-CaMKII-CREB axis.
Collapse
Affiliation(s)
- Qing Han
- Department of Traditional Chinese Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| | - Feng Wang
- Department of Traditional Chinese Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
4
|
Gong Z, Guo J, Liu B, Guo Y, Cheng C, Jiang Y, Liang N, Hu M, Song T, Yang L, Li H, Zhang H, Zong X, Che Q, Shi N. Mechanisms of immune response and cell death in ischemic stroke and their regulation by natural compounds. Front Immunol 2024; 14:1287857. [PMID: 38274789 PMCID: PMC10808662 DOI: 10.3389/fimmu.2023.1287857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Ischemic stroke (IS), which is the third foremost cause of disability and death worldwide, has inflammation and cell death as its main pathological features. IS can lead to neuronal cell death and release factors such as damage-related molecular patterns, stimulating the immune system to release inflammatory mediators, thereby resulting in inflammation and exacerbating brain damage. Currently, there are a limited number of treatment methods for IS, which is a fact necessitating the discovery of new treatment targets. For this review, current research on inflammation and cell death in ischemic stroke was summarized. The complex roles and pathways of the principal immune cells (microglia, astrocyte, neutrophils, T lymphocytes, and monocytes/macrophage) in the immune system after IS in inflammation are discussed. The mechanisms of immune cell interactions and the cytokines involved in these interactions are summarized. Moreover, the cell death mechanisms (pyroptosis, apoptosis, necroptosis, PANoptosis, and ferroptosis) and pathways after IS are explored. Finally, a summary is provided of the mechanism of action of natural pharmacological active ingredients in the treatment of IS. Despite significant recent progress in research on IS, there remain many challenges that need to be overcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Qianzi Che
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nannan Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Flores AE, Pascotini ET, Kegler A, Broetto N, Gabbi P, Duarte T, Prado ALC, Duarte MMMF, da Cruz IBM, Dos Santos ARS, Royes LFF, Fighera MR. Worst spasticity in patients post-stroke associated with MNSOD ALA16VAL polymorphism and interleukin-1β. Gene X 2022; 847:146880. [PMID: 36100117 DOI: 10.1016/j.gene.2022.146880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/23/2022] [Accepted: 09/05/2022] [Indexed: 11/04/2022] Open
Abstract
The MnSOD Ala16Val single nucleotide polymorphism (SNP) has shown to be associated to risk factors of several metabolic and vascular diseases. However, little is known about interaction between MnSOD Ala16Val SNP in stroke, a frequent neurologic disease that involves clinic manifestations such as motor deficits and spasticity. In this sense, we decided to investigate the relationship between MnSOD Ala16Val SNP with spasticity in stroke and also its influence on interleukin levels, BDNF, and glycolipid parameters. Eighty post-stroke subjects and 80 healthy controls were investigated. We showed a higher spasticity, levels of total cholesterol, LDL, IL-1β, IL-6, and INF-γ in VV post-stroke group. Interesting, we found a correlation between IL-1β levels and spasticity in VV post-stroke. Triglycerides, glucose levels and caspases (1 and 3) activation were significantly higher, as well as BDNF levels were lower in VV and AV post-stroke. DNA damage was higher in post-stroke group. Thus, we can suggest that the V allele has a worse glycolipid profile, which would facilitate changes in neurovascular homeostasis. These events associated with an increase in inflammatory markers and a reduction in BDNF can contribute with the stroke and a worse clinical evolution in relation to spasticity in patients with VV genotype.
Collapse
Affiliation(s)
- Ariane Ethur Flores
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Eduardo Tanuri Pascotini
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Aline Kegler
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil
| | - Núbia Broetto
- Curso de Fisioterapia, Departamento de Ginástica e Saúde, Universidade Federal de Pelotas, RS, Brazil
| | - Patricia Gabbi
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Thiago Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Marta M M F Duarte
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Ivana B M da Cruz
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | | | - Luiz Fernando Freire Royes
- Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Centro de Ciências da Saúde, Departamento de Neuropsiquiatria, Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências Naturais e Exatas, Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, RS, Brazil; Centro de Educação Física e Desportos, Laboratório de Bioquímica do Exercício (BIOEX), Universidade Federal de Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, RS, Brazil.
| |
Collapse
|
6
|
Khan H, Kaur P, Singh TG, Grewal AK, Sood S. Adenosine as a Key Mediator of Neuronal Survival in Cerebral Ischemic Injury. Neurochem Res 2022; 47:3543-3555. [PMID: 36042141 DOI: 10.1007/s11064-022-03737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022]
Abstract
Several experimental studies have linked adenosine's neuroprotective role in cerebral ischemia. During ischemia, adenosine is formed due to intracellular ATP breakdown into ADP, further when phosphate is released from ADP, the adenosine monophosphate is formed. It acts via A1, A2, and A3 receptors found on neurons, blood vessels, glial cells, platelets, and leukocytes. It is related to various effector systems such as adenyl cyclase and membrane ion channels via G-proteins. Pharmacological manipulation of adenosine receptors by agonists (CCPA, ADAC, IB-MECA) increases ischemic brain damage in various in vivo and in vitro models of cerebral ischemia whereas, agonist can also be neuroprotective. Mainly, receptor antagonists (CGS15943, MRS1706) indicated neuroprotection. Later, various studies also revealed that the downregulation or upregulation of specific adenosine receptors is necessary during the recovery of cerebral ischemia by activating several downstream signaling pathways. In the current review, we elaborate on the dual roles of adenosine and its receptor subtypes A1, A2, and A3 and their involvement in the pathobiology of cerebral ischemic injury. Adenosine-based therapies have the potential to improve the outcomes of cerebral injury patients, thereby providing them with a more optimistic future.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Parneet Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurejet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Shreya Sood
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| |
Collapse
|
7
|
Biological Effects and Mechanisms of Caspases in Early Brain Injury after Subarachnoid Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3345637. [PMID: 35847583 PMCID: PMC9277153 DOI: 10.1155/2022/3345637] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 06/22/2022] [Indexed: 12/12/2022]
Abstract
Caspases are an evolutionarily conserved family of proteases responsible for mediating and initiating cell death signals. In the past, the dysregulated activation of caspases was reported to play diverse but equally essential roles in neurodegenerative diseases, such as brain injury and neuroinflammatory diseases. A subarachnoid hemorrhage (SAH) is a traumatic event that is either immediately lethal or induces a high risk of stroke and neurological deficits. Currently, the prognosis of SAH after treatment is not ideal. Early brain injury (EBI) is considered one of the main factors contributing to the poor prognosis of SAH. The mechanisms of EBI are complex and associated with oxidative stress, neuroinflammation, blood-brain barrier disruption, and cell death. Based on mounting evidence, caspases are involved in neuronal apoptosis or death, endothelial cell apoptosis, and increased inflammatory cytokine-induced by apoptosis, pyroptosis, and necroptosis in the initial stages after SAH. Caspases can simultaneously mediate multiple death modes and regulate each other. Caspase inhibitors (including XIAP, VX-765, and Z-VAD-FMK) play an essential role in ameliorating EBI after SAH. In this review, we explore the related pathways mediated by caspases and their reciprocal regulation patterns after SAH. Furthermore, we focus on the extensive crosstalk of caspases as a potential area of research on therapeutic strategies for treating EBI after SAH.
Collapse
|
8
|
Lu Y, Wang J, Tang F, Pratap UP, Sareddy GR, Dhandapani KM, Capuano A, Arvanitakis Z, Vadlamudi RK, Brann DW. Regulation and Role of Neuron-Derived Hemoglobin in the Mouse Hippocampus. Int J Mol Sci 2022; 23:5360. [PMID: 35628182 PMCID: PMC9140924 DOI: 10.3390/ijms23105360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022] Open
Abstract
Hemoglobin (Hb) is the oxygen transport protein in erythrocytes. In blood, Hb is a tetramer consisting of two Hb-alpha (Hb-α) chains and two Hb-beta (Hb-β) chains. A number of studies have also shown that Hb-α is also expressed in neurons in both the rodent and human brain. In the current study, we examined for age-related regulation of neuronal Hb-α and hypoxia in the hippocampus and cerebral cortex of intact male and female mice. In addition, to confirm the role and functions of neuronal Hb-α, we also utilized lentivirus CRISPR interference-based Hb-α knockdown (Hb-α CRISPRi KD) in the non-ischemic and ischemic mouse hippocampus and examined the effect on neuronal oxygenation, as well as induction of hypoxia-inducible factor-1α (HIF-1α) and its downstream pro-apoptotic factors, PUMA and NOXA, and on neuronal survival and neurodegeneration. The results of the study revealed an age-related decrease in neuronal Hb-α levels and correlated increase in hypoxia in the hippocampus and cortex of intact male and female mice. Sex differences were observed with males having higher neuronal Hb-α levels than females in all brain regions at all ages. In vivo Hb-α CRISPRi KD in the mouse hippocampus resulted in increased hypoxia and elevated levels of HIF-1α, PUMA and NOXA in the non-ischemic and ischemic mouse hippocampus, effects that were correlated with a significant decrease in neuronal survival and increased neurodegeneration. As a whole, these findings indicate that neuronal Hb-α decreases with age in mice and has an important role in regulating neuronal oxygenation and neuroprotection.
Collapse
Affiliation(s)
- Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (Y.L.); (K.M.D.)
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.W.); (F.T.)
| | - Fulei Tang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.W.); (F.T.)
| | - Uday P. Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (U.P.P.); (G.R.S.); (R.K.V.)
| | - Gangadhara R. Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (U.P.P.); (G.R.S.); (R.K.V.)
| | - Krishnan M. Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (Y.L.); (K.M.D.)
| | - Ana Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA; (A.C.); (Z.A.)
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA; (A.C.); (Z.A.)
| | - Ratna K. Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antonio, TX 78229, USA; (U.P.P.); (G.R.S.); (R.K.V.)
| | - Darrell W. Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.W.); (F.T.)
| |
Collapse
|
9
|
Aronowski J, Sansing LH, Xi G, Zhang JH. Mechanisms of Damage After Cerebral Hemorrhage. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
10
|
Post-Ischemic Neurodegeneration of the Hippocampus Resembling Alzheimer's Disease Proteinopathy. Int J Mol Sci 2021; 23:ijms23010306. [PMID: 35008731 PMCID: PMC8745293 DOI: 10.3390/ijms23010306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 12/26/2021] [Accepted: 12/26/2021] [Indexed: 12/14/2022] Open
Abstract
In this review, we summarize, inter alia, the protein and gene changes associated with Alzheimer’s disease and their role in post-ischemic hippocampal neurodegeneration. In the hippocampus, studies have revealed dysregulation of the genes for the amyloid protein precursor metabolism and tau protein that is identical in nature to Alzheimer’s disease. Data indicate that amyloid and tau protein, derived from brain tissue and blood due to increased permeability of the blood–brain barrier after ischemia, play a key role in post-ischemic neurodegeneration of the hippocampus, with concomitant development of full-blown dementia. Thus, the knowledge of new neurodegenerative mechanisms that cause neurodegeneration of the hippocampus after ischemia, resembling Alzheimer’s disease proteinopathy, will provide the most important therapeutic development goals to date.
