1
|
Lee NT, Savvidou I, Selan C, Calvello I, Vuong A, Wright DK, Brkljaca R, Willcox A, Chia JSJ, Wang X, Peter K, Robson SC, Medcalf RL, Nandurkar HH, Sashindranath M. Development of endothelial-targeted CD39 as a therapy for ischemic stroke. J Thromb Haemost 2024; 22:2331-2344. [PMID: 38754782 DOI: 10.1016/j.jtha.2024.04.023] [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: 12/21/2023] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Ischemic stroke is characterized by a necrotic lesion in the brain surrounded by an area of dying cells termed the penumbra. Salvaging the penumbra either with thrombolysis or mechanical retrieval is the cornerstone of stroke management. At-risk neuronal cells release extracellular adenosine triphosphate, triggering microglial activation and causing a thromboinflammatory response, culminating in endothelial activation and vascular disruption. This is further aggravated by ischemia-reperfusion injury that follows all reperfusion therapies. The ecto-enzyme CD39 regulates extracellular adenosine triphosphate by hydrolyzing it to adenosine, which has antithrombotic and anti-inflammatory properties and reverses ischemia-reperfusion injury. OBJECTIVES The objective off the study was to determine the efficacy of our therapeutic, anti-VCAM-CD39 in ischaemic stroke. METHODS We developed anti-VCAM-CD39 that targets the antithrombotic and anti-inflammatory properties of recombinant CD39 to the activated endothelium of the penumbra by binding to vascular cell adhesion molecule (VCAM)-1. Mice were subjected to 30 minutes of middle cerebral artery occlusion and analyzed at 24 hours. Anti-VCAM-CD39 or control agents (saline, nontargeted CD39, or anti-VCAM-inactive CD39) were given at 3 hours after middle cerebral artery occlusion. RESULTS Anti-VCAM-CD39 treatment reduced neurologic deficit; magnetic resonance imaging confirmed significantly smaller infarcts together with an increase in cerebrovascular perfusion. Anti-VCAM-CD39 also restored blood-brain barrier integrity and reduced microglial activation. Coadministration of anti-VCAM-CD39 with thrombolytics (tissue plasminogen activator [tPA]) further reduced infarct volumes and attenuated blood-brain barrier permeability with no associated increase in intracranial hemorrhage. CONCLUSION Anti-VCAM-CD39, uniquely targeted to endothelial cells, could be a new stroke therapy even when administered 3 hours postischemia and may further synergize with thrombolytic therapy to improve stroke outcomes.
Collapse
Affiliation(s)
- Natasha Ting Lee
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia; Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Ioanna Savvidou
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Carly Selan
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Ilaria Calvello
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Amy Vuong
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - David K Wright
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Robert Brkljaca
- Monash Biomedical Imaging, Monash University, Clayton, Victoria, Australia
| | - Abbey Willcox
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Joanne S J Chia
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia; Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Simon C Robson
- Center for Inflammation Research, Department of Anesthesia, Critical Care & Pain Medicine and Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert L Medcalf
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Harshal H Nandurkar
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia
| | - Maithili Sashindranath
- Australian Centre for Blood Diseases, School of Translational Medicine, Monash University, Alfred Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
2
|
Bagheri SM, Allahtavakoli M, Hakimizadeh E. Neuroprotective effect of ischemic postconditioning against hyperperfusion and its mechanisms of neuroprotection. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:31. [PMID: 39239075 PMCID: PMC11376715 DOI: 10.4103/jrms.jrms_341_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/13/2023] [Accepted: 06/27/2023] [Indexed: 09/07/2024]
Abstract
Background In recent years, stroke and ischemia-reperfusion injury has motivated researchers to find new ways to reduce the complications. Although reperfusion is essential for brain survival, it is like a double-edged sword that may cause further damage to the brain. Ischemic postconditioning (IPostC) refers to the control of blood flow in postischemia-reperfusion that can reduce ischemia-reperfusion injuries. Materials and Methods Articles were collected by searching for the terms: Ischemic postconditioning and neuroprotective and ischemic postconditioning and hyperperfusion. Suitable articles were collected from electronic databases, including ISI Web of Knowledge, Medline/PubMed, ScienceDirect, Embase, Scopus, Biological Abstract, Chemical Abstract, and Google Scholar. Results New investigations show that IPostC has protection against hyperperfusion by reducing the amount of blood flow during reperfusion and thus reducing infarction volume, preventing the blood-brain barrier damage, and reducing the rate of apoptosis through the activation of innate protective systems. Numerous mechanisms have been suggested for IPostC, which include reduction of free radical production, apoptosis, inflammatory factors, and activation of endogenous protective pathways. Conclusion It seems that postconditioning can prevent damage to the brain by reducing the flow and blood pressure caused by hyperperfusion. It can protect the brain against damages such as stroke and hyperperfusion by activating various endogenous protection systems. In the present review article, we tried to evaluate both useful aspects of IPostC, neuroprotective effects, and fight against hyperperfusion.
Collapse
Affiliation(s)
- Seyyed Majid Bagheri
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Hakimizadeh
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
3
|
Behzadifard M, Aboutaleb N, Dolatshahi M, Khorramizadeh M, Mirshekari Jahangiri H, Kord Z, Nazarinia D. Neuroprotective Effects of Conditioned Medium of Mesenchymal Stem Cells (MSC-CM) as a Therapy for Ischemic Stroke Recovery: A Systematic Review. Neurochem Res 2022; 48:1280-1292. [PMID: 36581731 DOI: 10.1007/s11064-022-03848-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/31/2022]
Abstract
It has been reported that the therapeutic potential of stem cells is mainly mediated by their paracrine factors. In order to identify the effects of conditioned medium of mesenchymal stem cells (MSC-CM) against stroke, a systematic review was conducted. We searched PubMed, Scopus, and ISI Web of Science databases for all available articles relevant to the effects of MSC-CM against the middle cerebral artery occlusion (MCAO) model of ischemic stroke until August 2022. The quality of the included studies was evaluated using The STAIR scale. During the systematic search, a total of 356 published articles were found. A total of 15 datasets were included following screening for eligibility. The type of cerebral ischemia was the MCAO model and CM was obtained from MSCs. The results showed that the therapeutic time window can be considered a crucial factor when researchers use MSC-CM for stroke therapy. In addition, MSC-CM therapy contributes to functional recovery and reduces infarct volume after stroke by targeting different cellular signaling pathways. Our findings showed that MSC-CM therapy has the ability to improve functional recovery and attenuate brain infarct volume after ischemic stroke in preclinical studies. We hope our study accelerates needed progress towards clinical trials.
