51
|
Shi YH, Li Y, Wang Y, Xu Z, Fu H, Zheng GQ. Ginsenoside-Rb1 for Ischemic Stroke: A Systematic Review and Meta-analysis of Preclinical Evidence and Possible Mechanisms. Front Pharmacol 2020; 11:285. [PMID: 32296332 PMCID: PMC7137731 DOI: 10.3389/fphar.2020.00285] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background Ischemic stroke is the most common type of stroke, while pharmacological therapy options are limited. Ginsenosides are the major bioactive compounds in Ginseng and have been found to have various pharmacological effects in the nervous system. In the present study, we sought to evaluate the effects of Ginsenoside-Rb1 (G-Rb1), an important ingredient of ginsenosides, and the probable neuroprotective mechanisms in experimental ischemic strokes. Methods Studies of G-Rb1 on ischemic stroke animal models were identified from 7 databases. No clinical trials were included in the analysis. The primary outcome measures were neurological function scores, infarct volume, evans blue content and/or brain water content (BWC). The second outcome measures were the possible neuroprotective mechanisms. All the data were analyzed by Rev Man 5.3. Result Pooled preclinical data showed that compared with the controls, G-Rb1 could improve neurological function (Zea Longa (n = 367, P < 0.01); mNSS (n = 70, P < 0.01); Water maze test (n = 48, P < 0.01); Bederson (n = 16, P < 0.01)), infarct area (TTC (n = 211, P < 0.01); HE (n = 26, P < 0.01)), as well as blood-brain barrier function (BWC (n = 64, P < 0.01); Evans blue content (n=26, P < 0.05)). It also can increase BDNF (n = 26, P < 0.01), Gap-43 (n = 16, P < 0.01), SOD (n = 30, P < 0.01), GSH (n = 16, P < 0.01), Nissl-positive cells (n = 12, P < 0.01), Nestin-positive cells (n = 10, P < 0.05), and reduce Caspase-3 (n = 36, P < 0.01), IL-1 (n = 32, P < 0.01), TNF-α (n = 72, P < 0.01), MDA (n = 18, P < 0.01), NO (n = 44, P < 0.01), NOX (n = 32, P < 0.05), ROS (n = 6, P < 0.05), NF-κB (P < 0.05) and TUNEL-positive cells (n = 52, P < 0.01). Conclusion Available findings demonstrated the preclinical evidence that G-Rb1 has a potential neuroprotective effect, largely through attenuating brain water content, promoting the bioactivities of neurogenesis, anti-apoptosis, anti-oxidative, anti-inflammatory, energy supplement and cerebral circulation.
Collapse
Affiliation(s)
- Yi-Hua Shi
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yong Wang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhen Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huan Fu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
52
|
Shu L, Shen X, Zhao Y, Zhao R, He X, Yin J, Su J, Li Q, Liu J. Mechanisms of transformation of nicotinamide mononucleotides to cerebral infarction hemorrhage based on MCAO model. Saudi J Biol Sci 2020; 27:899-904. [PMID: 32127769 PMCID: PMC7042676 DOI: 10.1016/j.sjbs.2019.12.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/16/2019] [Accepted: 12/12/2019] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The study aims at discussing the effect of nicotinamide mononucleotides on protecting hemorrhagic transformation of cerebral infarction in the middle cerebral artery occlusion (MCAO) model. METHOD Male mice aged 4-5 weeks and weighing about 22-35 g in Shanghai Ninth People's Hospital are divided into three groups: sham group, collagenase intracerebral hemorrhage model (cICH + Vehicle) group and collagenase nicotinamide mononucleotide (cICH + NMN) group. Then, the intervention therapy research is carried out. After 24 h, the neurological function, brain edema, hematoma volume, body weight, hemorrhage volume, RNA expression level, apoptosis, inflammatory factors and reactive oxygen species (ROS) content in surrounding tissues of mice are analyzed comprehensively. RESULTS Compared with the other two groups, nicotinamide mononucleotides in MCAO model have significant effects on improving neurological function, brain edema, inflammatory factors, body weight and cell apoptosis in mice, but have no significant effect on hemorrhage volume and hematoma volume in mice. CONCLUSION Nicotinamide mononucleotides can significantly improve the collagenase-induced intracerebral hemorrhage (ICH) model in mice under MCAO model, and they can protect the brain tissue of mice from RNA level to tissue cell level or mouse body weight and volume level.