Collapse
|
11
|
Wasan H, Singh D, Joshi B, Upadhyay D, Sharma U, Dinda AK, Reeta KH. Dihydromyricetin alleviates cerebral ischemia-reperfusion injury by attenuating apoptosis and astrogliosis in peri-infarct cortex. Neurol Res 2021; 44:403-414. [PMID: 34882069 DOI: 10.1080/01616412.2021.1997010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVES In ischemic stroke, reperfusion after thrombolysis is associated with secondary brain damage. Dihydromyricetin (DHM), a flavonoid, has shown neuroprotective effects through anti-oxidant, anti-inflammatory and anti-apoptotic properties. This study investigates the potential of DHM, given postreperfusion in middle cerebral artery occlusion (MCAo) model of stroke in rats. METHODS MCAo surgery was performed in male Wistar rats. Reperfusion was performed after 90 min of ischemia. DHM (50 and 100 mg/kg) was administered 10-15 min and 2 h postreperfusion followed by daily dosing for 2 more days. Neurobehavioral parameters and infarct size (TTC staining) were assessed after 72 h. The effective dose (100 mg/kg) was then used to study reduction in infarct size (measured by MRI) and effect on apoptosis (evaluated by protein expression of Bax, Bcl-2 and cleaved caspase-3 and TUNEL assay) in peri-infarct cortex. Furthermore, effects of DHM on neuronal damage and activation of astrocytes were studied by immunofluorescence. RESULTS Poststroke DHM (100 mg/kg) administered for 3 days showed significant improvements in motor-coordination and infarct damage (TTC staining and MRI). MCAo-induced altered apoptotic proteins were normalized to a significant extent in peri-infarct cortex with DHM treatment. Data from TUNEL assay were complementary to the effects on apoptotic proteins. Additionally, DHM caused a significant reduction in the number of reactive astrocytes when compared with the MCAo group. DISCUSSION This study demonstrated the efficacy of subacute DHM treatment in ischemia/reperfusion injury by modulating apoptosis and astrogliosis in the peri-infarct cortex. This suggests the potential of DHM in attenuating disease progression.
Collapse
Affiliation(s)
- Himika Wasan
- Department of Pharmacology, AIIMS, New Delhi, India
| | | | - Balu Joshi
- Department of Pharmacology, AIIMS, New Delhi, India
| | | | - Uma Sharma
- Department of NMR, AIIMS, New Delhi, India
| | | | - K H Reeta
- Department of Pharmacology, AIIMS, New Delhi, India
| |
Collapse
|
12
|
Protective effects of melatonin and L-carnitine against methotrexate-induced toxicity in isolated rat hepatocytes. Naunyn Schmiedebergs Arch Pharmacol 2021; 395:87-97. [PMID: 34821957 DOI: 10.1007/s00210-021-02176-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
The present study was designed to evaluate the possible protective effects of melatonin (MEL) and/or L-carnitine (L-CAR) against methotrexate (MTX)-induced toxicity in isolated rat hepatocytes. Hepatocytes were prepared using collagenase techniques of perfusion and digestion of rat liver. Trypan blue uptake, as well as, glutathione (GSH), lipid peroxidation (LPO), nitric oxide (NO), and tumor necrosis factor-alpha (TNF-α) levels were measured. Caspase-3 activity was also assessed. Pre-incubation of hepatocytes with MEL (1 mM) and/or L-CAR (10 mM) 30 min prior to intoxication with MTX, significantly protected hepatocytes against toxicity. In addition, LPO, NO, TNF-α levels, and caspase-3 activity were decreased in comparison to the MTX-intoxicated group. Furthermore, the two drugs increased the MTX-depleted GSH level. MEL and L-CAR prevented MTX-induced hepatocytotoxicity, at least partly, by their antioxidative, antiinflammatory, and antiapoptotic effects. Further studies are recommended on the clinical pharmacologic and toxicologic effects of MEL and L-CAR in patients receiving MTX.
Collapse
|
13
|
Wiklund L, Sharma A, Patnaik R, Muresanu DF, Sahib S, Tian ZR, Castellani RJ, Nozari A, Lafuente JV, Sharma HS. Upregulation of hemeoxygenase enzymes HO-1 and HO-2 following ischemia-reperfusion injury in connection with experimental cardiac arrest and cardiopulmonary resuscitation: Neuroprotective effects of methylene blue. PROGRESS IN BRAIN RESEARCH 2021; 265:317-375. [PMID: 34560924 DOI: 10.1016/bs.pbr.2021.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Oxidative stress plays an important role in neuronal injuries after cardiac arrest. Increased production of carbon monoxide (CO) by the enzyme hemeoxygenase (HO) in the brain is induced by the oxidative stress. HO is present in the CNS in two isoforms, namely the inducible HO-1 and the constitutive HO-2. Elevated levels of serum HO-1 occurs in cardiac arrest patients and upregulation of HO-1 in cardiac arrest is seen in the neurons. However, the role of HO-2 in cardiac arrest is not well known. In this review involvement of HO-1 and HO-2 enzymes in the porcine brain following cardiac arrest and resuscitation is discussed based on our own observations. In addition, neuroprotective role of methylene blue- an antioxidant dye on alterations in HO under in cardiac arrest is also presented. The biochemical findings of HO-1 and HO-2 enzymes using ELISA were further confirmed by immunocytochemical approach to localize selective regional alterations in cardiac arrest. Our observations are the first to show that cardiac arrest followed by successful cardiopulmonary resuscitation results in significant alteration in cerebral concentrations of HO-1 and HO-2 levels indicating a prominent role of CO in brain pathology and methylene blue during CPR followed by induced hypothermia leading to superior neuroprotection after return of spontaneous circulation (ROSC), not reported earlier.
Collapse
Affiliation(s)
- Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Yao Y, Ji Y, Ren J, Liu H, Khanna R, Sun L. Inhibition of autophagy by CRMP2-derived peptide ST2-104 (R9-CBD3) via a CaMKKβ/AMPK/mTOR pathway contributes to ischemic postconditioning-induced neuroprotection against cerebral ischemia-reperfusion injury. Mol Brain 2021; 14:123. [PMID: 34362425 PMCID: PMC8344221 DOI: 10.1186/s13041-021-00836-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/28/2021] [Indexed: 01/03/2023] Open
Abstract
Cerebral ischemia, a common cerebrovascular disease, is characterized by functional deficits and apoptotic cell death. Autophagy, a type of programmed cell death, plays critical roles in controlling neuronal damage and metabolic homeostasis, and has been inextricably linked to cerebral ischemia. We previously identified a short peptide aptamer from collapsin response mediator protein 2 (CRMP2), designated the Ca2+ channel-binding domain 3 (CBD3) peptide, that conferred protection against excitotoxicity and traumatic brain injury. ST2-104, a nona-arginine (R9)-fused CBD3 peptide, exerted beneficial effects on neuropathic pain and was neuroprotective in a model of Alzheimer's disease; however, the effect of ST2-104 on cerebral ischemia and its mechanism of action have not been studied. In this study, we modeled cerebral ischemia-reperfusion injury in rats with the middle cerebral artery occlusion (MCAO) as well as challenged SH-SY5Y neuroblastoma cells with glutamate to induce toxicity to interrogate the effects of ST2-104 on autophagy following ischemic/excitotoxic insults. ST2-104 reduced the infarct volume and improved the neurological score of rats subjected to MCAO. ST2-104 protected SH-SY5Y cells from death following glutamate exposure via blunting apoptosis and autophagy as well as limiting excessive calcium entry. 3-Methyladenine (3-MA), an inhibitor of autophagy, promoted the effects of ST2-104 in inhibiting apoptosis triggered by glutamate while rapamycin, an activator of autophagy, failed to do so. ST2-104 peptide reversed glutamate-induced apoptosis via inhibiting Ca2+/CaM-dependent protein kinase kinase β (CaMKKβ)-mediated autophagy, which was partly enhanced by STO-609 (an inhibitor of CaMKKβ). ST2-104 attenuated neuronal apoptosis by inhibiting autophagy through CaMKKβ/AMPK/mTOR pathway. Our results suggest that the neuroprotective effect of ST2-104 are due to actions on the crosstalk between apoptosis and autophagy via the CaMKKβ/AMPK/mTOR signaling pathway. The findings present novel insights into the potential neuroprotection of ST2-104 in cerebral ischemia.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021 People’s Republic of China
| | - Yingshi Ji
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021 People’s Republic of China
| | - Jinghong Ren
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021 People’s Republic of China
| | - Huanyu Liu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021 People’s Republic of China
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 North Campbell Drive, P.O. Box 245050, Tucson, AZ 85724 USA
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital, Jilin University, Changchun, Jilin 130021 People’s Republic of China
| |
Collapse
|
15
|
Emam AM, Saad MA, Ahmed NA, Zaki HF. Vortioxetine mitigates neuronal damage by restricting PERK/eIF2α/ATF4/CHOP signaling pathway in rats subjected to focal cerebral ischemia-reperfusion. Life Sci 2021; 283:119865. [PMID: 34358549 DOI: 10.1016/j.lfs.2021.119865] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
AIMS Stroke has risen to the fifth and third most common causes of death in the United States and the rest of the world, respectively. Vortioxetine (VTX) is a multimodal antidepressant agent that balances 5-HT receptors and represses the serotonin transporter. Our study aimed to examine the neuroprotective impacts of VTX against cerebral ischemia caused by occluding the middle cerebral artery (MCA). MAIN METHODS Until the middle cerebral artery occlusion (MCAO) induction, VTX (10 mg/kg/day) was taken orally for 14 days. Behavioral assessments were carried out 24 h after the MCAO technique. The hippocampal and cortical tissues of the brain were isolated to assess the histological changes and the levels of the biochemical parameters. KEY FINDINGS MCAO damage led to severe neurological deficits and histopathological damage. However, VTX improved MCAO-induced neurological deficits and ameliorated histopathological changes in both hippocampal and cortical tissues of MCAO rats. Western blot analysis showed increments of p-PERK, CHOP, ASK-1, NICD, HES-1, HES-5, and p-eIF2α expression levels in MCAO rats. Moreover, ELISA revealed an increase in the levels of ATF4, IRE1, Apaf-1, and HIF-1α, while VTX administration ameliorated most of these perturbations induced after MCAO injury. SIGNIFICANCE This research suggests that VTX could be a potent neuroprotective agent against ischemic stroke by inhibiting a variety of oxidative, apoptotic, inflammatory, and endoplasmic reticulum stress pathways.