Collapse
Affiliation(s)
- Mahin Behzadifard
- Department of Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Nahid Aboutaleb
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Dolatshahi
- Department of Physiology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Maryam Khorramizadeh
- Department of Medical Physics, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | | | - Zeynab Kord
- Department of Anaesthesiology, School of Allied Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Donya Nazarinia
- Department of Laboratory Sciences, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran. .,Department of Physiology, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
| |
Collapse
|
4
|
Involvement of T-bet and GATA3 transcription factors in Mesenchymal stem cells and royal jelly combination treatment in brain stroke. J Stroke Cerebrovasc Dis 2022; 31:106678. [PMID: 35882098 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Mesenchymal stem cells (MSC) therapy is a promising therapeutic strategy to overcome the brain stroke side effects. However, it may be associated with long-term complications, including induction of inflammation. This project was designed to examine the effects of MSC administration and its combination with royal jelly (RJ) on the differentiation of T helper subsets. MATERIAL AND METHODS In this project, the mice were divided to the six groups, including control (healthy without stroke), stroke (mice model of middle cerebral artery occlusion (MCAO)), treated with mouse MSC (mMSC), royal jelly (RJ), combination of mMSC and RJ (mMSC + RJ) and MSC conditioned medium (SUP). Thereafter, sticky test, brain mRNA levels of T-bet (transcription factor for Th1 subset), GATA3 (transcription factor for Th2 subset), and ROR-γ (transcription factor for Th17 subset) and percentage of myeloperoxidase (MPO) activities were explored in the groups. RESULTS Administration of mMSC and mMSC + RJ improved the sticky test times and decreased the MPO activities. Using mMSCs and RJ was associated with increased expression of T-bet and GATA3 transcription factors. Transplantation of mMSCs in combination with RJ reduced expression of T-bet in the infarcted tissue. CONCLUSION Using mMSC may be associated with Th1-related inflammation in the long term. RJ co-administration significantly reduced the risks, hence, to decrease the plausible side effects of MSCs, it can be proposed to use RJ in combination with MSC to reduce stroke complications.
Collapse
|
5
|
Kangisser L, Tan E, Bellomo R, Deane AM, Plummer MP. Neuroprotective Properties of Vitamin C: A Scoping Review of Pre-Clinical and Clinical Studies. J Neurotrauma 2021; 38:2194-2205. [PMID: 33544035 DOI: 10.1089/neu.2020.7443] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
There is a need for novel neuroprotective therapies. We aimed to review the evidence for exogenous vitamin C as a neuroprotective agent. MEDLINE, Embase, and Cochrane library databases were searched from inception to May 2020. Pre-clinical and clinical reports evaluating vitamin C for acute neurological injury were included. Twenty-two pre-clinical and 11 clinical studies were eligible for inclusion. Pre-clinical studies included models of traumatic and hypoxic brain injury, subarachnoid and intracerebral hemorrhage, and ischemic stroke. The median [IQR] maximum daily dose of vitamin C in animal studies was 120 [50-500] mg/kg. Twenty-one animal studies reported improvements in biomarkers, functional outcome, or both. Clinical studies included single reports in neonatal hypoxic encephalopathy, traumatic brain injury, and subarachnoid hemorrhage and eight studies in ischemic stroke. The median maximum daily dose of vitamin C was 750 [500-1000] mg, or ∼10 mg/kg for an average-size adult male. Apart from one case series of intracisternal vitamin C administration in subarachnoid hemorrhage, clinical studies reported no patient-centered benefit. Although pre-clinical trials suggest that exogenous vitamin C improves biomarkers of neuroprotection, functional outcome, and mortality, these results have not translated to humans. However, clinical trials used approximately one tenth of the vitamin C dose of animal studies.
Collapse
Affiliation(s)
- Lauren Kangisser
- Department of Intensive Care, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Elinor Tan
- Department of Intensive Care, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Rinaldo Bellomo
- Department of Intensive Care, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Centre for Integrated Critical Care, Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Adam M Deane
- Department of Intensive Care, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Centre for Integrated Critical Care, Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark P Plummer
- Department of Intensive Care, Royal Melbourne Hospital, Parkville, Victoria, Australia
- Centre for Integrated Critical Care, Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Zhang N, Zhao W, Hu ZJ, Ge SM, Huo Y, Liu LX, Gao BL. Protective effects and mechanisms of high-dose vitamin C on sepsis-associated cognitive impairment in rats. Sci Rep 2021; 11:14511. [PMID: 34267240 PMCID: PMC8282649 DOI: 10.1038/s41598-021-93861-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/30/2021] [Indexed: 01/29/2023] Open
Abstract
Sepsis survivors present long-term cognitive deficits. The present study was to investigate the effect of early administration of high-dose vitamin C on cognitive function in septic rats and explore its possible cerebral protective mechanism. Rat sepsis models were established by cecal ligation and puncture (CLP). Ten days after surgery, the Morris water maze test was performed to evaluate the behavior and cognitive function. Histopathologic changes in the hippocampus were evaluated by nissl staining. The inflammatory cytokines, activities of antioxidant enzymes (superoxide dismutase or SOD) and oxidative products (malondialdehyde or MDA) in the serum and hippocampus were tested 24 h after surgery. The activity of matrix metalloproteinase-9 (MMP-9) and expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1(HO-1) in the hippocampus were measured 24 h after surgery. Compared with the sham group in the Morris water maze test, the escape latency of sepsis rats was significantly (P = 0.001) prolonged in the navigation test, whereas the frequency to cross the platform and the time spent in the target quadrant were significantly (P = 0.003) reduced. High-dose vitamin C significantly decreased the escape latency (P = 0.01), but increased the time spent in the target quadrant (P = 0.04) and the frequency to cross the platform (P = 0.19). In the CLP+ saline group, the pyramidal neurons were reduced and distributed sparsely and disorderly, the levels of inflammatory cytokines of tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10 in the serum and hippocampus were significantly increased (P = 0.000), the blood brain barrier (BBB) permeability in the hippocampus was significantly (P = 0.000) increased, the activities of SOD in the serum and hippocampus were significantly (P = 0.000 and P = 0.03, respectively) diminished while the levels of MDA in the serum and hippocampus were significantly (P = 0.007) increased. High-dose vitamin C mitigated hippocampus histopathologic changes, reduced systemic inflammation and neuroinflammation, attenuated BBB disruption, inhibited oxidative stress in brain tissue, and up-regulated the expression of nuclear and total Nrf2 and HO-1. High-dose vitamin C significantly (P < 0.05) decreased the levels of tumor necrosis factor- (TNF)-α, interleukin-6 (IL-6), MDA in the serum and hippocampus, and the activity of MMP-9 in the hippocampus, but significantly (P < 0.05) increased the levels of SOD, the anti-inflammatory cytokine (IL-10) in the serum and hippocampus, and nuclear and total Nrf2, and HO-1 in the hippocampus. In conclusion, high-dose vitamin C can improve cognition impairment in septic rats, and the possible protective mechanism may be related to inhibition of inflammatory factors, alleviation of oxidative stress, and activation of the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Wei Zhao
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Zhen-Jie Hu
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Sheng-Mei Ge
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Yan Huo
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| | - Li-Xia Liu
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China.
