1
|
Höfling C, Donkersloot P, Ulrich L, Burghardt S, Opitz M, Geissler S, Schilling S, Cynis H, Michalski D, Roßner S. Dipeptidyl peptidase 4 deficiency improves survival after focal cerebral ischemia in mice and ameliorates microglia activation and specific inflammatory markers. Neurobiol Dis 2024; 201:106671. [PMID: 39293688 DOI: 10.1016/j.nbd.2024.106671] [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: 06/17/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024] Open
Abstract
Dipeptidyl peptidase 4 (DPP4; CD26) is involved in the regulation of various metabolic, immunological, and neurobiological processes in healthy individuals. Observations based on epidemiological data indicate that DPP4 inhibition by gliptins, typically used in patients with diabetes, may reduce the risk for cerebral ischemia and may also improve related outcomes. However, as DPP4 inhibitor application is neither complete nor specific for suppression of DPP4 enzymatic activity and DPP4 has non-enzymatic functions as well, the variety of consequences is a matter of debate. Therefore, we here used DPP4 knock-out (KO) mice to analyze the specific contribution of DPP4 to cellular, immunological, and functional consequences of experimental focal cerebral ischemia. We observed a significantly higher survival rate of DPP4 KO mice after ischemia, which was accompanied by a lower abundance of the pro-inflammatory chemokine CCL2 and reduced activation of Iba1-positive microglia cells in brain tissue of DPP4 KO mice. In addition, after ischemia for 24 h to 72 h, decreased concentrations of CCL5 and CCL12 in plasma and of CCL17 in brain tissue of DPP4 KO mice were observed when compared to wild type mice. Other aspects analyzed, such as the functional Menzies score, astrocyte activation and chemokine levels in plasma and brain tissue were affected by ischemia but appeared to be unaffected by the DPP4 KO genotype. Taken together, experimental ablation of DPP4 functions in mice improves survival and ameliorates aspects of cellular and molecular inflammation after focal cerebral ischemia.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany; Department of Neurology, University of Leipzig, 04103 Leipzig, Germany
| | - Philippa Donkersloot
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Luise Ulrich
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Sina Burghardt
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Michael Opitz
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany
| | - Stefanie Geissler
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, 06120 Halle (Saale), Germany
| | - Stephan Schilling
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, 06120 Halle (Saale), Germany; Anhalt University of Applied Sciences, Faculty of Applied Biosciences and Process Engineering, 06366 Köthen, Germany
| | - Holger Cynis
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, 06120 Halle (Saale), Germany; Junior Research Group "Immunomodulation in Pathophysiological Processes" Faculty of Medicine, Martin Luther University Halle-Wittenberg, Germany
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, 04103 Leipzig, Germany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
2
|
Bindal P, Kumar V, Kapil L, Singh C, Singh A. Therapeutic management of ischemic stroke. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2651-2679. [PMID: 37966570 DOI: 10.1007/s00210-023-02804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023]
Abstract
Stroke is the third leading cause of years lost due to disability and the second-largest cause of mortality worldwide. Most occurrences of stroke are brought on by the sudden occlusion of an artery (ischemic stroke), but sometimes they are brought on by bleeding into brain tissue after a blood vessel has ruptured (hemorrhagic stroke). Alteplase is the only therapy the American Food and Drug Administration has approved for ischemic stroke under the thrombolysis category. Current views as well as relevant clinical research on the diagnosis, assessment, and management of stroke are reviewed to suggest appropriate treatment strategies. We searched PubMed and Google Scholar for the available therapeutic regimes in the past, present, and future. With the advent of endovascular therapy in 2015 and intravenous thrombolysis in 1995, the therapeutic options for ischemic stroke have expanded significantly. A novel approach such as vagus nerve stimulation could be life-changing for many stroke patients. Therapeutic hypothermia, the process of cooling the body or brain to preserve organ integrity, is one of the most potent neuroprotectants in both clinical and preclinical contexts. The rapid intervention has been linked to more favorable clinical results. This study focuses on the pathogenesis of stroke, as well as its recent advancements, future prospects, and potential therapeutic targets in stroke therapy.
Collapse
Affiliation(s)
- Priya Bindal
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Lakshay Kapil
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Chauras Campus, Distt. Tehri Garhwal, Uttarakhand, 246174, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India.
| |
Collapse
|
3
|
Chekol Abebe E, Mengstie MA, Seid MA, Gebeyehu NA, Adella GA, Kassie GA, Gesese MM, Tegegne KD, Anley DT, Feleke SF, Zemene MA, Dessie AM, Tesfa NA, Moges N, Chanie ES, Kebede YS, Bantie B, Dejenie TA. Comparison of circulating lipid profiles, D-dimer and fibrinogen levels between hypertensive patients with and without stroke. Metabol Open 2023; 19:100252. [PMID: 37559716 PMCID: PMC10407734 DOI: 10.1016/j.metop.2023.100252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/13/2023] [Accepted: 07/09/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Stroke is one of the leading causes of global mortality and disability, particularly in hypertensive patients. This study aimed to compare lipid profile, fibrinogen, and D-dimer levels between hypertensive patient with and without stroke. METHODS This was a facility-based cross-sectional study conducted from November 2022 to January 2023 among 115 hypertensive patients (70 patients without stroke and 45 with stroke) who had follow-up at Yikatit 12 Hospital Medical College, Ethiopia. All data analyses were done using SPSS version 25.0 and comparisons of variables between groups were made using the Chi-square test, independent sample t-test, and Mann-Whitney U test. Multiple logistic regression analysis was done to identify predictors of stroke among hypertensive patients. A p-value <0.05 was assumed to be statistically significant for all statistical tests. RESULTS Significantly elevated levels of TC, LDL-C, D-DI, and fibrinogen were observed in the stroke group than in the non-stroke group (p-value<0.05). The mean values of TC, D-DI, and fibrinogen were significantly higher in patients with ischemic stroke compared to those with hemorrhagic stroke. Duration of hypertension (AOR: 1.21; CI: 1.10, 2.09), TC (AOR:1.07; CI: 1.01, 1.22), D-DI (AOR: 1.15; CI: 1.05, 1.69) and fibrinogen (AOR:1.19; CI: 1.10, 2.89) were identified to be independent predictors of stroke in hypertensive patients. CONCLUSION The circulating levels of TC, LDL-C, D-DI and fibrinogen in hypertensive patients with stroke were significantly higher than in those without stroke. But only TC, D-DI, and fibrinogen were found to be predictors of stroke in hypertensives. Considerably higher TC, D-DI, and fibrinogen levels were also seen in patients with ischemic stroke than in those with hemorrhagic stroke. This confirms the key roles of dyslipidemia (hypercholesterolemia) and aberrant hemostatic activation to stroke development, notably ischemic stroke.