Collapse
Affiliation(s)
- Liang Shu
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaolei Shen
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yaxue Zhao
- Department of Neurology, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Rong Zhao
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xinwei He
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jiawen Yin
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jingjing Su
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qiang Li
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jianren Liu
- Department of Neurology Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
53
|
Teertam SK, Jha S, Prakash Babu P. Up-regulation of Sirt1/miR-149-5p signaling may play a role in resveratrol induced protection against ischemia via p53 in rat brain. J Clin Neurosci 2020; 72:402-411. [PMID: 31866350 DOI: 10.1016/j.jocn.2019.11.043] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/30/2019] [Indexed: 12/19/2022]
Abstract
Micro-RNA(miRNA) are well studied small noncoding RNA, which plays a diverse role in the regulation of vital elements in cell survival and apoptosis. However, the functional significance of miRNAs after the pathogenesis of ischemic stroke remains unclear. The present study is designed to investigate the regulatory role of miR-149-5p on Sirtuin-1/p53 axis during ischemic-reperfusion-induced injury. Middle cerebral artery occlusion (MCAO) was performed by nylon monofilament for 60 min. Resveratrol was administered via intraperitoneal (IP) route, 30 min before the MCAO. Our study demonstrated that the miR-149-5p levels were markedly decreased at 24 h after ischemic-reperfusion (I/R) injury. Further, we observed decreased p53 protein expression and increased miR-149-5p activity on sirtuin1 (Sirt1) activation with resveratrol after 24 h following MCAO. Moreover, immunohistochemistry studies found that resveratrol treatment significantly decreased the immunoreactivity of p53 and caspase-3 on activation of Sirt1/miR149-5p axis. In conclusion, our findings suggest that miR-149-5p could play a regulatory role in neuronal cell death via Sirt1/p53 axis, which offers a new target for novel therapeutic interventions during acute ischemic stroke.
Collapse
Affiliation(s)
- Sireesh Kumar Teertam
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad, 500 046 TS, India
| | - Shekhar Jha
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad, 500 046 TS, India
| | - Phanithi Prakash Babu
- Department of Biotechnology & Bioinformatics, School of Life Sciences, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad, 500 046 TS, India.
| |
Collapse
|
54
|
Hyperbaric oxygen therapy in acute stroke: is it time for Justitia to open her eyes? Neurol Sci 2020; 41:1381-1390. [PMID: 31925614 DOI: 10.1007/s10072-020-04241-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/06/2020] [Indexed: 01/27/2023]
Abstract
Hypoxia is a critical component of neuronal death in patients with stroke. Therefore increasing oxygenation of brain tissue seems to be a logical therapy against cerebral ischemia. Oxygen therapy exists in two modalities: normobaric hyperoxia therapy and hyperbaric oxygen therapy (HBO). HBO is a therapeutic procedure in which pure (100%) oxygen is administered at greater than atmospheric pressure in HBO therapy chambers. In this review article, we aimed to summarize the current knowledge regarding the therapeutic use of HBO in acute stroke patients. Literature review and electronic search were performed using PubMed, Medscape, and UpToDate with the keywords stroke, acute stroke, hyperbaric oxygen therapy, and hyperoxia. According to the reviewed literature, the use of HBO as routine stroke therapy cannot be justified in acute stage of stroke. More randomized, controlled studies are needed regarding safety and especially effectives of HBO in stroke patients. Also, standardized definitionof HBO should be proposed and used in all future studies.