| | - Bu-Lang Gao
- Department of Critical Care Medicine, The Fourth Hospital of Hebei Medical University, 12 Jiankang Road, Shijiazhuang, 050011, Hebei, China
| |
Collapse
|
7
|
Shabanizadeh A, Rahmani MR, Yousefi-Ahmadipour A, Asadi F, Arababadi MK. Mesenchymal Stem Cells: The Potential Therapeutic Cell Therapy to Reduce Brain Stroke Side Effects. J Stroke Cerebrovasc Dis 2021; 30:105668. [PMID: 33631477 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105668] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/27/2021] [Accepted: 02/05/2021] [Indexed: 01/06/2023] Open
Abstract
Tissue plasminogen activator (tPA) is the gold standard treatment for ischemic stroke in the time window of 3-4.5 hours after the onset of symptoms. However, tPA administration is associated with inflammation and neurotoxic effects. Mesenchymal stem cells (MSC)-based therapy is emerging as a promising therapeutic strategy to control different inflammatory conditions. This project was designed to examine the protective role of MSC administration alone or in combination with royal jelly (RJ) five hours after stroke onset. The mice model of middle cerebral artery occlusion (MCAO) was established and put to six groups, including intact (healthy mice without stroke), control (untreated stroke), treated with mouse MSC (mMSC), Sup (conditioned medium), RJ and combination of mMSC and RJ (mMSC/RJ). Thereafter, behavioral functions, serum and brain (in both infarcted and non-infarcted tissues) levels of interleukin (IL)-1β, IL-4, IL-10, tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) the sizes of brain infarction have been determined in the groups. Administration of mMSC and mMSC/RJ significantly improved the behavioral functions when compared to the controls. mMSC, RJ and mMSC/RJ significantly decreased the infarcted volumes. RJ and mMSC/RJ, but not mMSC, significantly decreased the brain edema. The infarction increased the serum levels of the cytokines, except TNF-α, and treatment with mMSC, Sup and RJ reduced serum levels of the pro-inflammatory cytokines. mMSC reduced IL-1β in the non-infarcted brain tissue. To conclude, data revealed that using mMSC/RJ combination significantly reduced stroke side effects, including brain edema and serum levels of pro-inflammatory cytokines, and suggested that combination therapy of MSCs with RJ may be considered as an effective stroke therapeutic strategy.
Collapse
|
8
|
Chelluboina B, Vemuganti R. Therapeutic potential of nutraceuticals to protect brain after stroke. Neurochem Int 2020; 142:104908. [PMID: 33220386 DOI: 10.1016/j.neuint.2020.104908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Stroke leads to significant neuronal death and long-term neurological disability due to synergistic pathogenic mechanisms. Stroke induces a change in eating habits and in many cases, leads to undernutrition that aggravates the post-stroke pathology. Proper nutritional regimen remains a major strategy to control the modifiable risk factors for cardiovascular and cerebrovascular diseases including stroke. Studies indicate that nutraceuticals (isolated and concentrated form of high-potency natural bioactive substances present in dietary nutritional components) can act as prophylactic as well as adjuvant therapeutic agents to prevent stroke risk, to promote ischemic tolerance and to reduce post-stroke consequences. Nutraceuticals are also thought to regulate blood pressure, delay neurodegeneration and improve overall vascular health. Nutraceuticals potentially mediate these effects by their powerful antioxidant and anti-inflammatory properties. This review discusses the studies that have highlighted the translational potential of nutraceuticals as stroke therapies.
Collapse
Affiliation(s)
- Bharath Chelluboina
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton Veterans Administration Hospital, Madison, WI, USA.
| |
Collapse
|
9
|
Early Electroacupuncture Extends the rtPA Time Window to 6 h in a Male Rat Model of Embolic Stroke via the ERK1/2-MMP9 Pathway. Neural Plast 2020. [DOI: 10.1155/2020/8851089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background. Recombinant tissue plasminogen activator (rtPA) is the only recommended pharmacological treatment for acute ischemic stroke, but it has a restricted therapeutic time window. When administered at time points greater than 4.5 h after stroke onset, rtPA disrupts the blood-brain barrier (BBB), which leads to serious brain edema and hemorrhagic transformation. Electroacupuncture (EA) exerts a neuroprotective effect on cerebral ischemia; however, researchers have not clearly determined whether EA increases the safety of thrombolysis and extends the therapeutic time window of rtPA administration following ischemic stroke. Objective. The present study was conducted to test the hypothesis that EA extends the therapeutic time window of rtPA for ischemic stroke in a male rat model of embolic stroke. Methods. SD rats were randomly divided into the sham operation group, model group, rtPA group, EA+rtPA group, and rtPA+MEK1/2 inhibitor group. An injection of rtPA was administered 6 h after ischemia. Rats were treated with EA at the Shuigou (GV26) and Neiguan (PC6) acupoints at 2 h after ischemia. Neurological function, infarct volume, BBB permeability, brain edema, and hemorrhagic transformation were assessed at 24 h after ischemia. Western blotting and immunofluorescence staining were performed to detect the levels of proteins involved in the ERK1/2 signaling pathway (MEK1/2 and ERK1/2), tight junction proteins (Claudin5 and ZO-1), and MMP9 in the ischemic penumbra at 24 h after stroke. Results. Delayed rtPA treatment aggravated hemorrhagic transformation and brain edema. However, treatment with EA plus rtPA significantly improved neurological function and reduced the infarct volume, hemorrhagic transformation, brain edema, and EB leakage in rats compared with rtPA alone. EA increased the levels of tight junction proteins, inhibited the activation of the ERK1/2 signaling pathway, and reduced MMP9 overexpression induced by delayed rtPA thrombolysis. Conclusions. EA potentially represents an effective adjunct method to increase the safety of thrombolytic therapy and extend the therapeutic time window of rtPA administration following ischemic stroke. This neuroprotective effect may be mediated by the inhibition of the ERK1/2-MMP9 pathway and alleviation of the destruction of the BBB.
Collapse
|
10
|
The protective effects of prolactin on brain injury. Life Sci 2020; 263:118547. [PMID: 33038380 DOI: 10.1016/j.lfs.2020.118547] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 11/22/2022]
Abstract
AIMS Brain injuries based on their causes are divided into two categories, TBI and NTBI. TBI is caused by damages such as head injury, but non-physical injury causes NTBI. Prolactin is one of the blood factors that increase during brain injury. It has been assumed to play a regenerative role in post-injury recovery. MATERIALS AND METHODS In this review, various valid papers from electronic sources (including Web of Science, Scopus, PubMed, SID, Google Scholar, and ISI databases) used, which in them the protective effect of prolactin on brain injury investigated. KEY FINDINGS Inflammation following brain injury with the production of pro-inflammatory cytokines in the affected area can even lead to excitotoxicity and cell death in the damaged area. Medical brain damage treatments are long-term, and can have several side effects. Therefore, it is better to consider medication treatments that have fewer side effects and greater efficacy. Research suggests that prolactin has numerous regenerative effects on brain injury, and prevents cell death. Prolactin is one of the hormones produced in the body; therefore it has fewer side effects and may be more effective because it increases during brain injury. SIGNIFICANCE Prolactin can be used peripherally and centrally, and exerts its neuro regenerative effects against further damage post-TBI and NTBI.