Collapse
Affiliation(s)
- Endeshaw Chekol Abebe
- Department of Biochemistry, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Misganaw Asmamaw Mengstie
- Department of Biochemistry, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Mohammed Abdu Seid
- Department of Physiology, College of Health Science, Debre Tabor University, Debre Tabor, Ethiopia
| | - Natnael Atnafu Gebeyehu
- Department of Midwifery, College of Medicine and Health Science, Wolaita Sodo University, Sodo, Ethiopia
| | - Getachew Asmare Adella
- Department of Reproductive Health and Nutrition, School of Public Health, Woliata Sodo University, Sodo, Ethiopia
| | - Gizachew Ambaw Kassie
- Department of Epidemiology and Biostatistics, School of Public Health, Woliata Sodo University, Sodo, Ethiopia
| | - Molalegn Mesele Gesese
- Department of Midwifery, College of Medicine and Health Science, Wolaita Sodo University, Sodo, Ethiopia
| | - Kirubel Dagnaw Tegegne
- Department of Nursing, College of Medicine and Health Science, Wollo University, Dessie, Ethiopia
| | - Denekew Tenaw Anley
- Department of Public Health, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Sefineh Fenta Feleke
- School of Medicine, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Melkamu Aderajew Zemene
- Department of Public Health, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Anteneh Mengist Dessie
- Department of Public Health, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Natnael Amare Tesfa
- School of Medicine, College of Health Sciences, Woldia University, Woldia, Ethiopia
| | - Natnael Moges
- Department of Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Ermias Sisay Chanie
- Department of Pediatrics and Child Health Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Yenealem Solomon Kebede
- Department of Medical Laboratory Science, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Berihun Bantie
- Department of Comprehensive Nursing, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Tadesse Asmamaw Dejenie
- Department of Medical Biochemistry, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
4
|
Höfling C, Roßner S, Flachmeyer B, Krueger M, Härtig W, Michalski D. Tricellulin, α-Catenin and Microfibrillar-Associated Protein 5 Exhibit Concomitantly Altered Immunosignals along with Vascular, Extracellular and Cytoskeletal Elements after Experimental Focal Cerebral Ischemia. Int J Mol Sci 2023; 24:11893. [PMID: 37569268 PMCID: PMC10418498 DOI: 10.3390/ijms241511893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/08/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Along with initiatives to understand the pathophysiology of stroke in detail and to identify neuroprotective targets, cell-stabilizing elements have gained increasing attention. Although cell culture experiments have indicated that tricellulin, α-catenin and microfibrillar-associated protein 5 (MFAP5) contribute to cellular integrity, these elements have not yet been investigated in the ischemic brain. Applying immunofluorescence labeling, this study explored tricellulin, MFAP5 and α-catenin in non-ischemic and ischemic brain areas of mice (24, 4 h of ischemia) and rats (4 h of ischemia), along with collagen IV and fibronectin as vascular and extracellular matrix constituents and microtubule-associated protein 2 (MAP2) and neurofilament light chain (NF-L) as cytoskeletal elements. Immunosignals of tricellulin and notably MFAP5 partially appeared in a fiber-like pattern, and α-catenin appeared more in a dotted pattern. Regional associations with vascular and extracellular constituents were found for tricellulin and α-catenin, particularly in ischemic areas. Due to ischemia, signals of tricellulin, MFAP5 and α-catenin decreased concomitantly with MAP2 and NF-L, whereby MFAP5 provided the most sensitive reaction. For the first time, this study demonstrated ischemia-related alterations in tricellulin, MFAP5 and α-catenin along with the vasculature, extracellular matrix and cytoskeleton. Confirmatory studies are needed, also exploring their role in cellular integrity and the potential for neuroprotective approaches in stroke.
Collapse
Affiliation(s)
- Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstr. 19, 04103 Leipzig, Germany; (C.H.); (S.R.); (W.H.)
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstr. 19, 04103 Leipzig, Germany; (C.H.); (S.R.); (W.H.)
| | - Bianca Flachmeyer
- Institute of Anatomy, University of Leipzig, Liebigstr. 13, 04103 Leipzig, Germany; (B.F.); (M.K.)
| | - Martin Krueger
- Institute of Anatomy, University of Leipzig, Liebigstr. 13, 04103 Leipzig, Germany; (B.F.); (M.K.)
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Liebigstr. 19, 04103 Leipzig, Germany; (C.H.); (S.R.); (W.H.)
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103 Leipzig, Germany
| |
Collapse
|
5
|
Gao X, Xu J, Yao T, Liu X, Zhang H, Zhan C. Peptide-decorated nanocarriers penetrating the blood-brain barrier for imaging and therapy of brain diseases. Adv Drug Deliv Rev 2022; 187:114362. [PMID: 35654215 DOI: 10.1016/j.addr.2022.114362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Blood-Brain Barrier (BBB) is one of the most important physiological barriers strictly restricting the substance exchange between blood and brain tissues. While the BBB protects the brain from infections and toxins and maintains brain homeostasis, it is also recognized as the main obstacle to the penetration of therapeutics and imaging agents into the brain. Due to high specificity and affinity, peptides are frequently exploited to decorate nanocarriers across the BBB for diagnosis and/or therapy purposes. However, there are still some challenges that restrict their clinical application, such as stability, safety and immunocompatibility. In this review, we summarize the biological and pathophysiological characteristics of the BBB, strategies across the BBB, and recent progress on peptide decorated nanocarriers for brain diseases diagnosis and therapy. The challenges and opportunities for their translation are also discussed.
Collapse
|
6
|
Kawadkar M, Mandloi AS, Saxena V, Tamadaddi C, Sahi C, Dhote VV. Noscapine alleviates cerebral damage in ischemia-reperfusion injury in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:669-683. [PMID: 33106921 DOI: 10.1007/s00210-020-02005-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
With high unmet medical needs, stroke remains an intensely focused research area. Although noscapine is a neuroprotective agent, its mechanism of action in ischemic-reperfusion (I-R) injury is yet to be ascertained. We investigated the effect of noscapine on the molecular mechanisms of cell damage using yeast, and its neuroprotection on cerebral I-R injury in rats. Yeast, both wild-type and Δtrx2 strains, was evaluated for cell growth and viability, and oxidative stress to assess the noscapine effect at 8, 10, 12, and 20 μg/ml concentrations. The neuroprotective activity of noscapine (5 and 10 mg/kg; po for 8 days) was investigated in rats using middle cerebral artery occlusion-induced I-R injury. Infarct volume, neurological deficit, oxidative stress, myeloperoxidase activity, and histological alterations were determined in I-R rats. In vitro yeast assays exhibited significant antioxidant activity and enhanced cell tolerance against oxidative stress after noscapine treatment. Similarly, noscapine pretreatment significantly reduced infarct volume and edema in the brain. The neurological deficit was decreased and oxidative stress biomarkers, superoxide dismutase activity and glutathione levels, were significantly increased while lipid peroxidation showed significant decrease in comparison to vehicle-treated I-R rats. Myeloperoxidase activity, an indicator of inflammation, was also reduced significantly in treated rats; histological changes were attenuated with noscapine. The study demonstrates the protective effect of noscapine in yeast and I-R rats by improving cell viability and attenuating neuronal damage, respectively. This protective activity of noscapine could be attributed to potent free radical scavenging and inhibition of inflammation in cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Manisha Kawadkar
- Department of Pharmacology, Faculty of Pharmacy, VNS Group of Institutions, VidyaVihar, Neelbud, Bhopal, Madhya Pradesh, 462044, India
| | - Avinash S Mandloi
- Department of Pharmacology, Faculty of Pharmacy, VNS Group of Institutions, VidyaVihar, Neelbud, Bhopal, Madhya Pradesh, 462044, India
| | - Vidhu Saxena
- Department of Pharmacology, Faculty of Pharmacy, VNS Group of Institutions, VidyaVihar, Neelbud, Bhopal, Madhya Pradesh, 462044, India
| | - Chetana Tamadaddi
- Chaperone and Stress Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh, 462066, India
| | - Chandan Sahi
- Chaperone and Stress Biology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, Madhya Pradesh, 462066, India
| | - Vipin V Dhote
- Department of Pharmacology, Faculty of Pharmacy, VNS Group of Institutions, VidyaVihar, Neelbud, Bhopal, Madhya Pradesh, 462044, India.