Collapse
|
55
|
Wang P, Lu Y, Han D, Wang P, Ren L, Bi J, Liang J. Neuroprotection by nicotinamide mononucleotide adenylyltransferase 1 with involvement of autophagy in an aged rat model of transient cerebral ischemia and reperfusion. Brain Res 2019; 1723:146391. [PMID: 31421130 DOI: 10.1016/j.brainres.2019.146391] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/20/2019] [Accepted: 08/13/2019] [Indexed: 11/18/2022]
Abstract
Recent researches suggest that autophagic degradation declines with age, and this leads to an accumulation of damage that contributes to age-related cellular dysfunction. Nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) shows therapeutic potential for cerebral ischemia in young-adult animals. This study investigated the role of NMNAT1 in focal cerebral ischemia in aged rats with a focus on neuronal autophagy. Focal cerebral ischemia was induced in aged rats by middle cerebral artery occlusion (MCAO). NMNAT1 levels in the peri-infarct penumbra increased at 12 and 24 h after ischemia in aged rats. Knockdown of NMNAT1 significantly increased infarct volume, whereas overexpression of NMNAT1 reduced ischemia-induced cerebral injuries in aged rats with acute ischemic stroke. Meanwhile, lentiviral overexpression of NMNAT1 increased autophagy, reduced the phosphorylation of mammalian target of rapamycin (mTOR), and enhanced the sirtuin 1 (SIRT1) protein level. In cultured cortical neurons, SIRT1 regulated the mTOR-mediated autophagy upon oxygen-glucose deprivation (OGD) stress and the effect of NMNAT1 on autophagy was blocked in cultured SIRT1-knockout neurons. Furthermore, autophagy inhibitor 3-methyladenine (3-MA) partly abolished the neuroprotection induced by NMNAT1 overexpression. The results suggest NMNAT1 protects against acute ischemic stroke in aged rats by inducing autophagy via regulating the SIRT1/mTOR pathway.
Collapse
Affiliation(s)
- Peng Wang
- Liaoning Key Laboratory of Neurodegenerative Diseases and Department of Neurobiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yijun Lu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Donghe Han
- Liaoning Key Laboratory of Neurodegenerative Diseases and Department of Neurobiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Pan Wang
- Liaoning Key Laboratory of Neurodegenerative Diseases and Department of Neurobiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lili Ren
- Liaoning Key Laboratory of Neurodegenerative Diseases and Department of Neurobiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jing Bi
- Liaoning Key Laboratory of Neurodegenerative Diseases and Department of Neurobiology, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jia Liang
- Institute of Life Science, Jinzhou Medical University, Jinzhou, Liaoning, China.
| |
Collapse
|
56
|
Gómez-de Frutos MC, Laso-García F, Diekhorst L, Otero-Ortega L, Fuentes B, Jolkkonen J, Detante O, Moisan A, Martínez-Arroyo A, Díez-Tejedor E, Gutiérrez-Fernández M. Intravenous delivery of adipose tissue-derived mesenchymal stem cells improves brain repair in hyperglycemic stroke rats. Stem Cell Res Ther 2019; 10:212. [PMID: 31315686 PMCID: PMC6637493 DOI: 10.1186/s13287-019-1322-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/14/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022] Open
Abstract
Background Over 50% of acute stroke patients have hyperglycemia, which is associated with a poorer prognosis and outcome. Our aim was to investigate the impact of hyperglycemia on behavioral recovery and brain repair of delivered human adipose tissue-derived mesenchymal stem cells (hAD-MSCs) in a rat model of permanent middle cerebral artery occlusion (pMCAO). Methods Hyperglycemia was induced in rats by the administration of nicotinamide and streptozotocin. The rats were then subjected to stroke by a pMCAO model. At 48 h post-stroke, 1 × 106 hAD-MSCs or saline were intravenously administered. We evaluated behavioral outcome, infarct size by MRI, and brain plasticity markers by immunohistochemistry (glial fibrillary acidic protein [GFAP], Iba-1, synaptophysin, doublecortin, CD-31, collagen-IV, and α-smooth muscle actin [α-SMA]). Results The hyperglycemic group exhibited more severe neurological deficits; lesion size and diffusion coefficient were larger compared with the non-hyperglycemic rats. GFAP, Iba-1, and α-SMA were increased in the hyperglycemic group. The hyperglycemic rats administered hAD-MSCs at 48 h after pMCAO had improved neurological impairment. Although T2-MRI did not show differences in lesion size between groups, the rADC values were lower in the treated group. Finally, the levels of GFAP, Iba-1, and arterial wall thickness were lower in the treated hyperglycemic group than in the nontreated hyperglycemic group at 6 weeks post-stroke. Conclusions Our data suggest that rats with hyperglycemic ischemic stroke exhibit increased lesion size and impaired brain repair processes, which lead to impairments in behavioral recovery after pMCAO. More importantly, hAD-MSC administration induced better anatomical tissue preservation, associated with a good behavioral outcome. Electronic supplementary material The online version of this article (10.1186/s13287-019-1322-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mari Carmen Gómez-de Frutos