Collapse
|
11
|
Chang CY, Chen JY, Wu MH, Hu ML. Therapeutic treatment with vitamin C reduces focal cerebral ischemia-induced brain infarction in rats by attenuating disruptions of blood brain barrier and cerebral neuronal apoptosis. Free Radic Biol Med 2020; 155:29-36. [PMID: 32450129 DOI: 10.1016/j.freeradbiomed.2020.05.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/16/2020] [Accepted: 05/16/2020] [Indexed: 01/15/2023]
Abstract
Stroke is a major public health problem and ranks third most common cause of death in adults worldwide. Thrombolysis with recombinant tissue plasminogen activator and endovascular thrombectomy are the main revascularization therapies for acute ischemic stroke. However, ischemia-reperfusion injury, mainly caused by oxidative/nitrosative stress injury, after revascularization therapy can result in worsening outcomes. For better clinical prognosis, more and more studies have focused on the pharmaceutical neuroprotective therapies against free radical damage. The impact of vitamin C (ascorbic acid) on oxidative stress-related diseases is moderate because of its limited oral bioavailability and rapid clearance. However, recent evidence of the clinical benefit of parenteral vitamin C administration has emerged, especially in critical care. In this study we demonstrated that parenteral administration of vitamin C significantly improved neurological deficits and reduced brain infarction and brain edema by attenuating the transient middle cerebral artery occlusion (tMCAO)-induced nitrosative stress, inflammatory responses, and the resultant disruptions of blood brain barrier and cerebral neuronal apoptosis. These results suggest that parenteral administration of vitamin C has potential as an adjuvant agent with intravenous thrombolysis or endovascular thrombectomy in acute treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chia-Yu Chang
- Department of Neurology, Chi Mei Medical Center, Tainan, Taiwan; Center for General Education, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
| | - Jen-Yin Chen
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan; Department of the Senior Citizen Service Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Ming-Hsiu Wu
- Department of Neurology, Liouying Chi Mei Hospital, Tainan, Taiwan; Department of Long-Term Care and Health Promotion, Min-Hwei Junior College of Health Care Management, Taiwan
| | - Miao-Lin Hu
- Department of Food Science and Applied Biotechnology, National Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|
12
|
Sarvari S, Moakedi F, Hone E, Simpkins JW, Ren X. Mechanisms in blood-brain barrier opening and metabolism-challenged cerebrovascular ischemia with emphasis on ischemic stroke. Metab Brain Dis 2020; 35:851-868. [PMID: 32297170 PMCID: PMC7988906 DOI: 10.1007/s11011-020-00573-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Stroke is the leading cause of disability among adults as well as the 2nd leading cause of death globally. Ischemic stroke accounts for about 85% of strokes, and currently, tissue plasminogen activator (tPA), whose therapeutic window is limited to up to 4.5 h for the appropriate population, is the only FDA approved drug in practice and medicine. After a stroke, a cascade of pathophysiological events results in the opening of the blood-brain barrier (BBB) through which further complications, disabilities, and mortality are likely to threaten the patient's health. Strikingly, tPA administration in eligible patients might cause hemorrhagic transformation and sustained damage to BBB integrity. One must, therefore, delineate upon stroke onset which cellular and molecular factors mediate BBB permeability as well as what key roles BBB rupture plays in the pathophysiology of stroke. In this review article, given our past findings of mechanisms underlying BBB opening in stroke animal models, we elucidate cellular, subcellular, and molecular factors involved in BBB permeability after ischemic stroke. The contribution of each factor to stroke severity and outcome is further discussed. Determinant factors in BBB permeability and stroke include mitochondria, miRNAs, matrix metalloproteinases (MMPs), immune cells, cytokines, chemokines, and adhesion proteins. Once these factors are interrogated and their roles in the pathophysiology of stroke are determined, novel targets for drug discovery and development can be uncovered in addition to novel therapeutic avenues for human stroke management.
Collapse
Affiliation(s)
- Sajad Sarvari
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Faezeh Moakedi
- Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Emily Hone
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA
| | - James W Simpkins
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Experimental Stroke Core Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Xuefang Ren
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA.
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA.
- Experimental Stroke Core Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
| |
Collapse
|
13
|
Whole body hypothermia extends tissue plasminogen activator treatment window in the rat model of embolic stroke. Life Sci 2020; 256:117450. [PMID: 32087233 DOI: 10.1016/j.lfs.2020.117450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 12/12/2022]
Abstract
Late treatment with tissue plasminogen activator (tPA) leads to reperfusion injury and poor outcome in ischemic stroke. We have recently shown the beneficial effects of local brain hypothermia after late thrombolysis. Herein, we investigated whether transient whole-body hypothermia was neuroprotective and could prevent the side effects of late tPA therapy at 5.5 h after embolic stroke. After induction of stroke, male rats were randomly assigned into four groups: Control, Hypothermia, tPA and Hypothermia+tPA. Hypothermia started at 5 h after embolic stroke and continued for 1 h. Thirty min after hypothermia, tPA was administrated. Infarct volume, brain edema, blood-brain barrier (BBB) and matrix metalloproteinase-9 (MMP-9) were assessed 48 h and neurological functions were assessed 24 and 48 hour post-stroke. Compared with the control or tPA groups, whole-body hypothermia decreased infarct volume (P < 0.01), BBB disruption (P < 0.05) and MMP-9 level (P < 0.05). However, compared with hypothermia alone a combination of hypothermia and tPA was more effective in reducing infarct volume. While hypothermia alone did not show any effect, its combination with tPA reduced brain edema (P < 0.05). Hypothermia alone or when combined with tPA decreased MMP-9 compared with control or tPA groups (P < 0.01). Although delayed tPA therapy exacerbated BBB integrity, general cooling hampered its leakage after late thrombolysis (P < 0.05). Moreover, only combination therapy significantly improved sensorimotor function as well as forelimb muscle strength at 24 or 48 h after stroke (P < 0.01). Transient whole-body hypothermia in combination with delayed thrombolysis therapy shows more neuroprotection and extends therapeutic time window of tPA up to 5.5 h.
Collapse
|
14
|
Bazmandegan G, Shamsizadeh A, FathiNajafi M, Assadollahi Z, Allahtavakoli M, Kamiab Z, Vakilian A, Moghadam-Ahmadi A, Amirteimoury M, Boroushaki MT. Iranian brown propolis possesses neuroprotective effect against ischemic neuronal damage in mice. JOURNAL OF HERBMED PHARMACOLOGY 2020. [DOI: 10.34172/jhp.2020.16] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Introduction: Stroke is one of the leading causes of death and disability worldwide. Propolis, a polyphenol-rich resinous product processed by honeybees from a variety of plant sources, has a set of biological activities. We investigated the neuroprotective effect of Iranian brown propolis (IBP) in a mouse model of permanent middle cerebral artery occlusion (MCAO). Methods: Experimentally, water extracts of propolis (WEPs) were obtained from Kerman (KeWEP) and Khorasan Razavi (KhWEP) provinces, Iran. The chemical characterization and total polyphenol content of WEPs were determined using the Folin–Ciocalteu assay and gas chromatography-mass spectrometry (GC-MS). Animals were divided into eight experimental groups including: sham, control, and three groups each of which KeWEP- and KhWEP-treated mice. The drugs were administered at doses of 30, 100 and 200 mg/kg, intraperitoneally (IP), during four different time points. Infarct volume and brain edema were measured at 48 h. Behavioral tests were evaluated at 4, 24 and 48-hour post stroke. Results: The total polyphenol content was 1100 and 1400 mg/L in KhWEP and KeWEP respectively. Compared to the control group, the doses of 100 and 200 mg/kg in both samples decreased infarct volume. Brain edema was also reduced in all treatment groups. The dose of 200 mg/kg in both samples and 100 mg/kg in the KeWEP-treated group significantly increased grasping ability. Sensory-motor function was improved in all groups, too. Conclusion: These results suggest that IBP may reduce ischemic brain injury by its neuroprotective effect on focal cerebral ischemia.