| |
Collapse
|
7
|
Michalski D, Spielvogel E, Puchta J, Reimann W, Barthel H, Nitzsche B, Mages B, Jäger C, Martens H, Horn AKE, Schob S, Härtig W. Increased Immunosignals of Collagen IV and Fibronectin Indicate Ischemic Consequences for the Neurovascular Matrix Adhesion Zone in Various Animal Models and Human Stroke Tissue. Front Physiol 2020; 11:575598. [PMID: 33192578 PMCID: PMC7649770 DOI: 10.3389/fphys.2020.575598] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/23/2020] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke causes cellular alterations in the “neurovascular unit” (NVU) comprising neurons, glia, and the vasculature, and affects the blood-brain barrier (BBB) with adjacent extracellular matrix (ECM). Limited data are available for the zone between the NVU and ECM that has not yet considered for neuroprotective approaches. This study describes ischemia-induced alterations for two main components of the neurovascular matrix adhesion zone (NMZ), i.e., collagen IV as basement membrane constituent and fibronectin as crucial part of the ECM, in conjunction with traditional NVU elements. For spatio-temporal characterization of these structures, multiple immunofluorescence labeling was applied to tissues affected by focal cerebral ischemia using a filament-based model in mice (4, 24, and 72 h of ischemia), a thromboembolic model in rats (24 h of ischemia), a coagulation-based model in sheep (2 weeks of ischemia), and human autoptic stroke tissue (3 weeks of ischemia). An increased fibronectin immunofluorescence signal demarcated ischemia-affected areas in mice, along with an increased collagen IV signal and BBB impairment indicated by serum albumin extravasation. Quantifications revealed a region-specific pattern with highest collagen IV and fibronectin intensities in most severely affected neocortical areas, followed by a gradual decline toward the border zone and non-affected regions. Comparing 4 and 24 h of ischemia, the subcortical fibronectin signal increased significantly over time, whereas neocortical areas displayed only a gradual increase. Qualitative analyses confirmed increased fibronectin and collagen IV signals in ischemic areas from all tissues and time points investigated. While the increased collagen IV signal was restricted to vessels, fibronectin appeared diffusely arranged in the parenchyma with focal accumulations associated to the vasculature. Integrin α5 appeared enriched in the vicinity of fibronectin and vascular elements, while most of the non-vascular NVU elements showed complementary staining patterns referring to fibronectin. This spatio-temporal characterization of ischemia-related alterations of collagen IV and fibronectin in various stroke models and human autoptic tissue shows that ischemic consequences are not limited to traditional NVU components and the ECM, but also involve the NMZ. Future research should explore more components and the pathophysiological properties of the NMZ as a possible target for novel neuroprotective approaches.
Collapse
Affiliation(s)
| | - Emma Spielvogel
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Joana Puchta
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany.,Department of Neuroradiology, University of Leipzig, Leipzig, Germany
| | - Willi Reimann
- Department of Neurology, University of Leipzig, Leipzig, Germany.,Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - Björn Nitzsche
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany.,Institute of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Bianca Mages
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Carsten Jäger
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | | | - Anja K E Horn
- Institute of Anatomy and Cell Biology I and German Center for Vertigo and Balance Disorders, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Schob
- Department of Neuroradiology, University of Leipzig, Leipzig, Germany
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
8
|
Thiebaut AM, Hedou E, Marciniak SJ, Vivien D, Roussel BD. Proteostasis During Cerebral Ischemia. Front Neurosci 2019; 13:637. [PMID: 31275110 PMCID: PMC6594416 DOI: 10.3389/fnins.2019.00637] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebral ischemia is a complex pathology involving a cascade of cellular mechanisms, which deregulate proteostasis and lead to neuronal death. Proteostasis refers to the equilibrium between protein synthesis, folding, transport, and protein degradation. Within the brain proteostasis plays key roles in learning and memory by controlling protein synthesis and degradation. Two important pathways are implicated in the regulation of proteostasis: the unfolded protein response (UPR) and macroautophagy (called hereafter autophagy). Both are necessary for cell survival, however, their over-activation in duration or intensity can lead to cell death. Moreover, UPR and autophagy can activate and potentiate each other to worsen the issue of cerebral ischemia. A better understanding of autophagy and ER stress will allow the development of therapeutic strategies for stroke, both at the acute phase and during recovery. This review summarizes the latest therapeutic advances implicating ER stress or autophagy in cerebral ischemia. We argue that the processes governing proteostasis should be considered together in stroke, rather than focusing either on ER stress or autophagy in isolation.