Collapse
Affiliation(s)
- Amr M Emam
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt.
| | - Muhammad A Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt; School of Pharmacy, New Giza University, Giza, Egypt
| | - Naglaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6th of October City, Giza, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| |
Collapse
|
16
|
Li C, He K, Yin M, Zhang Q, Lin J, Niu Y, Wang Q, Xu Q, Jiang N, Zhao G. LOX-1 Regulates Neutrophil Apoptosis and Fungal Load in A. Fumigatus Keratitis. Curr Eye Res 2021; 46:1800-1811. [PMID: 34264144 DOI: 10.1080/02713683.2021.1948063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE To determine whether LOX-1 regulates neutrophil apoptosis and fungal load in A. fumigatus keratitis. METHODS Fas, FasL, CASP3, CASP8, CASP9 and BCL2 were tested in normal and infected corneas of C57BL/6 mice. Mice corneas were infected with A. fumigatus with or without pretreatment of LOX-1 neutralizing antibody or inhibitor (Poly I). Clinical score was recored and HE staining was tested. Fungal load in mice corneas was observed by plate counting. Poly morphonuclear neutrophilic leukocytes (PMNs) were stimulated with 75% ethanol-killed A. fumigatus with or without pretreatment of LOX-1 neutralizing antibody or Poly I. PCR, western blot and immunostaining tested expression of Fas, FasL, CASP3, CASP8, CASP9, BCL2 and cleaved caspase-3. PMNs infiltration and TUNEL-positive cells were assessed by immunofluorescent staining. Flow cytometry assay tested the percentage of apoptosis neutrophils. RESULTS Fas, Fas ligand, caspase-8, caspase-9 and caspase-3 mRNA levels were significantly higher in C57BL/6 mice corneas infected with A. fumigatus than normal corneas. Poly I treatment alleviated the severity and decreased clinical score at 3, 5 and 7 days post infecrion (p.i.). HE staining showed less infiltration in corneal tissue after LOX-1 inhibition. Plate counting experiment showed that number of viable fungus in corneas of Poly I treated group was significantly less than control group. LOX-1 neutralizing antibody or Poly I treatment significantly decreased neutrophil infiltration, the quantity of TUNEL-positive cells, the expression of Fas, Fas ligand, caspase-8, caspase-9, caspase-3, cleaved caspase-3 and the percentage of apoptosis neutrophils compared with control corneas. LOX-1 neutralizing antibody treatment significantly decreased Fas, FasL, CASP3, CASP8, CASP9 and cleaved caspase-3 expression in neutrophils. CONCLUSION LOX-1 inhibition decrease neutrophil apoptosis and fungal load in A. fumigatus keratitis.
Collapse
Affiliation(s)
- Cui Li
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Kun He
- Department of Ophthalmology, Zhejiang Quhua Hospital Quzhou, Zhejiang, China
| | - Min Yin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qiuqiu Zhang
- Department of Ophthalmology, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China
| | - Jing Lin
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yawen Niu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qian Wang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qiang Xu
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Nan Jiang
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Guiqiu Zhao
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
17
|
Girnar GA, Mahajan HS. Cerebral ischemic stroke and different approaches for treatment of stroke. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00289-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Cerebral ischemia can be considered a lethal disease as it is a leading cause of death worldwide with no prompt line of treatment. The factors which make this disease more fatal are failure of drugs while crossing BBB, very low availability of the drug in the brain, inefficiency of drug molecule in the clinical studies, limited availability of clinical data, lack of awareness about this disease, and many more.
Main body
This review focuses on reasons and mechanisms of stroke, classification of brain ischemia; it also reveals the current scenario of stroke in India. Very few drugs are effective for the treatment of stroke. This compilation furnishes conventional and recent treatments of stroke along with their hurdles like the gap between preclinical and clinical studies. This review also suggests effective routes of administration of drugs for the treatment of brain ischemia specifically nose-to-brain route and effectiveness of different dosage forms precisely nanoformulations, as the most effective dosage form.
Conclusion
By following different guidelines and treatments, the risk of brain ischemia can be minimized as well as some advanced techniques for the treatment of this disease proving their efficiency. One of the important aspects in the success of the treatment for this disease is the route of administration of the drug. Among all routes, intranasal drug delivery presents a potential approach and is supposed to be the next-generation therapy for brain disorders. The nose-to-brain route is very effective, and it shows some promising results in case of stroke treatment. The strategy is still under investigation despite various successful lab-scale studies; there are numerous challenges to reach the product in the market. Research is going on to get a better understanding of this strategy. We believe that detailed studies to resolve pitfalls will lead to the successful development of an intranasal formulation for the management of ischemic brain injury such as stroke.
Collapse
|
18
|
Liu W, Qaed E, Zhu HG, Dong MX, Tang Z. Non-energy mechanism of phosphocreatine on the protection of cell survival. Biomed Pharmacother 2021; 141:111839. [PMID: 34174505 DOI: 10.1016/j.biopha.2021.111839] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
If mitochondrial energy availability or oxidative metabolism is altered, patients will suffer from insufficient energy supply Phosphocreatine (PCr) not only acts as an energy carrier, but also acts as an antioxidant and defensive agent to maintain the integrity and stability of the membrane, to maintain ATP homeostasis through regulating mitochondrial respiration. Meanwhile, PCr can enhance calcium balance and reduce morphological pathological changes, ultimately, PCr helps to reduce apoptosis. On the other aspect, the activities of ATP synthase and MitCK play a crucial role in the maintenance of cellular energy metabolic function. It is interesting to note, PCr not only rises the activities of ATP synthase as well as MitCK, but also promotes these two enzymatic reactions. Additionally, PCr can also inhibit mitochondrial permeability transition in a concentration-dependent manner, prevent ROS and CytC from spilling into the cytoplasm, thereby inhibit the release of proapoptotic factors caspase-3 and caspase-9, and eventually, effectively prevent LPS-induced apoptosis of cells. Understandably, PCr prevents the apoptosis caused by abnormal mitochondrial energy metabolism and has a protective role in a non-energy manner. Moreover, recent studies have shown that PCr protects cell survival through PI3K/Akt/eNOS, MAPK pathway, and inhibition of Ang II-induced NF-κB activation. Furthermore, PCr antagonizes oxidative stress through the activation of PI3K/Akt/GSK3b intracellular pathway, PI3K/AKT-PGC1α signaling pathway, while through the promotion of SIRT3 expression to maintain normal cell metabolism. Interestingly, PCr results in delaying the time to enter pathological metabolism through the delayed activation of AMPK pathway, which is different from previous studies, now we propose the hypothesis that the "miRNA-JAK2/STAT3 -CypD pathway" may take part in protecting cells from apoptosis, PCr may be further be involved in the dynamic relationship between CypD and STAT3. Furthermore, we believe that PCr and CypD would be the central link to maintain cell survival and maintain cell stability and mitochondrial repair under the mitochondrial dysfunction caused by oxidative stress. This review provides the modern progress knowledge and views on the molecular mechanism and molecular targets of PCr in a non-energy way.
Collapse
Affiliation(s)
- Wu Liu
- Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | - Eskandar Qaed
- Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | - Han Guo Zhu
- Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | - Ma Xiao Dong
- Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China
| | - ZeYao Tang
- Department of Pharmacology, Dalian Medical University, 9 West Section, South Road of Lushun, 116044 Dalian, China.
| |
Collapse
|
19
|
Khan MM, Badruddeen, Mujahid M, Akhtar J, Khan MI, Ahmad U. An Overview of Stroke: Mechanism, In vivo Experimental Models Thereof, and Neuroprotective Agents. Curr Protein Pept Sci 2021; 21:860-877. [PMID: 32552641 DOI: 10.2174/1389203721666200617133903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/22/2019] [Accepted: 07/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Stroke is one of the causes of death and disability globally. Brain attack is because of the acute presentation of stroke, which highlights the requirement for decisive action to treat it. OBJECTIVE The mechanism and in-vivo experimental models of stroke with various neuroprotective agents are highlighted in this review. METHOD The damaging mechanisms may proceed by rapid, nonspecific cell lysis (necrosis) or by the active form of cell death (apoptosis or necroptosis), depending upon the duration and severity and of the ischemic insult. RESULTS Identification of injury mediators and pathways in a variety of experimental animal models of global cerebral ischemia has directed to explore the target-specific cytoprotective strategies, which are critical to clinical brain injury outcomes. CONCLUSION The injury mechanism, available encouraging medicaments thereof, and outcomes of natural and modern medicines for ischemia have been summarized. In spite of available therapeutic agents (thrombolytics, calcium channel blockers, NMDA receptor antagonists and antioxidants), there is a need for an ideal drug for strokes.
Collapse
Affiliation(s)
- Mohd Muazzam Khan
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Badruddeen
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | - Mohd Mujahid
- Department of Pharmacology, College of Pharmacy, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| | | | - Usama Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
20
|
Participation of Amyloid and Tau Protein in Post-Ischemic Neurodegeneration of the Hippocampus of a Nature Identical to Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22052460. [PMID: 33671097 PMCID: PMC7957532 DOI: 10.3390/ijms22052460] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/05/2023] Open
Abstract
Recent evidence suggests that amyloid and tau protein are of vital importance in post-ischemic death of CA1 pyramidal neurons of the hippocampus. In this review, we summarize protein alterations associated with Alzheimer's disease and their gene expression (amyloid protein precursor and tau protein) after cerebral ischemia, as well as their roles in post-ischemic hippocampus neurodegeneration. In recent years, multiple studies aimed to elucidate the post-ischemic processes in the development of hippocampus neurodegeneration. Their findings have revealed the dysregulation of genes for amyloid protein precursor, β-secretase, presenilin 1 and 2, tau protein, autophagy, mitophagy, and apoptosis identical in nature to Alzheimer's disease. Herein, we present the latest data showing that amyloid and tau protein associated with Alzheimer's disease and their genes play a key role in post-ischemic neurodegeneration of the hippocampus with subsequent development of dementia. Therefore, understanding the underlying process for the development of post-ischemic CA1 area neurodegeneration in the hippocampus in conjunction with Alzheimer's disease-related proteins and genes will provide the most important therapeutic development goals to date.