Collapse
Affiliation(s)
- Gholamreza Bazmandegan
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Clinical Research Development Unit, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Shamsizadeh
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohsen FathiNajafi
- Razi Vaccine and Serum Research Institute, Mashhad Branch, Agricultural Research, Education and Extension Organization (AREEO), Mashhad, Iran
| | - Zahra Assadollahi
- Department of Epidemiology and Biostatistics, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Kamiab
- Clinical Research Development Unit, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Family Medicine, Ali-Ibn Abi-Talib Hospital , School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Alireza Vakilian
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Neurology, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Amir Moghadam-Ahmadi
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Neurology, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Morteza Amirteimoury
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Taher Boroushaki
- Pharmacological Research Center of Medicinal Plants, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Leichtle SW, Sarma AK, Strein M, Yajnik V, Rivet D, Sima A, Brophy GM. High-Dose Intravenous Ascorbic Acid: Ready for Prime Time in Traumatic Brain Injury? Neurocrit Care 2020; 32:333-339. [PMID: 31440996 DOI: 10.1007/s12028-019-00829-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading public health problems in the USA and worldwide. It is the number one cause of death and disability in children and adults between ages 1-44. Despite efforts to prevent TBIs, the incidence continues to rise. Secondary brain injury occurs in the first hours and days after the initial impact and is the most effective target for intervention. Inflammatory processes and oxidative stress play an important role in the pathomechanism of TBI and are exacerbated by impaired endogenous defense mechanisms, including depletion of antioxidants. As a reducing agent, free radical scavenger, and co-factor in numerous biosynthetic reactions, ascorbic acid (AA, vitamin C) is an essential nutrient that rapidly becomes depleted in states of critical illness. The administration of high-dose intravenous (IV) AA has demonstrated benefits in numerous preclinical models in the areas of trauma, critical care, wound healing, and hematology. A safe and inexpensive treatment, high-dose IV AA administration gained recent attention in studies demonstrating an associated mortality reduction in septic shock patients. High-quality data on the effects of high-dose IV AA on TBI are lacking. Historic data in a small number of patients demonstrate acute and profound AA deficiency in patients with central nervous system pathology, particularly TBI, and a strong correlation between low AA concentrations and poor outcomes. While replenishing deficient AA stores in TBI patients should improve the brain's ability to tolerate oxidative stress, high-dose IV AA may prove an effective strategy to prevent or mitigate secondary brain injury due to its ability to impede lipid peroxidation, scavenge reactive oxygen species, suppress inflammatory mediators, stabilize the endothelium, and reduce brain edema. The existing preclinical data and limited clinical data suggest that high-dose IV AA may be effective in lowering oxidative stress and decreasing cerebral edema. Whether this translates into improved clinical outcomes will depend on identifying the ideal target patient population and possible treatment combinations, factors that need to be evaluated in future clinical studies. With its excellent safety profile and low cost, high-dose IV AA is ready to be evaluated in the early treatment of TBI patients to mitigate secondary brain injury and improve outcomes.
Collapse
Affiliation(s)
- Stefan W Leichtle
- Division of Acute Care Surgical Services, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, USA.
| | - Anand K Sarma
- Department of Neurology, Wake Forest School of Medicine, Winston-Salem, USA
| | - Micheal Strein
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University School of Pharmacy, Richmond, USA
| | - Vishal Yajnik
- Division of Critical Care, Department of Anesthesiology, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Dennis Rivet
- Department of Neurosurgery, Virginia Commonwealth University School of Medicine, Richmond, USA
| | - Adam Sima
- Department of Biostatistics, Virginia Commonwealth University, Richmond, USA
| | - Gretchen M Brophy
- Department of Pharmacotherapy and Outcomes Sciences, Virginia Commonwealth University School of Pharmacy, Richmond, USA
- Department of Neurosurgery, Virginia Commonwealth University School of Medicine, Richmond, USA
| |
Collapse
|
16
|
Morris-Blanco KC, Kim T, Lopez MS, Bertogliat MJ, Chelluboina B, Vemuganti R. Induction of DNA Hydroxymethylation Protects the Brain After Stroke. Stroke 2019; 50:2513-2521. [PMID: 31327315 DOI: 10.1161/strokeaha.119.025665] [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: 12/11/2022]
Abstract
Background and Purpose- Epigenetics play a significant role in brain pathologies. We currently evaluated the role of a recently discovered brain-enriched epigenetic modification known as 5-hydroxymethylcytosine (5hmC) in regulating transcriptomic and pathogenic mechanisms after focal ischemic injury. Methods- Young and aged male and female mice were subjected to transient middle cerebral artery occlusion, and the peri-infarct region was analyzed at various times of reperfusion. Two days before middle cerebral artery occlusion, short-interfering RNA against an isoform of the 5hmC producing enzyme TET (ten-eleven translocase) was injected intracerebrally. Ascorbate was injected intraperitoneally at 5 minutes, 30 minutes, or 2 hours of reperfusion. Motor function was tested with rotarod and beam-walk test. Results- Focal ischemia rapidly induced the activity of TET, the enzyme that catalyzes the formation of 5hmC and preferentially increased expression of the TET3 isoform in the peri-infarct region of the ischemic cortex. Levels of 5hmC were increased in a TET3-dependent manner, and inhibition of TET3 led to wide-scale reductions in the postischemic expression of neuroprotective genes involved in antioxidant defense and DNA repair. TET3 knockdown in adult male and female mice further increased brain degeneration after focal ischemia, demonstrating a role for TET3 and 5hmC in endogenous protection against stroke. Ascorbate treatment after focal ischemia enhanced TET3 activity and 5hmC enrichment in the peri-infarct region. TET3 activation by ascorbate provided robust protection against ischemic injury in young and aged mice of both sexes. Moreover, ascorbate treatment improved motor function recovery in both male and female mice. Conclusions- Collectively, these results indicate the potential of TET3 and 5hmC as novel stroke therapeutic targets. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Kahlilia C Morris-Blanco
- From the Department of Neurological Surgery (K.C.M.-B., T.K., M.S.L., M.J.B., B.C., R.V.), University of Wisconsin-Madison, Madison, WI.,William S. Middleton Veterans Administration Hospital, Madison, WI, (K.C.M.-B., T.K., R.V.)
| | - TaeHee Kim
- From the Department of Neurological Surgery (K.C.M.-B., T.K., M.S.L., M.J.B., B.C., R.V.), University of Wisconsin-Madison, Madison, WI.,William S. Middleton Veterans Administration Hospital, Madison, WI, (K.C.M.-B., T.K., R.V.)