Collapse
Affiliation(s)
- Audrey M Thiebaut
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| | - Elodie Hedou
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| | - Stefan J Marciniak
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom.,Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Denis Vivien
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France.,Department of Clinical Research, University of Caen Normandy, Caen, France
| | - Benoit D Roussel
- INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, University of Caen Normandy, Caen, France
| |
Collapse
|
9
|
Reutov VP, Samosudova NV, Sorokina EG. A Model of Glutamate Neurotoxicity and Mechanisms of the Development of the Typical Pathological Process. Biophysics (Nagoya-shi) 2019. [DOI: 10.1134/s0006350919020143] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
10
|
Martire A, Lambertucci C, Pepponi R, Ferrante A, Benati N, Buccioni M, Dal Ben D, Marucci G, Klotz KN, Volpini R, Popoli P. Neuroprotective potential of adenosine A 1 receptor partial agonists in experimental models of cerebral ischemia. J Neurochem 2019; 149:211-230. [PMID: 30614535 DOI: 10.1111/jnc.14660] [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: 11/19/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 01/16/2023]
Abstract
Cerebral ischemia is the second most common cause of death and a major cause of disability worldwide. Available therapies are based only on anticoagulants or recombinant tissue plasminogen activator. Extracellular adenosine increases during ischemia and acts as a neuroprotective endogenous agent mainly by activating adenosine A1 receptors (A1 Rs) which control calcium influx, glutamate release, membrane potential, and metabolism. Accordingly, in many experimental paradigms it has been already demonstrated that the stimulation of A1 R with full agonists is able to reduce ischemia-related structural and functional brain damage; unfortunately, cardiovascular side effects and desensitization of A1 R induced by these compounds have strongly limited their exploitation in stroke therapy so far. Among the newly emerging compounds, A1 R partial agonists could be almost free of side effects and equally effective. Therefore, we decided to evaluate the neuroprotective potential of two A1 R partial agonists, namely 2'-dCCPA and 3'-dCCPA, in in vitro and ex vivo experimental models of cerebral ischemia. Within the experimental paradigm of oxygen-glucose deprivation in vitro in human neuroblastoma (SH-SY5Y) cells both A1 R partial agonists increased cell viability. Considering the high level of expression of A1 Rs in the hippocampus and the susceptibility of CA1 region to hypoxia, we performed electrophysiological experiments in this subfield. The application of 7 min of oxygen-glucose deprivation constantly produces an irreversible synaptic failure in all the C57Bl/6 mice hippocampal slices evaluated; both tested compounds allowed a significant recovery of synaptic transmission. These findings demonstrate that A1 R and its partial agonists are still of interest for cerebral ischemia therapy. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Alberto Martire
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Catia Lambertucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Rita Pepponi
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Antonella Ferrante
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Nicholas Benati
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Michela Buccioni
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Diego Dal Ben
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Gabriella Marucci
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Karl-Norbert Klotz
- Institut für Pharmakologie und Toxikologie, Universität Würzburg, Würzburg, Germany
| | - Rosaria Volpini
- Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
11
|
Furtado D, Björnmalm M, Ayton S, Bush AI, Kempe K, Caruso F. Overcoming the Blood-Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801362. [PMID: 30066406 DOI: 10.1002/adma.201801362] [Citation(s) in RCA: 389] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/09/2018] [Indexed: 05/24/2023]
Abstract
Therapies directed toward the central nervous system remain difficult to translate into improved clinical outcomes. This is largely due to the blood-brain barrier (BBB), arguably the most tightly regulated interface in the human body, which routinely excludes most therapeutics. Advances in the engineering of nanomaterials and their application in biomedicine (i.e., nanomedicine) are enabling new strategies that have the potential to help improve our understanding and treatment of neurological diseases. Herein, the various mechanisms by which therapeutics can be delivered to the brain are examined and key challenges facing translation of this research from benchtop to bedside are highlighted. Following a contextual overview of the BBB anatomy and physiology in both healthy and diseased states, relevant therapeutic strategies for bypassing and crossing the BBB are discussed. The focus here is especially on nanomaterial-based drug delivery systems and the potential of these to overcome the biological challenges imposed by the BBB. Finally, disease-targeting strategies and clearance mechanisms are explored. The objective is to provide the diverse range of researchers active in the field (e.g., material scientists, chemists, engineers, neuroscientists, and clinicians) with an easily accessible guide to the key opportunities and challenges currently facing the nanomaterial-mediated treatment of neurological diseases.
Collapse
Affiliation(s)
- Denzil Furtado
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Materials, Department of Bioengineering, and the Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
- Cooperative Research Center for Mental Health, Parkville, Victoria, 3052, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
12
|
Lv B, Cheng X, Sharp FR, Ander BP, Liu DZ. MicroRNA-122 Mimic Improves Stroke Outcomes and Indirectly Inhibits NOS2 After Middle Cerebral Artery Occlusion in Rats. Front Neurosci 2018; 12:767. [PMID: 30405345 PMCID: PMC6207613 DOI: 10.3389/fnins.2018.00767] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/03/2018] [Indexed: 01/19/2023] Open
Abstract
Aim: Our previous study demonstrated miR-122 mimic decreased NOS2 expression in blood leucocytes and improved stroke outcomes when given immediately after middle cerebral artery occlusion (MCAO) in rats. Since NOS2 is associated with neuro-inflammation in stroke and decreasing NOS2 expression alone in leucocytes is insufficient to improve stroke outcomes, we hypothesized that miR-122 mimic may also decrease NOS2 expression in brain microvascular endothelial cells (BMVECs) even at extended time windows. Methods: We administered PEG-liposome wrapped miR-122 mimic (2.4 mg/kg, i.v.) 0 or 6 h after MCAO, and assessed stroke volume and NOS2 expression in BMVECs 24 h following MCAO in rats. Luciferase reporter assays were used to determine if miR-122 binds to 3′ untranslated regions (3′UTR) of NOS2. Results: The data showed that miR-122 mimic decreased infarct volumes and decreased MCAO-induced NOS2 over-expression in BMVECs. However, miR-122 did not bind to 3′UTR of NOS2 in the luciferase assays. Conclusion: The data show the 6-h period of therapeutic efficacy of miR-122 mimic which could relate to indirect knockdown of NOS2 in both BMVECs and leucocytes.
Collapse
Affiliation(s)
- Bo Lv
- Department of Neurology, University of California, Davis, Davis, CA, United States.,Department of Critical Care Medicine and Emergency, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiyuan Cheng
- Department of Neurology, University of California, Davis, Davis, CA, United States
| | - Frank R Sharp
- Department of Neurology, University of California, Davis, Davis, CA, United States
| | - Bradley P Ander
- Department of Neurology, University of California, Davis, Davis, CA, United States
| | - Da Zhi Liu
- Department of Neurology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
13
|
Yu J, Li X, Matei N, McBride D, Tang J, Yan M, Zhang JH. Ezetimibe, a NPC1L1 inhibitor, attenuates neuronal apoptosis through AMPK dependent autophagy activation after MCAO in rats. Exp Neurol 2018; 307:12-23. [PMID: 29852178 DOI: 10.1016/j.expneurol.2018.05.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/22/2018] [Accepted: 05/27/2018] [Indexed: 12/25/2022]
Abstract
Autophagy activation exerts neuroprotective effects in the ischemic stroke model. Ezetimibe (Eze), a Niemann-Pick disease type C1-Like 1 (NPC1L1) pharmacological inhibitor, has been reported to protect hepatocytes from apoptosis via autophagy activation. In this study, we explored whether Eze could attenuate neuronal apoptosis in the rat model of middle cerebral artery occlusion (MCAO), specifically via activation of the AMPK/ULK1/autophagy pathway. Two hundred and one male Sprague-Dawley rats were subjected to transient MCAO followed by reperfusion. Eze was administered 1 h after MCAO. To elucidate the underlying molecular mechanism, Dorsomorphin, a selective AMPK inhibitor, and 3-methyladenine (3-MA), an autophagy inhibitor, were injected intracerebroventricularly before MCAO. Infarct volume, neurological score, brain cholesterol levels, immunofluorescence staining, Western blot, and Fluoro-Jade C (FJC) staining were used to evaluate the effects of Eze. The endogenous NPC1L1 expression increased and mainly expressed in neurons after MCAO. Intranasal administration of Eze reduced brain infarct volume at 24 and 72 h after MCAO, with improved short and long-term neurological functions after MCAO. Eze reduced brain cholesterol levels (total cholesterol, free cholesterol and cholesteryl esters) and the number of FJC-positive neurons. The expression of phosphorylated AMPK (p-AMPK) and downstream ULK1, Beclin1, LC3BII, Bcl-2, and Bcl-xl increased, while P62 and proapoptotic Bax decreased after treatment with Eze. Pretreatment with Dorsomorphin and 3-MA reversed the beneficial effects of Eze. These findings suggest that intranasal administration of Eze plays neuroprotective role through autophagy activation after MCAO in rats. Lowered cholesterol levels and AMPK activation may act in conjunction to induce autophagy after treatment with Eze. Eze merits further investigation as a potential therapeutic agent in ischemic stroke patients.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Nathanael Matei
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Devin McBride
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA; The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jiping Tang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China.