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Fernando Laso-García
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Luke Diekhorst
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Laura Otero-Ortega
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Blanca Fuentes
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Jukka Jolkkonen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland.,NeuroCenter, Kuopio University Hospital, Kuopio, Finland
| | - Olivier Detante
- Neurology Department, Stroke Unit, Grenoble Hospital, Grenoble, France.,Grenoble Institute of Neurosciences, Inserm U1216, Grenoble Alpes University, Grenoble, France
| | - Anaick Moisan
- Grenoble Institute of Neurosciences, Inserm U1216, Grenoble Alpes University, Grenoble, France.,Cell Therapy and Engineering Unit, EFS Auvergne Rhône Alpes, Saint-Ismier, France
| | - Arturo Martínez-Arroyo
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María Gutiérrez-Fernández
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Autonoma University of Madrid, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | | |
Collapse
|
57
|
Zuo X, Lu J, Manaenko A, Qi X, Tang J, Mei Q, Xia Y, Hu Q. MicroRNA-132 attenuates cerebral injury by protecting blood-brain-barrier in MCAO mice. Exp Neurol 2019; 316:12-19. [PMID: 30930097 DOI: 10.1016/j.expneurol.2019.03.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/24/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022]
Abstract
MicroRNAs (miRNAs) have been widely reported to induce posttranscriptional gene silencing and led to an explosion of new strategies for the treatment of human disease. It has been reported that the expression of MicroRNA-132 (miR-132) are altered both in the blood and brain after stroke. However, the effect of miR-132 on blood-brain barrier (BBB) disruption in ischemia stroke has not been studied. Here we will investigate the effects of miR-132 on the permeability of BBB after ischemic stroke and explore the potential mechanism underlying observed protection. Eight week-old mice were injected intracerebroventricularly with miR-132, antagomir-132 or agomir negative control (agomir-NC) 2 h before middle cerebral artery occlusion (MCAO), followed by animal behavior tests and infraction volume measurement at 24 h after MCAO. BBB permeability and integrity were measured by Evan's blue extravasation and brain water content. The expression of tight junction proteins was detected by immnostaining and Western blots. The level of MiR-132 and its targeted gene Mmp9 were assayed. Treatment with exogenous MiR-132 (agomir-132) decreased the infraction volume, reduced brain edema, and improved neurological functions compared to control mice. Agomir-132 increased the level of MiR-132 in brain tissue, suppressed the expression of MMP-9 mRNA and decreased the degradation of tight junction proteins VE-cadherin and β-Catenin in ischemic stroke mice. Inhibition of MMP-9 has a similar protective effect to agomir-132 on infraction volume, brain edema, and tight-junction protein expression after MCAO. Our results indicated that miR-132/MMP-9 axis might be a novel therapeutic target for BBB protection in ischemic stroke.
Collapse
Affiliation(s)
- Xiaokun Zuo
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, Haikou, China; Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anatol Manaenko
- Departments of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Xin Qi
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiping Tang
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, the Second Military Medical University, Shanghai, China.
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya Medical College of Central South University, Haikou, China.
| | - Qin Hu
- Discipline of Neuroscience, Department of Anatomy, Histology and Embryology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
58
|
Elgebaly MM, Arreguin J, Storke N. Targets, Treatments, and Outcomes Updates in Diabetic Stroke. J Stroke Cerebrovasc Dis 2019; 28:1413-1420. [PMID: 30904470 DOI: 10.1016/j.jstrokecerebrovasdis.2019.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 02/11/2019] [Indexed: 12/13/2022] Open
Abstract
GOAL Due to multiple failures to translate basic research, the need for novel therapeutic targets and strategies is still urgent to save a larger number of the stroke patients' population and to reduce the toxicity of the current stroke therapy. METHOD We summarize the most recent, within past 5 years, basic and clinical diabetic stroke research findings. FINDINGS We aim to examine the most current understanding of stroke and neurovascular unit integrity, especially in presence of hyperglycemia and/or diabetes mellitus. From there, we are comparing the meaningful findings that aim at treating diabetic stroke to see where they differ, where they succeed, and where they open questions for new therapeutic strategies. CONCLUSION The need for more clinically effective neuroprotective strategies is still mismatched with the bench side findings.