| | - Mary S Lopez
- From the Department of Neurological Surgery (K.C.M.-B., T.K., M.S.L., M.J.B., B.C., R.V.), University of Wisconsin-Madison, Madison, WI.,Cellular and Molecular Pathology Training Program (M.S.L., R.V.), University of Wisconsin-Madison, Madison, WI
| | - Mario J Bertogliat
- From the Department of Neurological Surgery (K.C.M.-B., T.K., M.S.L., M.J.B., B.C., R.V.), University of Wisconsin-Madison, Madison, WI
| | - Bharath Chelluboina
- From the Department of Neurological Surgery (K.C.M.-B., T.K., M.S.L., M.J.B., B.C., R.V.), University of Wisconsin-Madison, Madison, WI
| | - Raghu Vemuganti
- From the Department of Neurological Surgery (K.C.M.-B., T.K., M.S.L., M.J.B., B.C., R.V.), University of Wisconsin-Madison, Madison, WI.,Cellular and Molecular Pathology Training Program (M.S.L., R.V.), University of Wisconsin-Madison, Madison, WI.,William S. Middleton Veterans Administration Hospital, Madison, WI, (K.C.M.-B., T.K., R.V.)
| |
Collapse
|
17
|
Ascorbic acid attenuates cognitive impairment and brain oxidative stress in ovariectomized mice. Pharmacol Rep 2019; 71:133-138. [DOI: 10.1016/j.pharep.2018.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/15/2018] [Accepted: 10/02/2018] [Indexed: 12/14/2022]
|
18
|
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: 8.2] [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]
|
19
|
Kuan CY, Lin YY, Chen CY, Yang CC, Chi CY, Li CH, Dong GC, Lin FH. The preparation of oxidized methylcellulose crosslinked by adipic acid dihydrazide loaded with vitamin C for traumatic brain injury. J Mater Chem B 2019. [DOI: 10.1039/c9tb00816k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oxi-MC-ADH-VC can open up a new avenue for clinical TBI treatment and rehabilitation.
Collapse
Affiliation(s)
- Che-Yung Kuan
- PhD Program in Tissue Engineering and Regenerative Medicine
- National Chung Hsing University
- Taiwan
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
| | - Yu-Ying Lin
- PhD Program in Tissue Engineering and Regenerative Medicine
- National Chung Hsing University
- Taiwan
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
| | - Ching-Yun Chen
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
- Taiwan
| | - Chun-Chen Yang
- Institute of Biomedical Engineering
- College of Medicine and College of Engineering
- National Taiwan University
- Taipei
- Taiwan
| | - Chih-Ying Chi
- PhD Program in Tissue Engineering and Regenerative Medicine
- National Chung Hsing University
- Taiwan
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
| | - Chi-Han Li
- PhD Program in Tissue Engineering and Regenerative Medicine
- National Chung Hsing University
- Taiwan
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
| | - Guo-Chung Dong
- PhD Program in Tissue Engineering and Regenerative Medicine
- National Chung Hsing University
- Taiwan
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
| | - Feng-Huei Lin
- PhD Program in Tissue Engineering and Regenerative Medicine
- National Chung Hsing University
- Taiwan
- Institute of Biomedical Engineering and Nanomedicine
- National Health Research Institutes
| |
Collapse
|
20
|
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: 5.1] [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
|
21
|
Monoacylglycerol Lipase Inhibitor is Safe when Combined with Delayed r-tPA Administration in Treatment of Stroke. Inflammation 2018; 41:2052-2059. [DOI: 10.1007/s10753-018-0848-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Rahmani MR, Shamsizadeh A, Moghadam-Ahmadi A, Bazmandegan G, Allahtavakoli M. JZL184, as a monoacylglycerol lipase inhibitor, down-regulates inflammation in a cannabinoid pathway dependent manner. Biomed Pharmacother 2018; 103:1720-1726. [DOI: 10.1016/j.biopha.2018.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023] Open
|
23
|
Shekhar S, Cunningham MW, Pabbidi MR, Wang S, Booz GW, Fan F. Targeting vascular inflammation in ischemic stroke: Recent developments on novel immunomodulatory approaches. Eur J Pharmacol 2018; 833:531-544. [PMID: 29935175 DOI: 10.1016/j.ejphar.2018.06.028] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/19/2018] [Indexed: 10/28/2022]
Abstract
Ischemic stroke is a devastating and debilitating medical condition with limited therapeutic options. However, accumulating evidence indicates a central role of inflammation in all aspects of stroke including its initiation, the progression of injury, and recovery or wound healing. A central target of inflammation is disruption of the blood brain barrier or neurovascular unit. Here we discuss recent developments in identifying potential molecular targets and immunomodulatory approaches to preserve or protect barrier function and limit infarct damage and functional impairment. These include blocking harmful inflammatory signaling in endothelial cells, microglia/macrophages, or Th17/γδ T cells with biologics, third generation epoxyeicosatrienoic acid (EET) analogs with extended half-life, and miRNA antagomirs. Complementary beneficial pathways may be enhanced by miRNA mimetics or hyperbaric oxygenation. These immunomodulatory approaches could be used to greatly expand the therapeutic window for thrombolytic treatment with tissue plasminogen activator (t-PA). Moreover, nanoparticle technology allows for the selective targeting of endothelial cells for delivery of DNA/RNA oligonucleotides and neuroprotective drugs. In addition, although likely detrimental to the progression of ischemic stroke by inducing inflammation, oxidative stress, and neuronal cell death, 20-HETE may also reduce susceptibility of onset of ischemic stroke by maintaining autoregulation of cerebral blood flow. Although the interaction between inflammation and stroke is multifaceted, a better understanding of the mechanisms behind the pro-inflammatory state at all stages will hopefully help in developing novel immunomodulatory approaches to improve mortality and functional outcome of those inflicted with ischemic stroke.
Collapse
Affiliation(s)
- Shashank Shekhar
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS, USA; Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Mark W Cunningham
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Shaoxun Wang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
24
|
NLRP3 Inflammasome Is Involved in Q-VD-OPH Induced Necroptosis Following Cerebral Ischemia-Reperfusion Injury. Neurochem Res 2018; 43:1200-1209. [DOI: 10.1007/s11064-018-2537-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 03/16/2018] [Accepted: 04/19/2018] [Indexed: 02/02/2023]
|
25
|
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: 3.5] [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
|
26
|
Transient brain hypothermia reduces the reperfusion injury of delayed tissue plasminogen activator and extends its therapeutic time window in a focal embolic stroke model. Brain Res Bull 2017; 134:85-90. [DOI: 10.1016/j.brainresbull.2017.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/02/2017] [Accepted: 07/07/2017] [Indexed: 11/18/2022]
|
27
|
Vitamin C, Aging and Alzheimer's Disease. Nutrients 2017; 9:nu9070670. [PMID: 28654021 PMCID: PMC5537785 DOI: 10.3390/nu9070670] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence in mice models of accelerated senescence indicates a rescuing role of ascorbic acid in premature aging. Supplementation of ascorbic acid appeared to halt cell growth, oxidative stress, telomere attrition, disorganization of chromatin, and excessive secretion of inflammatory factors, and extend lifespan. Interestingly, ascorbic acid (AA) was also found to positively modulate inflamm-aging and immunosenescence, two hallmarks of biological aging. Moreover, ascorbic acid has been shown to epigenetically regulate genome integrity and stability, indicating a key role of targeted nutrition in healthy aging. Growing in vivo evidence supports the role of ascorbic acid in ameliorating factors linked to Alzheimer’s disease (AD) pathogenesis, although evidence in humans yielded equivocal results. The neuroprotective role of ascorbic acid not only relies on the general free radical trapping, but also on the suppression of pro-inflammatory genes, mitigating neuroinflammation, on the chelation of iron, copper, and zinc, and on the suppression of amyloid-beta peptide (Aβ) fibrillogenesis. Epidemiological evidence linking diet, one of the most important modifiable lifestyle factors, and risk of Alzheimer's disease is rapidly increasing. Thus, dietary interventions, as a way to epigenetically modulate the human genome, may play a role in the prevention of AD. The present review is aimed at providing an up to date overview of the main biological mechanisms that are associated with ascorbic acid supplementation/bioavailability in the process of aging and Alzheimer’s disease. In addition, we will address new fields of research and future directions.