| | - John H Zhang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
14
|
An P, Wu T, Yu H, Fang K, Ren Z, Tang M. Hispidulin Protects Against Focal Cerebral Ischemia Reperfusion Injury in Rats. J Mol Neurosci 2018; 65:203-212. [PMID: 29799104 DOI: 10.1007/s12031-018-1086-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/11/2018] [Indexed: 02/02/2023]
Abstract
Focal cerebral ischemia is associated with ischemia/reperfusion (I/R) injury. Hispidulin is a flavonoid compound with a variety of pharmacological properties. The neuroprotective effects of hispidulin have not been fully elucidated. Herein, we demonstrated that pretreatment of animals with hispidulin improved the neurological outcomes and decreased the infarct size and brain edema in the cerebral focal I/R model. Mechanistically, we showed in vivo and in vitro that hispidulin exerted a protective effect against I/R injury by inducing the Nrf2 antioxidant pathway through modulation of AMPK/GSK3β signaling. Taken together, our results suggest that hispidulin may be a useful neuroprotective agent against ischemia/reperfusion (I/R) injury.
Collapse
Affiliation(s)
- Pengpeng An
- The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Renming Road 4, Qingdao, 266000, China
| | - Tianhui Wu
- Qingdao Fifth People's Hospital (Shandong Qingdao Hospital of Integrated Traditional and Western Medicine), Qingdao, China
| | - Huanqing Yu
- Qingdao Fifth People's Hospital (Shandong Qingdao Hospital of Integrated Traditional and Western Medicine), Qingdao, China
| | - Kun Fang
- Qingdao Fifth People's Hospital (Shandong Qingdao Hospital of Integrated Traditional and Western Medicine), Qingdao, China
| | - Zhizhen Ren
- Community Health Service Center of Shinan District, Qingdao, China
| | - Ming Tang
- The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Renming Road 4, Qingdao, 266000, China.
| |
Collapse
|
15
|
Yu L, Wan HF, Li C, Yang JH, Zhou HF, Wan HT, He Y. Pharmacokinetics of Active Components From Guhong Injection in Normal and Pathological Rat Models of Cerebral Ischemia: A Comparative Study. Front Pharmacol 2018; 9:493. [PMID: 29867497 PMCID: PMC5962683 DOI: 10.3389/fphar.2018.00493] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/25/2018] [Indexed: 11/13/2022] Open
Abstract
Background and Objectives: Guhong Injection (GHI) is usually administered for the treatment of stroke in clinics. Aceglutamide and hydroxyl safflower yellow A (HSYA) are its key ingredients for brain protective effect. To investigate the pharmacokinetics of aceglutamide and HSYA under pathological and normal conditions, the pharmacokinetic parameters and characteristics of middle cerebral artery occlusion (MCAO) and normal rats given the same dosage of GHI were studied compared. Methods: 12 SD rats were divided into two groups, namely, MCAO and normal groups. Both groups were treated with GHI in the same dosage. Plasma samples were collected from the jaw vein at different time points and subsequently tested by high-performance liquid chromatography (HPLC). Results: After administration of GHI, both aceglutamide and HSYA were immediately detected in the plasma. Ninety percent of aceglutamide and HSYA was eliminated within 3 h. For aceglutamide, statistically significant differences in the parameters including AUC(0-t), AUC(0-∞), AUMC(0-t), AUMC(0-∞), Cmax (P < 0.01), and Vz (P < 0.05). Meanwhile, compared with the MCAO group, in the normal group, the values of AUC(0-t), AUMC(0-t), VRT(0-t), and Cmax (P < 0.01) for HSYA were significantly higher, whereas the value of MRT(0-t) was significantly lower in the normal group. Conclusions: The in vivo trials based on the different models showed that, the pharmacokinetic behaviors and parameters of aceglutamide and HSYA in GHI were completely different. These results suggest that the pathological damage of ischemia-reperfusion has a significant impact on the pharmacokinetic traits of aceglutamide and HSYA.
Collapse
Affiliation(s)
- Li Yu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hao-Fang Wan
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chang Li
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie-Hong Yang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui-Fen Zhou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hai-Tong Wan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
16
|
Recent progress in the structural modification and pharmacological activities of ligustrazine derivatives. Eur J Med Chem 2018; 147:150-162. [PMID: 29432947 DOI: 10.1016/j.ejmech.2018.01.097] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 11/22/2022]
Abstract
Ligustrazine is a main active fraction of the traditional medicine known as Ligusticum chuanxiong hort, which has been used as clinical medication for cerebral thrombosis, coronary heart disease and stenocardia recently. The rapid metabolism and short half-life of ligustrazine seriously limits its application in clinical practice. Therefore, derivatives of ligustrazine are designed and synthesized in our and other labs, including piperazine, cinnamic acid, styrene, acylguanidine, amides, curcumin and triterpenes derivatives of ligustrazine. Most of these compounds present better pharmacodynamics activities and more favorable pharmacokinetic properties compared to the parent compound. Besides, some new biological activities of these compounds are discovered. Hence, this review continues the previous review of our group as well as aims to highlight recent prominent advances in this field in the past ten years.
Collapse
|
17
|
Wang P, Zhao R, Yan W, Zhang X, Zhang H, Xu B, Chu F, Han Y, Li G, Liu W, Zhang Y, Lei H. Neuroprotection by new ligustrazine-cinnamon acid derivatives on CoCl 2-induced apoptosis in differentiated PC12 cells. Bioorg Chem 2018; 77:360-369. [PMID: 29421712 DOI: 10.1016/j.bioorg.2018.01.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/15/2018] [Accepted: 01/24/2018] [Indexed: 10/18/2022]
Abstract
A new series of ligustrazine-cinnamon acid derivatives had been designed and synthesized as potential neuro-protective agents. Among the derivatives, 3a exhibited the promising neuroprotective activity (EC50 = 3.68 μM). Moreover, with the deep research of the drug pathway, it (the further mechanism researches) suggested compound 3a could inhibit the apoptosis of injured PC12 cells via blocking the mitochondria apoptosis pathway including up-regulation the ratio of Bcl-2/Bax, down-regulation the expression of cytochrome-c (Cyt-c) and inhibition of the activity of caspase-9 and -3. In addition, the structure-activity relationships (SARs) of novel compounds were also discussed.