Collapse
|
21
|
Zhang LN, Zhang XW, Li CQ, Guo J, Chen YP, Chen SL. Vagal Nerve Stimulation Protects Against Cerebral Ischemia-Reperfusion Injury in Rats by Inhibiting Autophagy and Apoptosis. Neuropsychiatr Dis Treat 2021; 17:905-913. [PMID: 33790559 PMCID: PMC8008252 DOI: 10.2147/ndt.s300535] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cumulative evidence suggests that neuronal death including autophagy, apoptosis, and necrosis is closely related to the occurrence and development of cerebral ischemia-reperfusion (I/R) injury. Moreover, vagal nerve stimulation (VNS) is involved in many different neuroprotective and neuroplasticity pathways. Thus, VNS may be a novel approach for treating various neurodegenerative diseases. The present study aims to determine whether VNS protects against cerebral I/R injury in rats by inhibiting autophagy and apoptosis. METHODS Cerebral I/R injury is induced by middle cerebral artery occlusion (MCAO) and VNS is carried out. Infarct volume, neurological deficit, autophagy, and apoptosis are examined 24 h after reperfusion. RESULTS Vagal nerve stimulation decreases infarct volume and suppresses neurological deficit. Moreover, obvious autophagy and apoptosis are detected in rats that have undergone I/R, and VNS inhibits autophagy and apoptosis. CONCLUSION Vagal nerve stimulation exerts neuroprotective effects following I/R injury by inhibiting autophagy and apoptosis.
Collapse
Affiliation(s)
- Li-Na Zhang
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, 404000, People's Republic of China
| | - Xian-Wei Zhang
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, 404000, People's Republic of China
| | - Chang-Qing Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jing Guo
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, 404000, People's Republic of China
| | - Yong-Ping Chen
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, 404000, People's Republic of China
| | - Sheng-Li Chen
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, 404000, People's Republic of China
| |
Collapse
|
22
|
Balion Z, Ramanauskienė K, Jekabsone A, Majienė D. The Role of Mitochondria in Brain Cell Protection from Ischaemia by Differently Prepared Propolis Extracts. Antioxidants (Basel) 2020; 9:antiox9121262. [PMID: 33322707 PMCID: PMC7763930 DOI: 10.3390/antiox9121262] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/06/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are both the primary targets and mediators of ischaemic damage in brain cells. Insufficient oxygen causes reactive oxygen species that damage the mitochondria, leading to the loss of functionality and viability of highly energy-demanding neurons. We have recently found that aqueous (AqEP), polyethylene glycol-aqueous (Pg-AqEP) and ethanolic propolis extracts (EEP) can modulate mitochondria and ROS production in C6 cells of astrocytic origin. The aim of this study was to investigate the effect of the extracts on viability, mitochondrial efficiency and superoxide generation, and inflammatory cytokine release in primary rat cerebellar neuronal-glial cell cultures affected by ischaemia (mimicked by hypoxia +/- deoxyglucose). AqEP and Pg-AqEP (15-60 µg/mL of phenolic compounds, or PC) significantly increased neuronal viability in ischaemia-treated cultures, and this was accompanied by a reduction in mitochondrial superoxide levels. Less extended protection against ischaemia-induced superoxide production and death was exhibited by 2 to 4 µg/mL of PC EEP. Both Pg-AqEP and Ag-EP (but not EEP) significantly protected the cultures from hypoxia-induced elevation of TNF-α, IL-1β and IL-6. Only Pg-AqEP (but not AqEP or EEP) prevented hypoxia-induced loss of the mitochondrial basal and ATP-coupled respiration rate, and significantly increased the mitochondrial respiratory capacity. Summarising, the study revealed that hydrophilic propolis extracts might protect brain cells against ischaemic injury by decreasing the level of mitochondrial superoxide and preventing inflammatory cytokines, and, in the case of Pg-AqEP, by protecting mitochondrial function.
Collapse
Affiliation(s)
- Zbigniev Balion
- Laboratory of Pharmaceutical Sciences, Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Sukilėlių ave. 13, LT 50162 Kaunas, Lithuania; (Z.B.); (A.J.)
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Eivenių str. 4, LT-50161 Kaunas, Lithuania
| | - Kristina Ramanauskienė
- Department of Clinical Pharmacy, Lithuanian University of Health Sciences, Sukilėlių ave. 13, LT 50162 Kaunas, Lithuania;
| | - Aistė Jekabsone
- Laboratory of Pharmaceutical Sciences, Institute of Pharmaceutical Technologies, Lithuanian University of Health Sciences, Sukilėlių ave. 13, LT 50162 Kaunas, Lithuania; (Z.B.); (A.J.)
- Laboratory of Preclinical Drug Investigation, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių ave. 13, LT-50162 Kaunas, Lithuania
| | - Daiva Majienė
- Laboratory of Biochemistry, Neuroscience Institute, Lithuanian University of Health Sciences, Eivenių str. 4, LT-50161 Kaunas, Lithuania
- Department of Drug Technology and Social Pharmacy, Lithuanian University of Health Sciences, Sukilėlių ave. 13, LT 50162 Kaunas, Lithuania
- Correspondence: ; Tel.: +370-615-23993
| |
Collapse
|
23
|
Soliman GA, Abdel-Rahman RF, Ogaly HA, Althurwi HN, Abd-Elsalam RM, Albaqami FF, Abdel-Kader MS. Momordica charantia Extract Protects against Diabetes-Related Spermatogenic Dysfunction in Male Rats: Molecular and Biochemical Study. Molecules 2020; 25:molecules25225255. [PMID: 33187275 PMCID: PMC7698202 DOI: 10.3390/molecules25225255] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
More than 90% of diabetic patients suffer from sexual dysfunction, including diminished sperm count, sperm motility, and sperm viability, and low testosterone levels. The effects of Momordica charantia (MC) were studied by estimating the blood levels of insulin, glucose, glycosylated hemoglobin (HbA1c), testosterone (TST), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) in diabetic rats treated with 250 and 500 mg/kg b.w. of the total extract. Testicular antioxidants, epididymal sperm characteristics, testicular histopathology, and lesion scoring were also investigated. Testicular mRNA expression of apoptosis-related markers such as antiapoptotic B-cell lymphoma-2 (Bcl-2) and proapoptotic Bcl-2-associated X protein (Bax) were evaluated by real-time PCR. Furthermore, caspase-3 protein expression was evaluated by immunohistochemistry. MC administration resulted in a significant reduction in blood glucose and HbA1c and marked elevation of serum levels of insulin, TST, and gonadotropins in diabetic rats. It induced a significant recovery of testicular antioxidant enzymes, improved histopathological changes of the testes, and decreased spermatogenic and Sertoli cell apoptosis. MC effectively inhibited testicular apoptosis, as evidenced by upregulation of Bcl-2 and downregulation of Bax and caspase-3. Moreover, reduction in apoptotic potential in MC-treated groups was confirmed by reduction in the Bax/Bcl-2 mRNA expression ratio.
Collapse
Affiliation(s)
- Gamal A. Soliman
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (G.A.S.); (H.N.A.); (F.F.A.)
- Department of Pharmacology, College of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | | | - Hanan A. Ogaly
- Department of Chemistry, College of Science, King Khalid University, Abha 61421, Saudi Arabia;
- Department of Biochemistry, College of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Hassan N. Althurwi
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (G.A.S.); (H.N.A.); (F.F.A.)
| | - Reham M. Abd-Elsalam
- Department of Pathology, College of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Faisal F. Albaqami
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (G.A.S.); (H.N.A.); (F.F.A.)
| | - Maged S. Abdel-Kader
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacognosy, College of Pharmacy, Alexandria University, Alexandria 21215, Egypt
- Correspondence: ; Tel.: +966-545539145
| |
Collapse
|
24
|
Neuroprotective Effects of Anti-proBDNF in a Rat Photothrombotic Ischemic Model. Neuroscience 2020; 446:261-270. [PMID: 32798590 DOI: 10.1016/j.neuroscience.2020.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 11/23/2022]
Abstract
Up-regulation of proBDNF in ischemic brain and the detrimental role of proBDNF on cellular survival has already been established. We propose that the up-regulated proBDNF may trigger the harmful events and evoke a secondary ischemic damage after ischemia. This study aimed to establish the neuroprotective effects of anti-proBDNF antibody in a rat photothrombotic ischemic model. Photothrombotic ischemic model was performed on Sprague Dawley rats and anti-proBDNF antibodies were administered intraperitoneally to the ischemic rats at a dose of 5 mg/kg after 6 hours (6 h) and on 3 days (3d) after ischemia. Behavioural tests were performed for sensorimotor functional analyses. Animals were euthanized at 7d for histochemical and biochemical studies. We observed higher proBDNF expression around the ischemic infarct. Higher level of apoptosis and inflammation was evident at 7d after ischemia on brain sections. Interestingly, the anti-proBDNF treatment instigated significant reduction of the infarction size as detected by Haematoxylin and Eosin (H&E) staining. Similar reduction of apoptotic signaling proteins in western blot and immunostaining after anti-proBDNF treatment was found. Up-regulation of synaptic protein expression was also observed after this treatment. Significant sensorimotor functional improvements were also noticed at 7d after anti-proBDNF treatment. We conclude that anti-proBDNF treatment is anti-apoptotic and anti-inflammatory, and plays advantageous role in promoting cellular growth and improving sensorimotor function after ischemic insult. Taken together, our study suggests that this anti-proBDNF treatment can be considered as a therapeutic approach for ischemic recovery.
Collapse
|
25
|
Wang J, Li P, Qin T, Sun D, Zhao X, Zhang B. Protective effect of epigallocatechin-3-gallate against neuroinflammation and anxiety-like behavior in a rat model of myocardial infarction. Brain Behav 2020; 10:e01633. [PMID: 32304289 PMCID: PMC7303397 DOI: 10.1002/brb3.1633] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/20/2020] [Accepted: 03/21/2020] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Individuals who experience myocardial infarction (MI) often experience anxiety. Green tea has potent antioxidative properties and, epigallocatechin-3-gallate (EGCG), which is a primary component of tea polyphenols, has advantageous effects on anxiety and depression. However, its mechanism of action regarding the inhibition of anxiety-like symptoms after MI remains unclear. This study examined whether EGCG alleviated anxiety-like behavior in MI rats and its possible mechanism. MATERIAL AND METHODS Rats were administered a daily gavage of EGCG (50 mg/kg) 7 days before and 14 consecutive days after the MI procedure. The open-field test and light/dark shuttle box were performed to evaluate anxiety-like behavior. Serum and hippocampus interleukin (IL)-6 levels were tested using ELISA. Caspase 3, caspase 8, caspase 9 and bcl-2 messenger RNA levels in the hippocampus were determined using quantitative polymerase chain reaction, and STAT3 protein was detected by Western blot. RESULTS Results of the open field test and light/dark shuttle box task demonstrated that the MI procedure induced anxiety-like behavior in the animals, and this impairment was improved by EGCG. Daily EGCG administration significantly decreased the level of IL-6 both in serum and hippocampus after MI. The administration of EGCG also significantly moderated the expression of caspases 3, 8, and 9 mRNA, which was related to apoptosis in the hippocampus. Furthermore, EGCG also downregulated the expression of STAT3, which was related to the activity of IL-6. These results suggest that EGCG alleviated anxiety-like behavior by inhibiting increases in neuroinflammation and apoptosis in the rat hippocampus. In addition, EGCG reversed alterations of IL-6 and STAT3 in the brain to alleviate apoptosis in the hippocampus. CONCLUSIONS Thus, EGCG reversed anxiety-like behavior through an anti-inflammation effect to alleviate apoptosis in neurons and may be a useful therapeutic material for anxiety-like behavior after MI.