Collapse
Affiliation(s)
- Mostafa M Elgebaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, Florida.
| | - Jennifer Arreguin
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, Florida
| | - Niko Storke
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, Florida
| |
Collapse
|
59
|
Nikseresht S, Khodagholi F, Ahmadiani A. Protective effects of ex-527 on cerebral ischemia-reperfusion injury through necroptosis signaling pathway attenuation. J Cell Physiol 2019; 234:1816-1826. [PMID: 30067864 DOI: 10.1002/jcp.27055] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022]
Abstract
Necroptosis, a novel type of programmed cell death, is involved in ischemia-reperfusion-induced brain injury. Sirtuin 1 (Sirt1), as a well-known member of histone deacetylase class III, plays pivotal roles in inflammation, metabolism, and neuron loss in cerebral ischemia. We explored the relationship between Sirt1 and the necroptosis signaling pathway and its downstream events by administration of ex-527, as a selective and potent inhibitor of Sirt1, and necrostatin-1 (nec-1), as a necroptosis inhibitor, in an animal model of focal cerebral ischemia. Our data showed different patterns of sirt1 and necroptosis critical regulators, including receptor-interacting protein kinase 3 and mixed lineage kinase domain-like protein gene expressions in the prefrontal cortex and the hippocampus after ischemia-reperfusion. We found that ex-527 microinjection reduces the infarction volume of ischemic brains and improves the survival rate, but not stroke-associated neurological deficits. Additionally, treatment with ex-527 effectively abolished the elevation of the critical regulators of necroptosis, whereas necroptosis inhibition through nec-1 microinjection did not influence Sirt1 expression levels. Our data also demonstrated that the ex-527 relieves ischemia-induced perturbation of necroptosis-associated metabolic enzymes activity in downstream. This study provides a new approach to the possible neuroprotective potential of ex-527 orchestrated by necroptosis pathway inhibition to alleviate ischemia-reperfusion brain injury.
Collapse
Affiliation(s)
- Sara Nikseresht
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
60
|
Lv G, Wang GQ, Xia ZX, Wang HX, Liu N, Wei W, Huang YH, Zhang WW. Influences of blood lipids on the occurrence and prognosis of hemorrhagic transformation after acute cerebral infarction: a case-control study of 732 patients. Mil Med Res 2019; 6:2. [PMID: 30665465 PMCID: PMC6341695 DOI: 10.1186/s40779-019-0191-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 01/02/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND To study the influence of blood lipid levels on hemorrhagic transformation (HT) and prognosis after acute cerebral infarction (ACI). METHODS Patients with ACI within 72 h of symptoms onset between January 1st, 2015, and December 31st, 2016, were retrospectively analyzed. Patients were divided into group A (without HT) and group B (HT). The outcomes were assessed after 3 months of disease onset using the modified Rankin Scale (mRS). An mRS score of 0-2 points indicated excellent prognosis, and an mRS score of 3-6 points indicated poor prognosis. RESULTS A total of 732 patients conformed to the inclusion criteria, including 628 in group A and 104 in group B. The incidence of HT was 14.2%, and the median onset time was 2 d (interquartile range, 1-7 d). The percentages of patients with large infarct size and cortex involvement in group B were 80.8 and 79.8%, respectively, which were both significantly higher than those in group A (28.7 and 33.4%, respectively). The incidence rate of atrial fibrillation (AF) in group B was significantly higher than that in group A (39.4% vs. 13.9%, P < 0.001). The adjusted multivariate analysis results showed that large infarct size, cortex involvement and AF were independent risk factors of HT, while total cholesterol (TC) was a protective factor of HT (OR = 0.359, 95% CI 0.136-0.944, P = 0.038). With every 1 mmol/L reduction in normal TC levels, the risk of HT increased by 64.1%. The mortality and morbidity at 3 months in group B (21.2 and 76.7%, respectively) were both significantly higher than those in group A (8.0 and 42.8%, respectively). The adjusted multivariate analysis results showed that large infarct size (OR = 12.178, 95% CI 5.390-27.516, P < 0.001) was an independent risk factor of long-term unfavorable outcomes, whereas low-density lipoprotein cholesterol (LDL-C) was a protective factor (OR = 0.538, 95% CI 0.300-0.964, P = 0.037). With every 1 mmol/L reduction in normal LDL-C levels, the risk of an unfavorable outcome increased by 46.2%. Major therapies, including intravenous recombinant human tissue plasminogen activator (rTPA), intensive lipid-lowering statins and anti-platelets, were not significantly related to either HT or long-term, post-ACI poor prognosis. CONCLUSION For patients with large infarct sizes, especially those with cortex involvement, AF, or lower levels of TC, the risk of HT might increase after ACI. The risk of a long-term unfavorable outcome in these patients might increase with a reduction in LDL-C.