Collapse
|
28
|
Kocot J, Luchowska-Kocot D, Kiełczykowska M, Musik I, Kurzepa J. Does Vitamin C Influence Neurodegenerative Diseases and Psychiatric Disorders? Nutrients 2017; 9:E659. [PMID: 28654017 PMCID: PMC5537779 DOI: 10.3390/nu9070659] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/15/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023] Open
Abstract
Vitamin C (Vit C) is considered to be a vital antioxidant molecule in the brain. Intracellular Vit C helps maintain integrity and function of several processes in the central nervous system (CNS), including neuronal maturation and differentiation, myelin formation, synthesis of catecholamine, modulation of neurotransmission and antioxidant protection. The importance of Vit C for CNS function has been proven by the fact that targeted deletion of the sodium-vitamin C co-transporter in mice results in widespread cerebral hemorrhage and death on post-natal day one. Since neurological diseases are characterized by increased free radical generation and the highest concentrations of Vit C in the body are found in the brain and neuroendocrine tissues, it is suggested that Vit C may change the course of neurological diseases and display potential therapeutic roles. The aim of this review is to update the current state of knowledge of the role of vitamin C on neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis and amyotrophic sclerosis, as well as psychiatric disorders including depression, anxiety and schizophrenia. The particular attention is attributed to understanding of the mechanisms underlying possible therapeutic properties of ascorbic acid in the presented disorders.
Collapse
Affiliation(s)
- Joanna Kocot
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Dorota Luchowska-Kocot
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Małgorzata Kiełczykowska
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Irena Musik
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| | - Jacek Kurzepa
- Chair and Department of Medical Chemistry, Medical University of Lublin, 4A Chodźki Street, 20-093 Lublin, Poland.
| |
Collapse
|
29
|
Brzica H, Abdullahi W, Ibbotson K, Ronaldson PT. Role of Transporters in Central Nervous System Drug Delivery and Blood-Brain Barrier Protection: Relevance to Treatment of Stroke. J Cent Nerv Syst Dis 2017; 9:1179573517693802. [PMID: 28469523 PMCID: PMC5392046 DOI: 10.1177/1179573517693802] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/22/2017] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality in the United States. The only approved pharmacologic treatment for ischemic stroke is thrombolysis via recombinant tissue plasminogen activator (r-tPA). A short therapeutic window and serious adverse events (ie, hemorrhage, excitotoxicity) greatly limit r-tPA therapy, which indicates an essential need to develop novel stroke treatment paradigms. Transporters expressed at the blood-brain barrier (BBB) provide a significant opportunity to advance stroke therapy via central nervous system delivery of drugs that have neuroprotective properties. Examples of such transporters include organic anion–transporting polypeptides (Oatps) and organic cation transporters (Octs). In addition, multidrug resistance proteins (Mrps) are transporter targets in brain microvascular endothelial cells that can be exploited to preserve BBB integrity in the setting of stroke. Here, we review current knowledge on stroke pharmacotherapy and demonstrate how endogenous BBB transporters can be targeted for improvement of ischemic stroke treatment.
Collapse
Affiliation(s)
- Hrvoje Brzica
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Kathryn Ibbotson
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
30
|
Peña ID, Borlongan C, Shen G, Davis W. Strategies to Extend Thrombolytic Time Window for Ischemic Stroke Treatment: An Unmet Clinical Need. J Stroke 2017; 19:50-60. [PMID: 28178410 PMCID: PMC5307939 DOI: 10.5853/jos.2016.01515] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/13/2022] Open
Abstract
To date, reperfusion with tissue plasminogen activator (tPA) remains the gold standard treatment for ischemic stroke. However, when tPA is given beyond 4.5 hours of stroke onset, deleterious effects of the drug ensue, especially, hemorrhagic transformation (HT), which causes the most significant morbidity and mortality in stroke patients. An important clinical problem at hand is to develop strategies that will enhance the therapeutic time window for tPA therapy and reduce the adverse effects (especially HT) of delayed tPA treatment. We reviewed the pharmacological agents which reduced the risk of HT associated with delayed (beyond 4.5 hours post-stroke) tPA treatment in preclinical studies, which we classified into those that putatively preserve the blood-brain barrier (e.g., minocycline, cilostazol, fasudil, candesartan, and bryostatin) and/or enhance vascularization and protect the cerebrovasculature (e.g., coumarin derivate IMM-H004 and granulocyte colony-stimulating factor). Recently, other new therapeutic modalities (e.g., oxygen transporters) have been reported which improved delayed tPA-associated outcomes by acting through other mechanisms. While the above-mentioned interventions unequivocally reduced delayed tPA-induced HT in stroke models, the long-term efficacy of these drugs are not yet established. Further optimization is required to expedite their future clinical application. The findings from this review indicate the need to explore the most ideal adjunctive interventions that will not only reduce delayed tPA–induced HT, but also preserve neurovascular functions. While waiting for the next breakthrough drug in acute stroke treatment, it is equally important to allocate considerable effort to find approaches to address the limitations of the only FDA-approved stroke therapy.
Collapse
Affiliation(s)
- Ike Dela Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, United States
| | - Cesar Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, United States
| | - Guofang Shen
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, United States
| | - Willie Davis
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University School of Pharmacy, Loma Linda, United States
| |
Collapse
|
31
|
Lam V, Hackett M, Takechi R. Antioxidants and Dementia Risk: Consideration through a Cerebrovascular Perspective. Nutrients 2016; 8:nu8120828. [PMID: 27999412 PMCID: PMC5188481 DOI: 10.3390/nu8120828] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/16/2016] [Accepted: 12/16/2016] [Indexed: 12/16/2022] Open
Abstract
A number of natural and chemical compounds that exert anti-oxidative properties are demonstrated to be beneficial for brain and cognitive function, and some are reported to reduce the risk of dementia. However, the detailed mechanisms by which those anti-oxidative compounds show positive effects on cognition and dementia are still unclear. An emerging body of evidence suggests that the integrity of the cerebrovascular blood-brain barrier (BBB) is centrally involved in the onset and progression of cognitive impairment and dementia. While recent studies revealed that some anti-oxidative agents appear to be protective against the disruption of BBB integrity and structure, few studies considered the neuroprotective effects of antioxidants in the context of cerebrovascular integrity. Therefore, in this review, we examine the mechanistic insights of antioxidants as a pleiotropic agent for cognitive impairment and dementia through a cerebrovascular axis by primarily focusing on the current available data from physiological studies. Conclusively, there is a compelling body of evidence that suggest antioxidants may prevent cognitive decline and dementia by protecting the integrity and function of BBB and, indeed, further studies are needed to directly examine these effects in addition to underlying molecular mechanisms.