Collapse
Affiliation(s)
- Penglong Wang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Rui Zhao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Wenqiang Yan
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Xinyu Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Huazheng Zhang
- Department of Pathology, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Fuhao Chu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Yaotian Han
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Guoliang Li
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China
| | - Wei Liu
- Department of Management, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yuzhong Zhang
- Department of Pathology, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
18
|
Thériault P, Le Béhot A, ElAli A, Rivest S. Sub-acute systemic erythropoietin administration reduces ischemic brain injury in an age-dependent manner. Oncotarget 2018; 7:35552-35561. [PMID: 27248662 PMCID: PMC5094944 DOI: 10.18632/oncotarget.9652] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/19/2016] [Indexed: 12/26/2022] Open
Abstract
Stroke is associated with neuroinflammation, neuronal loss and blood-brain barrier (BBB) breakdown. Thus far, recombinant tissue-type plasminogen activator (rtPA), the only approved treatment for acute ischemic stroke, increases the risk of intracerebral hemorrhage and is poorly efficient in disaggregating platelet-rich thrombi. Therefore, the development of safer and more efficient therapies is highly awaited. Encouraging neuroprotective effects were reported in mouse models of ischemic stroke following administration of erythropoietin (EPO). However, previous preclinical studies did not investigate the effects of EPO in focal ischemic stroke induced by a platelet-rich thrombus and did not consider the implication of age. Here, we performed middle cerebral artery occlusion by inducing platelet-rich thrombus formation in chimeric 5- (i.e. young) and 20- (i.e. aged) months old C57BL/6 mice, in which hematopoietic stem cells carried the green fluorescent protein (GFP)-tag. Recombinant human EPO (rhEPO) was administered 24 hours post-occlusion and blood-circulating monocyte populations were studied by flow cytometry 3 hours post-rhEPO administration. Twenty-four hours following rhEPO treatment, neuronal loss and BBB integrity were assessed by quantification of Fluoro-Jade B (FJB)-positive cells and extravasated serum immunoglobulins G (IgG), respectively. Neuroinflammation was determined by quantifying infiltration of GFP-positive bone marrow-derived cells (BMDC) and recruitment of microglial cells into brain parenchyma, along with monocyte chemotactic protein-1 (MCP-1) brain protein levels. Here, rhEPO anti-inflammatory properties rescued ischemic injury by reducing neuronal loss and BBB breakdown in young animals, but not in aged littermates. Such age-dependent effects of rhEPO must therefore be taken into consideration in future studies aiming to develop new therapies for ischemic stroke.
Collapse
Affiliation(s)
- Peter Thériault
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
| | - Audrey Le Béhot
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec City, QC, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
| |
Collapse
|
19
|
Jin GH, Lee JB. Effect of Recombinant Human Growth Hormone Add on Therapy on Acute Stroke Outcome. BRAIN & NEUROREHABILITATION 2018. [DOI: 10.12786/bn.2018.11.e4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ga-Heon Jin
- The Faculty of Beauty Health Sciences, Major in Ophthalmic Optics, Shinhan University, Uijeongbu, Korea
| | - Jun-Beom Lee
- Department of Neurology, Hongik Hospital, Seoul, Korea
| |
Collapse
|
20
|
Hexahydrocurcumin protects against cerebral ischemia/reperfusion injury, attenuates inflammation, and improves antioxidant defenses in a rat stroke model. PLoS One 2017; 12:e0189211. [PMID: 29220411 PMCID: PMC5722321 DOI: 10.1371/journal.pone.0189211] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/21/2017] [Indexed: 12/04/2022] Open
Abstract
The purpose of the present experiment was to investigate whether hexahydrocurcumin (HHC) attenuates brain damage and improves functional outcome via the activation of antioxidative activities, anti-inflammation, and anti-apoptosis following cerebral ischemia/reperfusion (I/R). In this study, rats with cerebral I/R injury were induced by a transient middle cerebral artery occlusion (MCAO) for 2 h, followed by reperfusion. The male Wistar rats were randomly divided into five groups, including the sham-operated, vehicle-treated, 10 mg/kg HHC-treated, 20 mg/kg HHC-treated, and 40 mg/kg HHC-treated I/R groups. The animals were immediately injected with HHC by an intraperitoneal administration at the onset of cerebral reperfusion. After 24 h of reperfusion, the rats were tested for neurological deficits, and the pathology of the brain was studied by 2,3,5-triphenyltetrazolium chloride (TTC) staining, hematoxylin and eosin (H&E) staining, and terminal deoxynucleotidyltransferase UTP nick end labeling (TUNEL) staining. In addition, the brain tissues were prepared for protein extraction for Western blot analysis, a malondialdehyde (MDA) assay, a nitric oxide (NO) assay, a superoxide dismutase (SOD) assay, a glutathione (GSH) assay, and a glutathione peroxidase (GSH-Px) assay. The data revealed that the neurological deficit scores and the infarct volume were significantly reduced in the HHC-treated rats at all doses compared to the vehicle group. Treatment with HHC significantly attenuated oxidative stress and inflammation, with a decreased level of MDA and NO and a decreased expression of NF-κB (p65) and cyclooxygenase-2 (COX-2) in the I/R rats. HHC also evidently increased Nrf2 (nucleus) protein expression, heme oxygenase-1 (HO-1) protein expression, the antioxidative enzymes, and the superoxide dismutase (SOD) activity. Moreover, the HHC treatment also significantly decreased apoptosis, with a decrease in Bax and cleaved caspase-3 and an increase in Bcl-XL, which was in accordance with a decrease in the apoptotic neuronal cells. Therefore, the HHC treatment protects the brain from cerebral I/R injury by diminishing oxidative stress, inflammation, and apoptosis. The antioxidant properties of HHC may play an important role in improving functional outcomes and may offer significant neuroprotection against I/R damage.
Collapse
|
21
|
Härtig W, Mages B, Aleithe S, Nitzsche B, Altmann S, Barthel H, Krueger M, Michalski D. Damaged Neocortical Perineuronal Nets Due to Experimental Focal Cerebral Ischemia in Mice, Rats and Sheep. Front Integr Neurosci 2017; 11:15. [PMID: 28860977 PMCID: PMC5559442 DOI: 10.3389/fnint.2017.00015] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/10/2017] [Indexed: 01/08/2023] Open
Abstract
As part of the extracellular matrix (ECM), perineuronal nets (PNs) are polyanionic, chondroitin sulfate proteoglycan (CSPG)-rich coatings of certain neurons, known to be affected in various neural diseases. Although these structures are considered as important parts of the neurovascular unit (NVU), their role during evolution of acute ischemic stroke and subsequent tissue damage is poorly understood and only a few preclinical studies analyzed PNs after acute ischemic stroke. By employing three models of experimental focal cerebral ischemia, this study was focused on histopathological alterations of PNs and concomitant vascular, glial and neuronal changes according to the NVU concept. We analyzed brain tissues obtained 1 day after ischemia onset from: (a) mice after filament-based permanent middle cerebral artery occlusion (pMCAO); (b) rats subjected to thromboembolic MACO; and (c) sheep at 14 days after electrosurgically induced focal cerebral ischemia. Multiple fluorescence labeling was applied to explore simultaneous alterations of NVU and ECM. Serial mouse sections labeled with the net marker Wisteria floribunda agglutinin (WFA) displayed largely decomposed and nearly erased PNs in infarcted neocortical areas that were demarcated by up-regulated immunoreactivity for vascular collagen IV (Coll IV). Subsequent semi-quantitative analyses in mice confirmed significantly decreased WFA-staining along the ischemic border zone and a relative decrease in the directly ischemia-affected neocortex. Triple fluorescence labeling throughout the three animal models revealed up-regulated Coll IV and decomposed PNs accompanied by activated astroglia and altered immunoreactivity for parvalbumin, a calcium-binding protein in fast-firing GABAergic neurons which are predominantly surrounded by neocortical PNs. Furthermore, ischemic neocortical areas in rodents simultaneously displayed less intense staining of WFA, aggrecan, the net components neurocan, versican and the cartilage link protein (CRTL) as well as markers in net-bearing neurons such as the potassium channel subunit Kv3.1b and neuronal nuclei (NeuN). In summary, theconsistent observations based on three different stroke models confirmed that PNs are highly sensitive constituents of the NVU along with impaired associated GABAergic neurons. These results suggest that PNs could be promising targets of future stroke treatment, and further studies should address their reorganization and plasticity in both stabilizing the acute stroke as well as supportive effects during the chronic phase of stroke.