Collapse
Affiliation(s)
- Gang Lv
- Department of General Surgery, 309 Hospital of Chinese People's Liberation Army, Beijing, 100091, China
| | - Guo-Qiang Wang
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China.
| | - Zhen-Xi Xia
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China
| | - Hai-Xia Wang
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China
| | - Nan Liu
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China
| | - Wei Wei
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China
| | - Yong-Hua Huang
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China
| | - Wei-Wei Zhang
- Department of Neurology, Chinese PLA Army General Hospital, Beijing, 100700, China
| |
Collapse
|
61
|
New progress in the approaches for blood–brain barrier protection in acute ischemic stroke. Brain Res Bull 2019; 144:46-57. [DOI: 10.1016/j.brainresbull.2018.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
|
62
|
Eser Ocak P, Ocak U, Sherchan P, Zhang JH, Tang J. Insights into major facilitator superfamily domain-containing protein-2a (Mfsd2a) in physiology and pathophysiology. What do we know so far? J Neurosci Res 2018; 98:29-41. [PMID: 30345547 DOI: 10.1002/jnr.24327] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 08/20/2018] [Accepted: 08/28/2018] [Indexed: 01/02/2023]
Abstract
Major facilitator superfamily domain-containing protein-2a (Mfsd2a) which was considered as an orphan transporter has recently gained attention for its regulatory role in the maintenance of proper functioning of the blood-brain barrier. Besides the major role of Mfsd2a in maintaining the barrier function, increasing evidence has emerged with regard to the contributions of Mfsd2a to various biological processes such as transport, cell fusion, cell cycle, inflammation and regeneration, managing tumor growth, functioning of other organs with barrier functions or responses to injury. The purpose of this article is to review the different roles of Mfsd2a and its involvement in the physiological and pathophysiological processes primarily in the central nervous system and throughout the mammalian body under the lights of the current literature.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Umut Ocak
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
63
|
Knecht T, Borlongan C, Dela Peña I. Combination therapy for ischemic stroke: Novel approaches to lengthen therapeutic window of tissue plasminogen activator. Brain Circ 2018; 4:99-108. [PMID: 30450415 PMCID: PMC6187940 DOI: 10.4103/bc.bc_21_18] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 12/24/2022] Open
Abstract
Tissue plasminogen activator (tPA) thrombolysis continues to be the gold standard therapy for ischemic stroke. Due to the time-limited treatment window, within 4.5 h of stroke onset, and a variety of potentially deadly complications related to delayed administration, particularly hemorrhagic transformation (HT), clinical use of tPA is limited. Combination therapies with other interventions, drug or nondrug, have been hypothesized as a logical approach to enhancing tPA effectiveness. Here, we discuss various potential pharmacological and nondrug treatments to minimize adverse effects, primarily HT, associated with delayed tPA administration. Pharmacological interventions include many that support the integrity of the blood–brain barrier (i.e., atorvastatin, batimastat, candesartan, cilostazol, fasudil, and minocycline), promote vascularization and preserve cerebrovasculature (i.e., coumarin derivative IMM-H004 and granulocyte-colony stimulating factor), employing other mechanisms of action (i.e., oxygen transporters and ascorbic acid). Nondrug treatments are comprised of stem cell transplantation and gas therapies with multi-faceted approaches. Combination therapy with tPA and the aforementioned treatments demonstrated promise for mitigating the adverse complications associated with delayed tPA treatment and rescuing stroke-induced behavioral deficits. Therefore, the conjunctive therapy method is a novel therapeutic approach that can attempt to minimize the limitations of tPA treatment and possibly increase the therapeutic window for ischemic stroke treatment.