Collapse
Affiliation(s)
- Jinpeng Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Ping Li
- Department of Paediatrics, The Second Hospital of Jilin University, Changchun, China
| | - Tian Qin
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dongjie Sun
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xin Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Beilin Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
26
|
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
|
27
|
Cell Death Pathways in Ischemic Stroke and Targeted Pharmacotherapy. Transl Stroke Res 2020; 11:1185-1202. [PMID: 32219729 DOI: 10.1007/s12975-020-00806-z] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is one of the significant causes of morbidity and mortality, affecting millions of people across the globe. Cell injury in the infarct region is an inevitable consequence of focal cerebral ischemia. Subsequent reperfusion exacerbates the harmful effect and increases the infarct volume. These cellular injuries follow either a regulated pathway involving tightly structured signaling cascades and molecularly defined effector mechanisms or a non-regulated pathway, also known as accidental cell death, where the process is biologically uncontrolled. Classical cell death pathways are long established and well reported in several articles that majorly define apoptotic cell death. A recent focus on cell death study also considers investigation on non-classical pathways that are tightly regulated, may or may not involve caspases, but non-apoptotic. Pathological cell death is a cardinal feature of different neurodegenerative diseases. Although ischemia cannot be classified as a neurodegenerative disease, it is a cerebrovascular event where the infarct region exhibits aberrant cell death. Over the past few decades, several therapeutic options have been implicated for ischemic stroke. However, their use has been hampered owing to the number of limitations that they possess. Ischemic penumbral neurons undergo apoptosis and become dysfunctional; however, they are salvageable. Thus, understanding the role of different cell death pathways is crucial to aid in the modern treatment of protecting apoptotic neurons.
Collapse
|
28
|
Rawlinson C, Jenkins S, Thei L, Dallas ML, Chen R. Post-Ischaemic Immunological Response in the Brain: Targeting Microglia in Ischaemic Stroke Therapy. Brain Sci 2020; 10:brainsci10030159. [PMID: 32168831 PMCID: PMC7139954 DOI: 10.3390/brainsci10030159] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/27/2020] [Accepted: 03/07/2020] [Indexed: 12/21/2022] Open
Abstract
Microglia, the major endogenous immune cells of the central nervous system, mediate critical degenerative and regenerative responses in ischaemic stroke. Microglia become "activated", proliferating, and undergoing changes in morphology, gene and protein expression over days and weeks post-ischaemia, with deleterious and beneficial effects. Pro-inflammatory microglia (commonly referred to as M1) exacerbate secondary neuronal injury through the release of reactive oxygen species, cytokines and proteases. In contrast, microglia may facilitate neuronal recovery via tissue and vascular remodelling, through the secretion of anti-inflammatory cytokines and growth factors (a profile often termed M2). This M1/M2 nomenclature does not fully account for the microglial heterogeneity in the ischaemic brain, with some simultaneous expression of both M1 and M2 markers at the single-cell level. Understanding and regulating microglial activation status, reducing detrimental and promoting repair behaviours, present the potential for therapeutic intervention, and open a longer window of opportunity than offered by acute neuroprotective strategies. Pharmacological modulation of microglial activation status to promote anti-inflammatory gene expression can increase neurogenesis and improve functional recovery post-stroke, based on promising preclinical data. Cell-based therapies, using preconditioned microglia, are of interest as a method of therapeutic modulation of the post-ischaemic inflammatory response. Currently, there are no clinically-approved pharmacological options targeting post-ischaemic inflammation. A major developmental challenge for clinical translation will be the selective suppression of the deleterious effects of microglial activity after stroke whilst retaining (or enhancing) the neurovascular repair and remodelling responses of microglia.
Collapse
Affiliation(s)
- Charlotte Rawlinson
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
| | - Stuart Jenkins
- School of Medicine, Keele University, Staffordshire ST5 5BG, UK;
| | - Laura Thei
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK; (L.T.); (M.L.D.)
| | - Mark L. Dallas
- School of Pharmacy, University of Reading, Reading RG6 6UB, UK; (L.T.); (M.L.D.)
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK;
- Correspondence: ; Tel.: +44-1782-733849; Fax: 44-1782-733326
| |
Collapse
|
29
|
Affiliation(s)
- Antonino Tuttolomondo
- Department of Promoting Health, Maternal Infant, Excellence and Internal & Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Palermo, Sicily, Italy
- Internal Medicine and Stroke Care Ward, Policlinico ‘P Giaccone’, University of Palermo, Palermo, Sicily, Italy
| | - Antonio Pinto
- Department of Promoting Health, Maternal Infant, Excellence and Internal & Specialized Medicine (ProMISE) G. D'Alessandro, University of Palermo, Palermo, Sicily, Italy
- Internal Medicine and Stroke Care Ward, Policlinico ‘P Giaccone’, University of Palermo, Palermo, Sicily, Italy
| |
Collapse
|
30
|
Park CW, Ahn JH, Lee TK, Park YE, Kim B, Lee JC, Kim DW, Shin MC, Park Y, Cho JH, Ryoo S, Kim YM, Won MH, Park JH. Post-treatment with oxcarbazepine confers potent neuroprotection against transient global cerebral ischemic injury by activating Nrf2 defense pathway. Biomed Pharmacother 2020; 124:109850. [PMID: 31981945 DOI: 10.1016/j.biopha.2020.109850] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/29/2019] [Accepted: 01/12/2020] [Indexed: 01/27/2023] Open
Abstract
Oxcarbazepine (OXC), a voltage-gated sodium channel blocker, is an antiepileptic medication and used for the bipolar disorders treatment. Some voltage-gated sodium channel blockers have been demonstrated to display strong neuroprotective properties in models of cerebral ischemia. However, neuroprotective effects and mechanisms of OXC have not yet been reported. Here, we investigated the protective effect of OXC and its mechanisms in the cornu ammonis 1 subfield (CA1) of gerbils subjected to 5 min of transient global cerebral ischemia (tGCI). tGCI led to death of most pyramidal neurons in CA1 at 5 days after ischemia. OXC (100 and 200 mg/kg) was intraperitoneally administered once at 30 min after tGCI. Treatment with 200 mg/kg, not 100 mg/kg OXC, significantly protected CA1 pyramidal neurons from tGCI-induced injury. OXC treatment significantly decreased superoxide anion production, 4-hydroxy-2-nonenal and 8-hydroxyguanine levels in ischemic CA1 pyramidal neurons. In addition, the treatment restored levels of superoxide dismutases, catalase, and glutathione peroxidase. Furthermore, the treatment distinctly inhibited tGCI-induced microglia activation and significantly reduced levels of pro-inflammatory cytokines (interleukin-1β and tumor necrosis factor-α). In particular, OXC treatment significantly enhanced expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream protein heme oxygenase-1 in ischemic CA1. The neuroprotective effects of OXC were abolished by brusatol (an inhibitor of Nrf2). Taken together, these results indicate that post-treatment of OXC can display neuroprotection against brain injuries following ischemic insults. This neuroprotection may be displayed by attenuation of oxidative stress and neuroinflammation, which can be mediated by activation of Nrf2 pathway.
Collapse
Affiliation(s)
- Cheol Woo Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Young Eun Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, and Research Institute of Oral Sciences, College of Dentistry, Gangnung-Wonju National University, Gangneung, Gangwon 25457, Republic of Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Yoonsoo Park
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Sungwoo Ryoo
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Republic of Korea.
| |
Collapse
|
31
|
Virdi JK, Bhanot A, Jaggi AS, Agarwal N. Investigation on beneficial role of l-carnosine in neuroprotective mechanism of ischemic postconditioning in mice: possible role of histidine histamine pathway. Int J Neurosci 2020; 130:983-998. [PMID: 31951767 DOI: 10.1080/00207454.2020.1715393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVE The present study was undertaken to investigate the possible role of histidine-histamine pathway in the neuroprotective effects produced by L-carnosine hand in hand with ischemic postconditioning in the animal model of cerebral ischemia. METHODS Cerebral ischemia was induced in swiss albino mice by performing BCCAO surgery. Morris water-maze test was utilized to assess the learning ability and memory of the animals. The whole brain acetylcholinesterase (AChE) activity, TBARS, GSH levels and MPO activity were evaluated as the biochemical parameters. For histopathological evaluation of the cerebral infarct size, TTC staining was employed. RESULTS Administration of L-carnosine (500 mg/kg, i.p.) successfully attenuated the manifestations of cerebral ischemia. Higher levels of AChE, TBARS, and MPO were observed in BCCAO treated animals, which were successfully attenuated by treatment with L-carnosine and ischemic postconditioning. Whereas administration of L-carnosine and ischemic postconditioning significantly increased the level of GSH in BCCAO treated animals. Moreover, treatment with ranitidine, an H2 blocker (30 NMol, i.c.v) antagonized the neuroprotective actions of L-carnosine evidenced by decrease in MWM performance, increase in the level of AChE and oxidative stress, while decrease in GSH level in brain. The cerebral infarct size was found to be more in BCCAO inflicted animals, which was improved by the administration of L-carnosine, while the cerebral infarct size worsened by treatment with ranitidine (3 nmol, i.c.v.). CONCLUSION It is concluded that L-carnosine exerts neuroprotective effect via involvement of histidine-histamine pathway since the beneficial effects of L-carnosine were abolished by the H2-blocker.