Collapse
Affiliation(s)
- Wolfgang Härtig
- Department of Pathophysiology of Neuroglia, Paul Flechsig Institute for Brain Research, University of LeipzigLeipzig, Germany
| | - Bianca Mages
- Department of Pathophysiology of Neuroglia, Paul Flechsig Institute for Brain Research, University of LeipzigLeipzig, Germany.,Department of Neurology, University of LeipzigLeipzig, Germany
| | - Susanne Aleithe
- Department of Pathophysiology of Neuroglia, Paul Flechsig Institute for Brain Research, University of LeipzigLeipzig, Germany.,Department of Neurology, University of LeipzigLeipzig, Germany
| | - Björn Nitzsche
- Department of Nuclear Medicine, University of LeipzigLeipzig, Germany.,Faculty of Veterinary Medicine, Institute of Anatomy, Histology and Embryology, University of LeipzigLeipzig, Germany
| | - Stephan Altmann
- Department of Pathophysiology of Neuroglia, Paul Flechsig Institute for Brain Research, University of LeipzigLeipzig, Germany.,Department of Neurology, University of LeipzigLeipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of LeipzigLeipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, University of LeipzigLeipzig, Germany
| | | |
Collapse
|
22
|
Gribkoff VK, Kaczmarek LK. The need for new approaches in CNS drug discovery: Why drugs have failed, and what can be done to improve outcomes. Neuropharmacology 2017; 120:11-19. [PMID: 26979921 PMCID: PMC5820030 DOI: 10.1016/j.neuropharm.2016.03.021] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/14/2016] [Accepted: 03/11/2016] [Indexed: 12/31/2022]
Abstract
An important goal of biomedical research is to translate basic research findings into useful medical advances. In the field of neuropharmacology this requires understanding disease mechanisms as well as the effects of drugs and other compounds on neuronal function. Our hope is that this information will result in new or improved treatment for CNS disease. Despite great progress in our understanding of the structure and functions of the CNS, the discovery of new drugs and their clinical development for many CNS disorders has been problematic. As a result, CNS drug discovery and development programs have been subjected to significant cutbacks and eliminations over the last decade. While there has been recent resurgence of interest in CNS targets, these past changes in priority of the pharmaceutical and biotech industries reflect several well-documented realities. CNS drugs in general have higher failure rates than non-CNS drugs, both preclinically and clinically, and in some areas, such as the major neurodegenerative diseases, the clinical failure rate for disease-modifying treatments has been 100%. The development times for CNS drugs are significantly longer for those drugs that are approved, and post-development regulatory review is longer. In this introduction we review some of the reasons for failure, delineating both scientific and technical realities, some unique to the CNS, that have contributed to this. We will focus on major neurodegenerative disorders, which affect millions, attract most of the headlines, and yet have witnessed the fewest successes. We will suggest some changes that, when coupled with the approaches discussed in the rest of this special volume, may improve outcomes in future CNS-targeted drug discovery and development efforts. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Valentin K Gribkoff
- Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
23
|
Khoshnam SE, Winlow W, Farzaneh M. The Interplay of MicroRNAs in the Inflammatory Mechanisms Following Ischemic Stroke. J Neuropathol Exp Neurol 2017; 76:548-561. [DOI: 10.1093/jnen/nlx036] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
24
|
Zhang C, Yan W, Zhao R, Xu B, Fang X, Yan M, Zhang Y, Wang P, Lei H. Design, synthesis and evaluation of new ligustrazine derivatives as potential plasma-stable neuroprotective agents. MEDCHEMCOMM 2017; 8:652-656. [PMID: 30108782 DOI: 10.1039/c7md00003k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/08/2017] [Indexed: 01/08/2023]
Abstract
A series of ligustrazine-phenolic acid esters which exhibited promising neuroprotective activities have previously been reported. Nevertheless, we found that these ester compounds (like T-VA) were not stable in plasma by further in vivo studies. To investigate plasma-stable neuroprotective agents, a series of new ligustrazine derivatives were synthesized by conjoining ligustrazine and phenols with ester, ether and amide bonds. Most of the compounds exhibited higher protective effects against CoCl2-induced neurotoxicity in differentiated PC12 cells than ligustrazine. Structure-activity relationships were also briefly discussed. We found that compound 2c (2-((2-methoxy-4-(((3,5,6-trimethylpyrazin-2-yl)methoxy) methyl)phenoxy)methyl)-3,5,6-trimethylpyrazine) displayed the highest protective effect on the PC12 cells damaged by CoCl2 (EC50 = 1.07 μM). Preliminary stability investigation in rat plasma was verified in vitro and better plasma stability was observed with 2c in comparison to T-VA.