Collapse
Affiliation(s)
- Talia Knecht
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| | - Cesar Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
64
|
Jackson CW, Escobar I, Xu J, Perez-Pinzon MA. Effects of ischemic preconditioning on mitochondrial and metabolic neruoprotection: 5' adenosine monophosphate-activated protein kinase and sirtuins. Brain Circ 2018; 4:54-61. [PMID: 30276337 PMCID: PMC6126241 DOI: 10.4103/bc.bc_7_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 02/07/2023] Open
Abstract
Stroke and cardiac arrest result in cerebral ischemia, a highly prevalent medical issue around the world, which is characterized by a reduction or loss of blood flow to the brain. The loss of adequate nutrient supply in the brain during ischemia results in neuronal cell death contributing to cognitive and motor deficits that are usually permanent. Current effective therapies for cerebral ischemia are only applicable after the fact. Thus, the development of preventative therapies of ischemia is imperative. A field of research that continues to show promise in developing therapies for cerebral ischemia is ischemic preconditioning (IPC). IPC is described as exposure to sublethal ischemic events, which induce adaptive changes that provide tolerance to future ischemic events. Through either transient sub-lethal ischemic events, or the actions of a preconditioning molecular mimetic, IPC typically results in augmented gene expression and cellular metabolism. A pivotal target of such changes in gene expression and metabolism is the mitochondrion. Direct and indirect effects on mitochondria by IPC can result in the activation of 5’ adenosine monophosphate-activated protein kinase (AMPK), a master regulator of cellular metabolism. Changes in the activity of the posttranslational modifiers, SIRT1 and SIRT5, also contribute to the overall adaptive processes in cellular metabolism and mitochondrial functioning. In this review, we present recently collected evidence to highlight the neuroprotective interactions of mitochondria with AMPK, SIRT1, and SIRT5 in IPC. To produce this review, we utilized PubMed and previous reviews to target and to consolidate the relevant studies and lines of evidence.
Collapse
Affiliation(s)
- Charles W Jackson
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Iris Escobar
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jing Xu
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
65
|
Liska GM, Lippert T, Russo E, Nieves N, Borlongan CV. A Dual Role for Hyperbaric Oxygen in Stroke Neuroprotection: Preconditioning of the Brain and Stem Cells. CONDITIONING MEDICINE 2018; 1:151-166. [PMID: 30079404 PMCID: PMC6075658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Stroke continues to be an extremely prevalent disease and poses a great challenge in developing safe and effective therapeutic options. Hyperbaric oxygen therapy (HBOT) has demonstrated significant pre-clinical effectiveness for the treatment of acute ischemic stroke, and limited potential in treating chronic neurological deficits. Reported benefits include reductions in oxidative stress, inflammation, neural apoptosis, and improved physiological metrics such as edema and oxygen perfusion, all of which contribute to improved functional recovery. This pre-clinical evidence has failed to translate into an effective evidence-based therapy, however, due in large part to significant inconsistencies in treatment protocols and design of clinical studies. While the medical community works to standardize clinical protocols in an effort to advance HBOT for acute stroke, pre-clinical investigations continue to probe novel applications of HBOT in an effort to optimize stroke neuroprotection. One such promising strategy is HBOT preconditioning. Based upon the premise of mild oxidative stress priming the brain for tolerating the full-blown oxidative stress inherent in stroke, HBOT preconditioning has displayed extensive efficacy. Here, we first review the pre-clinical and clinical evidence supporting HBOT delivery following ischemic stroke and then discuss the scientific basis for HBOT preconditioning as a neuroprotective strategy. Finally, we propose the innovative concept of stem cell preconditioning, in tandem with brain preconditioning, as a promising regenerative pathway for maximizing the application of HBOT for ischemic stroke treatment.