Collapse
Affiliation(s)
- Virginie Lam
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth WA 6845, Australia.
| | - Mark Hackett
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- Department of Chemistry, Faculty of Science and Engineering, Curtin University, Perth WA 6845, Australia.
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth WA 6845, Australia.
| |
Collapse
|
32
|
Brown propolis attenuates cerebral ischemia-induced oxidative damage via affecting antioxidant enzyme system in mice. Biomed Pharmacother 2016; 85:503-510. [PMID: 27889229 DOI: 10.1016/j.biopha.2016.11.057] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/28/2016] [Accepted: 11/14/2016] [Indexed: 11/22/2022] Open
Abstract
Oxidative stress plays a critical role in ischemic brain injury. Superoxide dismutase (SOD) and glutathione peroxidase (GPx) are the enzymes underlying the endogenous antioxidant mechanisms affected by stroke and are considered as oxidative stress biomarkers. Brown propolis (BP) is a bioactive natural product with a set of biological activities that in turn may differ depending on the area from which the substance is originated. The aim of this study was to investigate the effect of water-extracted brown propolis (WEBPs), from two regions of Iran, against cerebral ischemia-induced oxidative injury in a mouse model of stroke. Experimentally, the chemical characterization and total polyphenol content were determined using GC/MS and Folin-Ciocalteu assay respectively. Seventy-two adult male mice were randomly divided into the surgical sham group, control group (treated with vehicle), and four groups of WEBPs-treated animals. The WEBPs were administered at the doses of 100 and 200mg/kg IP, during four different time points. Oxidative stress biomarkers (SOD and GPx activity, SOD/GPx ratio), lipid peroxidation (LPO) index (malondialdehyde content) and infarct volume were measured 48h post stroke. Behavioral tests were evaluated 24 and 48h after stroke. WEBPs treatment resulted in significant restoration of antioxidant enzymes activity and a subsequent decrease in LPO as well as the infarct volume compared to the control group. Sensory-motor impairment and neurological deficits were improved significantly as well. These results indicate that Iranian BP confers neuroprotection on the stroke-induced neuronal damage via an antioxidant mechanism which seems to be mediated by the endogenous antioxidant system.
Collapse
|
33
|
Dong MX, Hu QC, Shen P, Pan JX, Wei YD, Liu YY, Ren YF, Liang ZH, Wang HY, Zhao LB, Xie P. Recombinant Tissue Plasminogen Activator Induces Neurological Side Effects Independent on Thrombolysis in Mechanical Animal Models of Focal Cerebral Infarction: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0158848. [PMID: 27387385 PMCID: PMC4936748 DOI: 10.1371/journal.pone.0158848] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 06/22/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Recombinant tissue plasminogen activator (rtPA) is the only effective drug approved by US FDA to treat ischemic stroke, and it contains pleiotropic effects besides thrombolysis. We performed a meta-analysis to clarify effect of tissue plasminogen activator (tPA) on cerebral infarction besides its thrombolysis property in mechanical animal stroke. METHODS Relevant studies were identified by two reviewers after searching online databases, including Pubmed, Embase, and ScienceDirect, from 1979 to 2016. We identified 6, 65, 17, 12, 16, 12 and 13 comparisons reporting effect of endogenous tPA on infarction volume and effects of rtPA on infarction volume, blood-brain barrier, brain edema, intracerebral hemorrhage, neurological function and mortality rate in all 47 included studies. Standardized mean differences for continuous measures and risk ratio for dichotomous measures were calculated to assess the effects of endogenous tPA and rtPA on cerebral infarction in animals. The quality of included studies was assessed using the Stroke Therapy Academic Industry Roundtable score. Subgroup analysis, meta-regression and sensitivity analysis were performed to explore sources of heterogeneity. Funnel plot, Trim and Fill method and Egger's test were obtained to detect publication bias. RESULTS We found that both endogenous tPA and rtPA had not enlarged infarction volume, or deteriorated neurological function. However, rtPA would disrupt blood-brain barrier, aggravate brain edema, induce intracerebral hemorrhage and increase mortality rate. CONCLUSIONS This meta-analysis reveals rtPA can lead to neurological side effects besides thrombolysis in mechanical animal stroke, which may account for clinical exacerbation for stroke patients that do not achieve vascular recanalization with rtPA.
Collapse
Affiliation(s)
- Mei-Xue Dong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Qing-Chuan Hu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Peng Shen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun-Xi Pan
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - You-Dong Wei
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Yun Liu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Fei Ren
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zi-Hong Liang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Li-Bo Zhao
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| |
Collapse
|
34
|
Lee JY, Choi HY, Yune TY. Fluoxetine and vitamin C synergistically inhibits blood-spinal cord barrier disruption and improves functional recovery after spinal cord injury. Neuropharmacology 2016; 109:78-87. [PMID: 27256500 DOI: 10.1016/j.neuropharm.2016.05.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 01/08/2023]
Abstract
Recently we reported that fluoxetine (10 mg/kg) improves functional recovery by attenuating blood spinal cord barrier (BSCB) disruption after spinal cord injury (SCI). Here we investigated whether a low-dose of fluoxetine (1 mg/kg) and vitamin C (100 mg/kg), separately not possessing any protective effect, prevents BSCB disruption and improves functional recovery when combined. After a moderate contusion injury at T9 in rat, a low-dose of fluoxetine and vitamin C, or the combination of both was administered intraperitoneally immediately after SCI and further treated once a day for 14 d. Co-treatment with fluoxetine and vitamin C significantly attenuated BSCB permeability at 1 d after SCI. When only fluoxetine or vitamin C was treated after injury, however, there was no effect on BSCB disruption. Co-treatment with fluoxetine and vitamin C also significantly inhibited the expression and activation of MMP-9 at 8 h and 1 d after injury, respectively, and the infiltration of neutrophils (at 1 d) and macrophages (at 5 d) and the expression of inflammatory mediators (at 2 h, 6 h, 8 h or 24 h after injury) were significantly inhibited by co-treatment with fluoxetine and vitamin C. Furthermore, the combination of fluoxetine and vitamin C attenuated apoptotic cell death at 1 d and 5 d and improved locomotor function at 5 weeks after SCI. These results demonstrate the synergistic effect combination of low-dose fluoxetine and vitamin C on BSCB disruption after SCI and furthermore support the effectiveness of the combination treatment regimen for the management of acute SCI.
Collapse
Affiliation(s)
- Jee Y Lee
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hae Y Choi
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Tae Y Yune
- Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
35
|
Maher EM, Ali AMA, Salem HF, Abdelrahman AA. In vitro/in vivo evaluation of an optimized fast dissolving oral film containing olanzapine co-amorphous dispersion with selected carboxylic acids. Drug Deliv 2016; 23:3088-3100. [DOI: 10.3109/10717544.2016.1153746] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Eman Magdy Maher
- Department of Pharmaceutics, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt,
| | - Ahmed Mahmoud Abdelhaleem Ali
- Department of Pharmaceutics, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt,
- Department of Pharmaceutics, Faculty of Pharmacy, Taif University, Taif, Saudi Arabia, and
| | - Heba Farouk Salem
- Department of Pharmaceutics, Faculty of Pharmacy, Beni Suef University, Beni Suef, Egypt,
| | | |
Collapse
|