Collapse
Affiliation(s)
- Jasleen Kaur Virdi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Amritansh Bhanot
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Neha Agarwal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| |
Collapse
|
32
|
Prieto-Moure B, Cejalvo-Lapeña D, Belda-Antolí M, Padrón-Sanz C, Lloris-Cejalvo JM, Lloris-Carsí JM. Combination Therapy of Allopurinol and Dantrolene and Its Role In The Prevention of Experimental Ischemia Reperfusion Injury Of The Small Intestine. J INVEST SURG 2020; 34:800-807. [PMID: 31906750 DOI: 10.1080/08941939.2019.1696904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The effect of different drugs on ischemia and reperfusion (I/R; induced oxygen free radical damage) was examined in small bowel tissue because the intestine is extremely sensitive to this pathology. Different drugs (allopurinol and dantrolene) can remove oxygen free radicals or inhibit the mechanisms leading to their generation, thus reducing mucosal lesions. We investigated the protective potential of combination therapy in the intestine against I/R damage. METHODS Forty-eight male Wistar rats were separated into 8 groups: one sham (control), one I/R (ischemia 60 min + reperfusion at 24 h), and 6 groups treated with allopurinol, dantrolene, or combination therapy. The grade of injury in the small bowel was established by the lipid peroxidation (MDA) and antioxidant enzymatic activity of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) in tissue samples. Moreover, the collected samples were subjected to histological study. RESULTS Combination therapy preserved normal enzymatic levels compared to the I/R groups (p < 0.05) for all parameters studied. The animals treated with combination therapy showed less severe small bowel damage than I/R group in accordance with the histological results. CONCLUSIONS Results obtained in the experimental process indicate that the administration of antioxidants protects against intestinal damage by I/R. Overall, combination therapy may protect intestinal tissue from I/R injury.
Collapse
Affiliation(s)
- Beatriz Prieto-Moure
- Department of Biomedical Sciences, European University of Valencia, Valencia, Spain
| | - Dolores Cejalvo-Lapeña
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | - Mariola Belda-Antolí
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | - Carolina Padrón-Sanz
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | - José Miguel Lloris-Cejalvo
- Research Group "Identification Therapeutic and Experimental Evaluation of Natural Products Bioprotectors," Faculty of Medicine and Experimental, Catholic University of Valencia, Valencia, Spain
| | | |
Collapse
|
33
|
Richard SA. Elucidating the novel biomarker and therapeutic potentials of High-mobility group box 1 in Subarachnoid hemorrhage: A review. AIMS Neurosci 2019; 6:316-332. [PMID: 32341986 PMCID: PMC7179354 DOI: 10.3934/neuroscience.2019.4.316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/21/2019] [Indexed: 12/13/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) frequently arises after an aneurysm in a cerebral artery ruptures, resulting into bleeding as well as clot formation. High-mobility group box 1 (HMGB1) is an extremely preserved, universal protein secreted in the nuclei of all cell varieties. This review explores the biomarker as well as therapeutic potentials of HMBG1 in SAH especially during the occurrence of cerebral vasospasms. Plasma HMGB1 levels have proven to be very useful prognosticators of effective outcome as well as death after SAH. Correspondingly, higher HMGB1 levels in the cerebrospinal fluid (CSF) of SAH patients correlated well with poor outcome; signifying that, CSF level of HMGB1 is a novel predictor of outcome following SAH. Nonetheless, the degree of angiographic vasospasm does not always correlate with the degree of neurological deficits in SAH patients. HMGB1 stimulated cerebral vasospasm, augmented gene as well as protein secretory levels of receptor for advance glycation end product (RAGE) in neurons following SAH; which means that, silencing HMGB1 during SAH could be of therapeutic value. Compounds like resveratrol, glycyrrhizin, rhinacanthin, purpurogallin, 4′-O-β-D-Glucosyl-5-O-Methylvisamminol (4OGOMV) as well as receptor-interacting serine/threonine-protein kinase 3 (RIPK3) gene are capable of interacting with HMGB1 resulting in therapeutic benefits following SAH.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Medicine, Princefield University, P. O. Box MA 128, Ho-Volta Region, Ghana West Africa
| |
Collapse
|
34
|
Costea L, Mészáros Á, Bauer H, Bauer HC, Traweger A, Wilhelm I, Farkas AE, Krizbai IA. The Blood-Brain Barrier and Its Intercellular Junctions in Age-Related Brain Disorders. Int J Mol Sci 2019; 20:ijms20215472. [PMID: 31684130 PMCID: PMC6862160 DOI: 10.3390/ijms20215472] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
With age, our cognitive skills and abilities decline. Maybe starting as an annoyance, this decline can become a major impediment to normal daily life. Recent research shows that the neurodegenerative disorders responsible for age associated cognitive dysfunction are mechanistically linked to the state of the microvasculature in the brain. When the microvasculature does not function properly, ischemia, hypoxia, oxidative stress and related pathologic processes ensue, further damaging vascular and neural function. One of the most important and specialized functions of the brain microvasculature is the blood-brain barrier (BBB), which controls the movement of molecules between blood circulation and the brain parenchyma. In this review, we are focusing on tight junctions (TJs), the multiprotein complexes that play an important role in establishing and maintaining barrier function. After a short introduction of the cell types that modulate barrier function via intercellular communication, we examine how age, age related pathologies and the aging of the immune system affects TJs. Then, we review how the TJs are affected in age associated neurodegenerative disorders: Alzheimer's disease and Parkinson's disease. Lastly, we summarize the TJ aspects of Huntington's disease and schizophrenia. Barrier dysfunction appears to be a common denominator in neurological disorders, warranting detailed research into the molecular mechanisms behind it. Learning the commonalities and differences in the pathomechanism of the BBB injury in different neurological disorders will predictably lead to development of new therapeutics that improve our life as we age.
Collapse
Affiliation(s)
- Laura Costea
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania.
| | - Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, 6726 Szeged, Hungary.
| | - Hannelore Bauer
- Department of Biological Sciences, University of Salzburg, 5020 Salzburg, Austria.
| | - Hans-Christian Bauer
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University-Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria.
| | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University-Spinal Cord Injury and Tissue Regeneration Center Salzburg, 5020 Salzburg, Austria.
| | - Imola Wilhelm
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania.
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary.
| | - Attila E Farkas
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary.
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, 6726 Szeged, Hungary.
| | - István A Krizbai
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania.
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary.
| |
Collapse
|
35
|
Zaric M, Drakulic D, Dragic M, Gusevac Stojanovic I, Mitrovic N, Grkovic I, Martinovic J. Molecular Alterations and Effects of Acute Dehydroepiandrosterone Treatment Following Brief Bilateral Common Carotid Artery Occlusion: Relevance to Transient Ischemic Attack. Neuroscience 2019; 410:128-139. [PMID: 31095985 DOI: 10.1016/j.neuroscience.2019.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/01/2019] [Accepted: 05/05/2019] [Indexed: 10/26/2022]
Abstract
Transient ischemic attack (TIA) represents brief neurological dysfunction of vascular origin without detectable infarction. Despite major clinical relevance characterization of post-TIA molecular changes using appropriate experimental model is lacking and no therapeutic agent has been established yet. Neurosteroid dehydroepiandrosterone (DHEA) arose as one of the candidates for cerebral ischemia treatment but its effects on TIA-like condition remain unknown. Seeking an animal model applicable for investigation of molecular alterations in mild ischemic conditions such as TIA, 15-min bilateral common carotid artery occlusion with 24-h reperfusion was performed to induce ischemia/ reperfusion (I/R) injury in adult male Wistar rats. Additionally, effects of 4-h post-operative DHEA treatment (20 mg/kg) were investigated in physiological and I/R conditions in hippocampus (HIP) and prefrontal cortex (PFC). The study revealed absence of sensorimotor deficits, cerebral infarcts and neurodegeneration along with preserved HIP and PFC overall neuronal morphology and unaltered malondialdehyde and reduced glutathione level following I/R and/or DHEA treatment. I/R induced nitric oxide burst in HIP and PFC was accompanied with increased neuronal nitric oxide synthase protein level exclusively in HIP. DHEA had no effects in physiological conditions, while increase of Bax/Bcl2 ratio and dissipation of mitochondrial membrane potential in treated I/R group suggested DHEA-mediated exacerbation of post-ischemic changes that might lead to pro-apoptotic events in HIP. Interestingly, DHEA restored I/R-induced NO to the control level in PFC. Obtained results indicated that I/R may serve as an appropriate model for investigation of molecular changes and treatment outcome following mild ischemic conditions such as TIA.
Collapse
Affiliation(s)
- Marina Zaric
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia.