Collapse
Affiliation(s)
| | | | | | | | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL
| |
Collapse
|
66
|
Acute Hyperbaric Oxygenation, Contrary to Intermittent Hyperbaric Oxygenation, Adversely Affects Vasorelaxation in Healthy Sprague-Dawley Rats due to Increased Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7406027. [PMID: 29854092 PMCID: PMC5949176 DOI: 10.1155/2018/7406027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/27/2017] [Accepted: 02/15/2018] [Indexed: 02/07/2023]
Abstract
The present study was aimed at assessing endothelium-dependent vasorelaxation, at measuring superoxide production in the aorta and femoral artery, and at determining antioxidative enzyme expression and activity in aortas of male Sprague-Dawley rats (N = 135), randomized to an A-HBO2 group exposed to a single hyperbaric oxygenation session (120′ of 100% O2 at 2.0 bars), a 24H-HBO2 group (single session, examined 24 h after exposure), a 4D-HBO2 group (4 consecutive days of single sessions), and a CTRL group (untreated group). Vasorelaxation of aortic rings in response to acetylcholine (AChIR) and to reduced pO2 (HIR) was tested in vitro in the absence/presence of NOS inhibitor L-NAME and superoxide scavenger TEMPOL. eNOS, iNOS, antioxidative enzyme, and NADPH oxidase mRNA expression was assessed by qPCR. Serum oxidative stress markers and enzyme activity were assessed by spectrometry, and superoxide production was determined by DHE fluorescence. Impaired AChIR and HIR in the A-HBO2 group were restored by TEMPOL. L-NAME inhibited AChIR in all groups. Serum oxidative stress and superoxide production were increased in the A-HBO2 group compared to all other groups. The mRNA expression of iNOS was decreased in the A-HBO2 and 24H-HBO2 groups while SOD1 and 3 and NADPH oxidase were increased in the 4D-HBO2 group. The expression and activity of catalase and glutathione peroxidase were increased in the 4D-HBO2 group as well. AChIR was NO dependent. Acute HBO2 transiently impaired vasorelaxation due to increased oxidative stress. Vasorelaxation was restored and oxidative stress was normalized 24 h after the treatment.
Collapse
|
67
|
Adjunctive Therapy Approaches for Ischemic Stroke: Innovations to Expand Time Window of Treatment. Int J Mol Sci 2017; 18:ijms18122756. [PMID: 29257093 PMCID: PMC5751355 DOI: 10.3390/ijms18122756] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 12/12/2017] [Accepted: 12/16/2017] [Indexed: 12/18/2022] Open
Abstract
Tissue plasminogen activator (tPA) thrombolysis remains the gold standard treatment for ischemic stroke. A time-constrained therapeutic window, with the drug to be given within 4.5 h after stroke onset, and lethal side effects associated with delayed treatment, most notably hemorrhagic transformation (HT), limit the clinical use of tPA. Co-administering tPA with other agents, including drug or non-drug interventions, has been proposed as a practical strategy to address the limitations of tPA. Here, we discuss the pharmacological and non-drug approaches that were examined to mitigate the complications-especially HT-associated with delayed tPA treatment. The pharmacological treatments include those that preserve the blood-brain barrier (e.g., atovarstatin, batimastat, candesartan, cilostazol, fasudil, minocycline, etc.), enhance vascularization and protect the cerebrovasculature (e.g., coumarin derivate IMM-H004 and granulocyte-colony stimulating factor (G-CSF)), and exert their effects through other modes of action (e.g., oxygen transporters, ascorbic acid, etc.). The non-drug approaches include stem cell treatments and gas therapy with multi-pronged biological effects. Co-administering tPA with the abovementioned therapies showed promise in attenuating delayed tPA-induced side effects and stroke-induced neurological and behavioral deficits. Thus, adjunctive treatment approach is an innovative therapeutic modality that can address the limitations of tPA treatment and potentially expand the time window for ischemic stroke therapy.
Collapse
|
68
|
She DT, Jo DG, Arumugam TV. Emerging Roles of Sirtuins in Ischemic Stroke. Transl Stroke Res 2017; 8:10.1007/s12975-017-0544-4. [PMID: 28656393 DOI: 10.1007/s12975-017-0544-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is one of the leading causes of death worldwide. It is characterized by a sudden disruption of blood flow to the brain causing cell death and damage, which will lead to neurological impairments. In the current state, only one drug is approved to be used in clinical setting and new therapies that confer ischemic neuroprotection are desperately needed. Several targets and pathways have been indicated to be neuroprotective in ischemic stroke, among which the sirtuin family of nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases has emerged as important modulators of several processes in the normal physiology and pathological conditions such as stroke. Recent studies have identified some members of the sirtuin family are able to ameliorate the devastating consequences of ischemic stroke by conferring neuroprotection by means of reducing neuronal cell death, oxidative stress, and neuroinflammation whereas some sirtuins are found to be detrimental in the pathophysiology of ischemic stroke. This review summarizes implications of sirtuins in ischemic stroke and the experimental evidences that demonstrate the potential of sirtuin modulators as neuroprotective therapy for ischemic stroke.
Collapse
Affiliation(s)
- David T She
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Neurobiology/Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Neurobiology/Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|