| | - Dunja Drakulic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Milorad Dragic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia; Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Studentski trg 3, 11001 Belgrade, Republic of Serbia
| | - Ivana Gusevac Stojanovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Natasa Mitrovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Ivana Grkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| | - Jelena Martinovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11351 Belgrade, Republic of Serbia
| |
Collapse
|
36
|
Roffe C, Nevatte T, Bishop J, Sim J, Penaloza C, Jowett S, Ives N, Gray R, Ferdinand P, Muddegowda G. Routine low-dose continuous or nocturnal oxygen for people with acute stroke: three-arm Stroke Oxygen Supplementation RCT. Health Technol Assess 2019; 22:1-88. [PMID: 29595449 DOI: 10.3310/hta22140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Stroke is a major cause of death and disability worldwide. Hypoxia is common after stroke and is associated with worse outcomes. Oxygen supplementation could prevent hypoxia and secondary brain damage. OBJECTIVES (1) To assess whether or not routine low-dose oxygen supplementation in patients with acute stroke improves outcome compared with no oxygen; and (2) to assess whether or not oxygen given at night only, when oxygen saturation is most likely to be low, is more effective than continuous supplementation. DESIGN Multicentre, prospective, randomised, open, blinded-end point trial. SETTING Secondary care hospitals with acute stroke wards. PARTICIPANTS Adult stroke patients within 24 hours of hospital admission and 48 hours of stroke onset, without definite indications for or contraindications to oxygen or a life-threatening condition other than stroke. INTERVENTIONS Allocated by web-based minimised randomisation to: (1) continuous oxygen: oxygen via nasal cannula continuously (day and night) for 72 hours after randomisation at a flow rate of 3 l/minute if baseline oxygen saturation was ≤ 93% or 2 l/minute if > 93%; (2) nocturnal oxygen: oxygen via nasal cannula overnight (21:00-07:00) for three consecutive nights. The flow rate was the same as the continuous oxygen group; and (3) control: no routine oxygen supplementation unless required for reasons other than stroke. MAIN OUTCOME MEASURES Primary outcome: disability assessed by the modified Rankin Scale (mRS) at 3 months by postal questionnaire (participant aware, assessor blinded). Secondary outcomes at 7 days: neurological improvement, National Institutes of Health Stroke Scale (NIHSS), mortality, and the highest and lowest oxygen saturations within the first 72 hours. Secondary outcomes at 3, 6, and 12 months: mortality, independence, current living arrangements, Barthel Index, quality of life (European Quality of Life-5 Dimensions, three levels) and Nottingham Extended Activities of Daily Living scale by postal questionnaire. RESULTS In total, 8003 patients were recruited between 24 April 2008 and 17 June 2013 from 136 hospitals in the UK [continuous, n = 2668; nocturnal, n = 2667; control, n = 2668; mean age 72 years (standard deviation 13 years); 4398 (55%) males]. All prognostic factors and baseline characteristics were well matched across the groups. Eighty-two per cent had ischaemic strokes. At baseline the median Glasgow Coma Scale score was 15 (interquartile range 15-15) and the mean and median NIHSS scores were 7 and 5 (range 0-34), respectively. The mean oxygen saturation at randomisation was 96.6% in the continuous and nocturnal oxygen groups and 96.7% in the control group. Primary outcome: oxygen supplementation did not reduce disability in either the continuous or the nocturnal oxygen groups. The unadjusted odds ratio for a better outcome (lower mRS) was 0.97 [95% confidence interval (CI) 0.89 to 1.05; p = 0.5] for the combined oxygen groups (both continuous and nocturnal together) (n = 5152) versus the control (n = 2567) and 1.03 (95% CI 0.93 to 1.13; p = 0.6) for continuous versus nocturnal oxygen. Secondary outcomes: oxygen supplementation significantly increased oxygen saturation, but did not affect any of the other secondary outcomes. LIMITATIONS Severely hypoxic patients were not included. CONCLUSIONS Routine low-dose oxygen supplementation in stroke patients who are not severely hypoxic is safe, but does not improve outcome after stroke. FUTURE WORK To investigate the causes of hypoxia and develop methods of prevention. TRIAL REGISTRATION Current Controlled Trials ISRCTN52416964 and European Union Drug Regulating Authorities Clinical Trials (EudraCT) number 2006-003479-11. FUNDING DETAILS This project was funded by the National Institute for Health Research (NIHR) Research for Patient Benefit and Health Technology Assessment programmes and will be published in full in Health Technology Assessment; Vol. 22, No. 14. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Christine Roffe
- Institute for Applied Clinical Sciences, Keele University, Keele, UK
| | | | - Jon Bishop
- University of Birmingham, Birmingham, UK
| | | | | | - Susan Jowett
- Health Economics Unit, University of Birmingham, Birmingham, UK
| | | | | | | | - Girish Muddegowda
- Neurosciences Department, Royal Stoke University Hospital, Stoke-on-Trent, UK
| |
Collapse
|
37
|
Neurotherapeutic potential of kolaviron on neurotransmitter dysregulation, excitotoxicity, mitochondrial electron transport chain dysfunction and redox imbalance in 2-VO brain ischemia/reperfusion injury. Biomed Pharmacother 2019; 111:859-872. [DOI: 10.1016/j.biopha.2018.12.144] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/26/2018] [Accepted: 12/31/2018] [Indexed: 02/05/2023] Open
|
38
|
Zhang G, Zha J, Liu J, Di J. Minocycline impedes mitochondrial-dependent cell death and stabilizes expression of hypoxia inducible factor-1α in spinal cord injury. Arch Med Sci 2019; 15:475-483. [PMID: 30899301 PMCID: PMC6425201 DOI: 10.5114/aoms.2018.73520] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 01/01/2018] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION One of the crucial mechanisms following spinal cord injury is mitochondria-associated cell death. Minocycline, an anti-inflammatory drug, is well known to impede mitochondrial cell death. However, there has been no study on the effect of minocycline linking Fas cell surface death receptor (FAS)-mediated cell death and hypoxia inducible factor (HIF-1α), the targets involved in mitochondrial cell death. MATERIAL AND METHODS Male Sprague Dawley rats (N = 15, divided into three groups) were subjected to traumatic spinal cord injury and were injected with minocycline (n = 5) (90 mg/kg and later a 45 mg/kg dose twice a day (every 12 h)). Injection with sterile PBS in injured animals served as the vehicle (n = 5) and another group comprised healthy animals (n = 5). TUNEL assay was used to quantify cell death. The release of Smac/Diablo, cytochrome-c (cyt-c), HIF-1α, FAS ligand (FASL) and tumour necrosis factor-α (TNF-α) was measured using ELISA. Expression of HIF-1α, FASL and other cell death associated factors was quantified at the mRNA and protein level and confirmed with immunohistochemistry. RESULTS There was a marked reduction in the HIF-1α and FASL expression levels in the minocycline-treated group compared to the vehicle. The reduction of HIF-1α and FASL was associated with other factors linked to cell death (Smac/Diablo, cyt-c, TNF-α, p53, caspase-8 and BH3 interacting domain death agonist (BID)) (p < 0.5; *p < 0.05 vs. vehicle group, **p < 0.01 vs. vehicle group). CONCLUSIONS The present study focuses on the investigation of minocycline in inhibiting mitochondria-associated cell death by modulating FASL and HIF-1α expression, which are seemingly interlinked mechanisms contributing to cell death.
Collapse
Affiliation(s)
- Guolei Zhang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junpu Zha
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junchuan Liu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Di
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
39
|
Cui Y, Feng N, Gu X, Fu F, Li J, Guo H, Liu Y, Zhang S, Li J, Wang Y, Jia M, Yang L, Zhang F, Wang Y, Fan R, Pei J. κ-Opioid receptor stimulation reduces palmitate-induced apoptosis via Akt/eNOS signaling pathway. Lipids Health Dis 2019; 18:52. [PMID: 30764838 PMCID: PMC6376663 DOI: 10.1186/s12944-019-0989-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/28/2019] [Indexed: 12/02/2022] Open
Abstract
Background This study was designed to test the hypothesis that κ-opioid receptor (κ-OR) stimulation reduces palmitate-induced HUVECs apoptosis and to investigate its mechanisms. Methods HUVECs were subjected to sodium palmitate, apoptosis and cell viability were determined, HUVECs were treated with specific inhibitors to PI3K, Akt, eNOS and siRNAs targeting κ-OR and Akt. Groups were divided as follows: the control group, the sodium palmitate group, the sodium palmitate+U50,488H (a selective κ-OR agonist) group and the sodium palmitate+U50,488H + nor-BNI (a selective κ-OR antagonist) group. Results Treatment with sodium palmitate significantly reduced cell viability and increased apoptosis rate which were significantly alleviated by pretreatment with U50,488H, the effect of U50,488H was abolished by nor-BNI. Phosphorylation of Akt and eNOS, as well as NO production were attenuated and accompanied by an increased expression of caspase 3 when HUVECs were subjected to sodium palmitate, and all these changes were restored by pretreatment with U50,488H, the effects of U50,488H were abolished by nor-BNI, and specific inhibitors to PI3K, Akt, eNOS, respectively. SiRNAs targeting κ-OR or Akt abolished the effects of U50,488H on phosphorylation of Akt and eNOS as well as the expressions of caspase 3, Bax and Bcl-2. SiRNAs targeting Akt elicited no effect on the expression of κ-OR. Conclusion This study provides the evidence for the first time that κ-OR stimulation possesses anti-palmitate-induced apoptosis effect, which is mediated by PI3K/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Yan Cui
- Department of Nursing, Medical College of Xi'an Peihua University, Xi'an, 710125, Shaanxi Province, China
| | - Na Feng
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Jun Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Haitao Guo
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Yuanbo Wang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Min Jia
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Lu Yang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Fuyang Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Yuemin Wang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China
| | - Rong Fan
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China.
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi Province, China.
| |
Collapse
|
40
|
Xu L, Ding L, Su Y, Shao R, Liu J, Huang Y. Neuroprotective effects of curcumin against rats with focal cerebral ischemia-reperfusion injury. Int J Mol Med 2019; 43:1879-1887. [PMID: 30816425 DOI: 10.3892/ijmm.2019.4094] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/07/2019] [Indexed: 11/06/2022] Open
Affiliation(s)
- Lu Xu
- School of Pharmacy, Chongqing Medical and Pharmaceutical College, Chongqing 401331, P.R. China
| | - Ling Ding
- Pharmacy Department, The Central Hospital of Jiangjin, Chongqing 402260, P.R. China
| | - Yuanqi Su
- School of Pharmacy, Chongqing Medical and Pharmaceutical College, Chongqing 401331, P.R. China
| | - Ruyue Shao
- School of Pharmacy, Chongqing Medical and Pharmaceutical College, Chongqing 401331, P.R. China
| | - Jie Liu
- Pharmacy Department, The Central Hospital of Jiangjin, Chongqing 402260, P.R. China
| | - Yan Huang
- Scientific Research and Teaching Department, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| |
Collapse
|
41
|
Şahin S, Gürgen SG, Yazar U, İnce İ, Kamaşak T, Acar Arslan E, Diler Durgut B, Dilber B, Cansu A. Vitamin D protects against hippocampal apoptosis related with seizures induced by kainic acid and pentylenetetrazol in rats. Epilepsy Res 2019; 149:107-116. [DOI: 10.1016/j.eplepsyres.2018.12.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/04/2018] [Accepted: 12/14/2018] [Indexed: 11/26/2022]
|
42
|
Diao Y, Yan W, Sun W, Luo Y, Li J, Yin Y. The dual role of KCNQ/M channels upon OGD or OGD/R insults in cultured cortical neurons of mice: Timing is crucial in targeting M-channels against ischemic injur ies. J Cell Physiol 2018; 234:12714-12726. [PMID: 30523632 DOI: 10.1002/jcp.27889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 01/09/2023]
Abstract
KCNQ/M potassium channels play a vital role in neuronal excitability; however, it is required to explore their pharmacological modulation on N-Methyl- d-aspartic acid receptors (NMDARs)-mediated glutamatergic transmission of neurons upon ischemic insults. In the current study, both presynaptic glutamatergic release and activities of NMDARs were measured by NMDAR-induced miniature excitatory postsynaptic currents (mEPSCs) in cultured cortical neurons of C57 mice undergoing oxygen and glucose deprivation (OGD) or OGD/reperfusion (OGD/R). The KCNQ/M-channel opener, retigabine (RTG), suppressed the overactivation of postsynaptic NMDARs induced by OGD and then NO transient; RTG also decreased OGD-induced neuronal death measured with MTT assay, suggesting the beneficial role of KCNQ/M-channels for the neurons exposed to ischemic insults. However, when the neurons exposed to the subsequent reperfusion, KCNQ/M-channels played a differential role from its protective effect. OGD/R increased presynaptic glutamatergic release, which was further augmented by RTG or decreased by KCNQ/M-channel blocker, XE991. Reactive oxygen species (ROS) were produced partly in a NO-dependent manner. In addition, XE991 decreased neuronal injuries upon reperfusion measured with DCF and PI staining. Meanwhile, the addition of RTG upon OGD or XE991 upon reperfusion can reverse OGD or OGD/R-reduced mitochondrial membrane potential. Our present study indicates the dual role of KCNQ/M-channels in OGD and OGD/R, which will decide the fate of neurons. Provided that activation of KCNQ/M-channels has differential effects on neuronal injuries during OGD or OGD/R, we propose that therapy targeting KCNQ/M-channels may be effective for ischemic injuries but the proper timing is so crucial for the corresponding treatment.
Collapse
Affiliation(s)
- Yu Diao
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Weijie Yan
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanlin Luo
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanling Yin
- Department of Neurobiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Pascotini MET, Flores DAE, Kegler MA, Konzen MV, Fornari MAL, Arend MJ, Gabbi MP, Gobo MLA, Bochi DGV, Prado DALC, de Carvalho DLM, Duarte DMM, da Cruz DIBM, Moresco DRN, dos Santos DARS, Royes DLFF, Fighera DMR. Brain-Derived Neurotrophic Factor Levels are Lower in Chronic Stroke Patients: A Relation with Manganese-dependent Superoxide Dismutase ALA16VAL Single Nucleotide Polymorphism through Tumor Necrosis Factor-α and Caspases Pathways. J Stroke Cerebrovasc Dis 2018; 27:3020-3029. [DOI: 10.1016/j.jstrokecerebrovasdis.2018.06.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/03/2018] [Accepted: 06/24/2018] [Indexed: 12/19/2022] Open
|
44
|
Tsai YR, Chang CF, Lai JH, Wu JCC, Chen YH, Kang SJ, Hoffer BJ, Tweedie D, Luo W, Greig NH, Chiang YH, Chen KY. Pomalidomide Ameliorates H₂O₂-Induced Oxidative Stress Injury and Cell Death in Rat Primary Cortical Neuronal Cultures by Inducing Anti-Oxidative and Anti-Apoptosis Effects. Int J Mol Sci 2018; 19:ijms19103252. [PMID: 30347766 PMCID: PMC6213994 DOI: 10.3390/ijms19103252] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022] Open
Abstract
Due to its high oxygen demand and abundance of peroxidation-susceptible lipid cells, the brain is particularly vulnerable to oxidative stress. Induced by a redox state imbalance involving either excessive generation of reactive oxygen species (ROS) or dysfunction of the antioxidant system, oxidative stress plays a central role in a common pathophysiology that underpins neuronal cell death in acute neurological disorders epitomized by stroke and chronic ones such as Alzheimer’s disease. After cerebral ischemia, for example, inflammation bears a key responsibility in the development of permanent neurological damage. ROS are involved in the mechanism of post-ischemic inflammation. The activation of several inflammatory enzymes produces ROS, which subsequently suppress mitochondrial activity, leading to further tissue damage. Pomalidomide (POM) is a clinically available immunomodulatory and anti-inflammatory agent. Using H2O2-treated rat primary cortical neuronal cultures, we found POM displayed neuroprotective effects against oxidative stress and cell death that associated with changes in the nuclear factor erythroid derived 2/superoxide dismutase 2/catalase signaling pathway. POM also suppressed nuclear factor kappa-light-chain-enhancer (NF-κB) levels and significantly mitigated cortical neuronal apoptosis by regulating Bax, Cytochrome c and Poly (ADP-ribose) polymerase. In summary, POM exerted neuroprotective effects via its anti-oxidative and anti-inflammatory actions against H2O2-induced injury. POM consequently represents a potential therapeutic agent against brain damage and related disorders and warrants further evaluation.
Collapse
Affiliation(s)
- Yan-Rou Tsai
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
| | - Cheng-Fu Chang
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei City Hospital, Zhongxiao Branch, Taipei 11556, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jing-Huei Lai
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - John Chung-Che Wu
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Yen-Hua Chen
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Shuo-Jhen Kang
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Barry J Hoffer
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA.
| | - Weiming Luo
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA.
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 20892, USA.
| | - Yung-Hsiao Chiang
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 11031, Taiwan.
| | - Kai-Yun Chen
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 11031, Taiwan.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
45
|
Lixisenatide ameliorates cerebral ischemia-reperfusion injury via GLP-1 receptor dependent/independent pathways. Eur J Pharmacol 2018; 833:145-154. [DOI: 10.1016/j.ejphar.2018.05.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023]
|
46
|
Lv J, Liang Y, Tu Y, Chen J, Xie Y. Hypoxic preconditioning reduces propofol-induced neuroapoptosis via regulation of Bcl-2 and Bax and downregulation of activated caspase-3 in the hippocampus of neonatal rats. Neurol Res 2018; 40:767-773. [PMID: 29790425 DOI: 10.1080/01616412.2018.1477545] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Evidence has shown that propofol may cause widespread apoptotic neurodegeneration. Hypoxic preconditioning (HPC) was previously demonstrated to provide neuroprotection and brain recovery from either acute or chronic neurodegeneration in several cellular and animal models. Therefore, the present study was designed to investigate the protective effects of hypoxic preconditioning on apoptosis caused by propofol in neonatal rats. METHODS Propofol (100 mg/kg) was given to 7-day-old (P7) Sprague Dawley pups. Before the propofol injection, hypoxic preconditioning was administered by subjecting rats to five cycles of 10 min of hypoxia (8% O2) and 10 min of normoxia (21% O2), then 2 h of room air. We detected neuronal structure changes and apoptosis by hematoxylin and eosin (HE) staining and TUNEL assay, respectively. Bcl-2, Bax and cleaved-caspase-3 levels were quantified using Western blotting and immunohistochemistry. RESULT After treatment with propofol, Bcl-2 levels decreased and Bax and cleaved-caspase-3 levels increased. However, our results suggest that hypoxic preconditioning could reverse this change. Conclusion: Our results indicate that pretreatment with hypoxic preconditioning prevents propofol-induced neuroapoptosis by increasing the levels of Bcl-2 and decreasing the levels of Bax and cleaved-caspase-3.
Collapse
Affiliation(s)
- Jing Lv
- a Department of Anesthesiology , The First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Yubing Liang
- b Department of Anesthesiology , The Affiliated tumor hospital of Guangxi Medical University , Nanning , China
| | - Youbing Tu
- a Department of Anesthesiology , The First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Jing Chen
- a Department of Anesthesiology , The First Affiliated Hospital of Guangxi Medical University , Nanning , China
| | - Yubo Xie
- a Department of Anesthesiology , The First Affiliated Hospital of Guangxi Medical University , Nanning , China
| |
Collapse
|
47
|
Wang Y, Wang J, Li Y, Wang S, Zhu X. Platelet-rich Plasma Protects HUVECs against oX-LDL-induced Injury. Open Med (Wars) 2018; 13:41-52. [PMID: 29607413 PMCID: PMC5874509 DOI: 10.1515/med-2018-0007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/13/2017] [Indexed: 01/26/2023] Open
Abstract
Platelet-rich plasma (PRP) contains a variety of cytokines, some of which ameliorate oX-LDL (oxidized low-density lipoprotein)-induced endothelial cell (EC) injury. Therefore, we hypothesized that PRP might alleviate oX-LDL-induced injury. METHODOLOGY Human umbilical vein endothelial cells (HUVECs) were divided into four groups: a PPP (platelet-poor plasma) group, an oX-LDL group, an oX-LDL+PRP group and a PRP group. CCK-8 (Cell Counting Kit) assay, Annexin V-FITC/7-AAD and Hochest 33342 staining were performed to assess cell proliferation and apoptosis. Tube formation and cell migration assays were performed to evaluate HUVEC-mediated vasculogenesis and migration. Expression levels of Bcl-2, Bax, caspase-3, cleaved caspase-3, PI3K, Akt, eNOS p-Akt, p-eNOS, IL-6 and IL-1 were detected by western blotting or immunofluorescence. PRINCIPAL FINDINGS PRP promoted HUVEC proliferation in a non-linear pattern, protected HUVECs against oX-LDL-induced apoptosis and attenuated oX-LDL-mediated inhibition of HUVEC migration and vasculogenesis. Additionally, compared to the PPP group, PRP downregulated pro-apoptotic proteins (ratio of Bax/Bcl-2, caspase-3 and cleaved caspase-3) as well as IL-6 and IL-1. Moreover, the PI3K/Akt/eNOS pathway was activated by PRP and inactivated by oX-LDL. CONCLUSIONS It was demonstrated that PRP protected HUVECs against oX-LDL-induced injury and that the PI3K/Akt/eNOS pathway was activated in this process.
Collapse
Affiliation(s)
- Yang Wang
- First Affiliated Hospital of Sun Yat-Sen University, GuangZhou, GuangDong, China
| | - Jinsong Wang
- First Affiliated Hospital of Sun Yat-Sen University, GuangZhou, GuangDong, China
| | - Yonghui Li
- First Affiliated Hospital of Sun Yat-Sen University, GuangZhou, GuangDong, China
| | - Shenming Wang
- First Affiliated Hospital of Sun Yat-Sen University, GuangZhou, GuangDong, China
| | - Xiaonan Zhu
- Sun Yat-sen University Zhongshan School of Medicine, GuangZhou, China
| |
Collapse
|
48
|
Zhang G, Zha J, Liu J, Di J. WITHDRAWN: Minocycline an antimicrobial agent attenuates the mitochondrial dependent cell death and stabilizes the expression of HIF-1α in spinal cord injury. Microb Pathog 2018:S0882-4010(18)30284-5. [PMID: 29530807 DOI: 10.1016/j.micpath.2018.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/02/2018] [Accepted: 03/08/2018] [Indexed: 10/17/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Guolei Zhang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Junpu Zha
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Junchuan Liu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Jun Di
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| |
Collapse
|
49
|
Kapoor M, Sharma N, Sandhir R, Nehru B. Effect of the NADPH oxidase inhibitor apocynin on ischemia-reperfusion hippocampus injury in rat brain. Biomed Pharmacother 2018; 97:458-472. [DOI: 10.1016/j.biopha.2017.10.123] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 01/23/2023] Open
|
50
|
Kim BJ, Zack DJ. The Role of c-Jun N-Terminal Kinase (JNK) in Retinal Degeneration and Vision Loss. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1074:351-357. [PMID: 29721963 DOI: 10.1007/978-3-319-75402-4_43] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
c-Jun N-terminal kinase (JNK), a member of stress-induced mitogen-activated protein (MAP) kinase family, has been shown to modulate a variety of biological processes associated with neurodegenerative pathology of the retina. In particular, various retinal cell culture and animal models related to glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa indicate that JNK signaling may contribute to disease pathogenesis. This mini-review discusses the impact of JNK signaling in retinal disease, with a focus on retinal ganglion cells (RGCs), photoreceptor cells, retinal pigment epithelial (RPE) cells, and animal studies, with particular attention to modulation of JNK signaling as a potential therapeutic target for the treatment of retinal disease.
Collapse
Affiliation(s)
- Byung-Jin Kim
- The Wilmer Eye Institute, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Donald J Zack
- The Wilmer Eye Institute, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|