Collapse
Affiliation(s)
- Chenze Zhang
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| | - Wenqiang Yan
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| | - Rui Zhao
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| | - Bing Xu
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| | - Xiong Fang
- School of Life Sciences , Tsinghua University , Beijing 100084 , China
| | - Mengmeng Yan
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| | - Yuzhong Zhang
- Department of Pathology , Beijing University of Chinese Medicine , Beijing 100102 , China
| | - Penglong Wang
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| | - Haimin Lei
- School of Chinese Pharmacy , Beijing University of Chinese Medicine , Beijing 100102 , China . ;
| |
Collapse
|
25
|
Up-regulation of neurofilament light chains is associated with diminished immunoreactivities for MAP2 and tau after ischemic stroke in rodents and in a human case. J Chem Neuroanat 2016; 78:140-148. [DOI: 10.1016/j.jchemneu.2016.09.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/09/2016] [Accepted: 09/09/2016] [Indexed: 11/18/2022]
|
26
|
Tian X, Peng J, Zhong J, Yang M, Pang J, Lou J, Li M, An R, Zhang Q, Xu L, Dong Z. β-Caryophyllene protects in vitro neurovascular unit against oxygen-glucose deprivation and re-oxygenation-induced injury. J Neurochem 2016; 139:757-768. [PMID: 27565895 DOI: 10.1111/jnc.13833] [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: 06/15/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 01/21/2023]
Abstract
β-Caryophyllene (BCP) mediates neuroprotection in cerebral ischemic animals. The neurovascular unit (NVU) acts as an intricate network to maintain the neuronal homeostatic microenvironment. However, the effects exerted by BCP on NVU remain unclear. Therefore, we established an in vitro NVU model to investigate the effects of BCP on oxygen-glucose deprivation and re-oxygenation (OGD/R)-induced injury. This model involved the co-culture of brain microvascular endothelial cells, neurons, and astrocytes. BCP (10 μmol/L) was applied for 24 h prior to OGD/R and maintained throughout OGD/R. Blood-brain barrier (BBB) integrity and neuronal apoptosis were analyzed. BCP pre-treatment prior to the initiation of OGD/R significantly (i) decreased BBB permeability and neuronal apoptosis, (ii) mitigated oxidative stress damage and the release of inflammatory cytokines, (iii) down-regulated Bax expression, metalloproteinase-9 activity and expression, and (iv) up-regulated claudin-5, occludin, ZO-1, growth-associated protein-43 and Bcl-2 expression. Thus, BCP pre-treatment exerted multiple protective effects on NVU in the context of OGD/R-induced injury. These protective effects potentially occur via reductions in oxidative stress damage and inflammatory cytokines that induce BBB breakdown, subsequently resulting in reduced neuronal apoptosis. The NVU serves as putative therapeutic targets for cerebral ischemia, and the results of this study provide new insights for the application of BCP as a neuroprotective agent.
Collapse
Affiliation(s)
- Xiaocui Tian
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Jianhua Peng
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianjun Zhong
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mei Yang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Jinwei Pang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Lou
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Minghang Li
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Ruidi An
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Qian Zhang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Lu Xu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Zhi Dong
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, College of Pharmacy, Chongqing Medical University, Yuzhong District, Chongqing, China
| |
Collapse
|
27
|
Ai J, Wan H, Shu M, Zhou H, Zhao T, Fu W, He Y. Guhong injection protects against focal cerebral ischemia-reperfusion injury via anti-inflammatory effects in rats. Arch Pharm Res 2016; 40:610-622. [PMID: 27624481 DOI: 10.1007/s12272-016-0835-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 09/07/2016] [Indexed: 11/30/2022]
Abstract
Guhong injection (GHI), composed of aceglutamide and safflower aqueous extract, has been used clinically for the treatment of cerebrovascular diseases such as cerebral embolism, hemorrhage and mental deterioration. In this paper, we reported the results of the first study on the anti-inflammatory effects of GHI in murine focal cerebral ischemia-reperfusion (I/R) injury. Adult male SD rats were randomly divided into six groups: Sham group, I/R group, GHI-L group (2.5 mL/kg), GHI-M group (5 mL/kg), GHI-H group (10 mL/kg) and Nimodipine group. I/R injury was induced by middle cerebral artery occlusion (MCAO) for 1.5 h followed by reperfusion for 24 h. Compared with I/R group, rats treated with GHI showed dose dependent reductions in neurological defect scores and cerebral infarct volume. GHI obviously down-regulated nitric oxide (NO), inducible NO synthase (iNOS), myeloperoxidase (MPO), interleukin-1β (IL-1β), TNF-α (tumor necrosis factor-α) and C reactive protein (CRP) levels in serum. Moreover, histological examination by H&E staining showed that clear cell outline, less vacuolated spaces and largely surviving neurons were observed in GHI-treated rats. The immunohistochemical staining revealed that GHI administration significantly diminished the positive expressions of intercellular cell adhesion molecule-1 (ICAM-1) and nuclear factor-κB p65 (NF-κB p65) in brain tissues. Western blot analysis for ICAM, NF-κB p65 and iNOS further solidified the above findings. All these results demonstrate that GHI exerts a strong and ameliorative effect on cerebral I/R injury in rats possibly through the inhibition of inflammation.
Collapse
Affiliation(s)
- Jinchao Ai
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Mingchun Shu
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China
| | - Tao Zhao
- Buchang Pharmaceutical Co., Ltd., Xi'an, 710075, China
| | - Wei Fu
- Buchang Pharmaceutical Co., Ltd., Xi'an, 710075, China
| | - Yu He
- Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
28
|
Zhang X, Xue X, Xian L, Guo Z, Ito Y, Sun W. Potential neuroprotection of protodioscin against cerebral ischemia-reperfusion injury in rats through intervening inflammation and apoptosis. Steroids 2016; 113:52-63. [PMID: 27343977 PMCID: PMC8725987 DOI: 10.1016/j.steroids.2016.06.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 06/09/2016] [Accepted: 06/15/2016] [Indexed: 11/16/2022]
Abstract
The aim of the current research is to investigate the cerebral-protection of protodioscin on a transient cerebral ischemia-reperfusion (I/R) model and to explore its possible underlying mechanisms. The rats were preconditioned with protodioscin at the doses of 25 and 50mgkg(-1) prior to surgery. Then the animals were subjected to right middle cerebral artery occlusion (MCAO) using an intraluminal method by inserting a thread (90min surgery). After the blood flow was restored in 24h via withdrawing the thread, some representative indicators for the cerebral injury were evaluated by various methods including TTC-staining, TUNEL, immunohistochemistry, and Western blotting. As compared with the operated rats without drug intervening, treatment with protodioscin apparently lowered the death rate and improved motor coordination abilities through reducing the deficit scores and cerebral infarct volume. What's more, an apparent decrease in neuron apoptosis detected in hippocampus CA1 and cortex of the ipsilateral hemisphere might attribute to alleviate the increase in Caspase-3 and Bax/Bcl-2 ratio. Meanwhile, concentrations of several main pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) in the serum were also significantly suppressed. Finally, the NF-κB and IκBa protein expressions in the cytoplasm of right injured brain were remarkably up-regulated, while NF-κB in nucleus was down-regulated. Therefore, these observed findings demonstrated that protodioscin appeared to reveal potential neuroprotection against the I/R injury due to its anti-inflammatory and anti-apoptosis properties. This therapeutic effect was probably mediated by the inactivation of NF-κB signal pathways.
Collapse
Affiliation(s)
- Xinxin Zhang
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xuanji Xue
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Liang Xian
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Zengjun Guo
- College of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China.
| | - Yoichiro Ito
- Laboratory of Bioseparation Technology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wenji Sun
- Biomedicine Key Laboratory of Shaanxi Province, Northwest University, Xi'an 710069, China.
| |
Collapse
|
29
|
Ishihara Y, Fujitani N, Sakurai H, Takemoto T, Ikeda-Ishihara N, Mori-Yasumoto K, Nehira T, Ishida A, Yamazaki T. Effects of sex steroid hormones and their metabolites on neuronal injury caused by oxygen-glucose deprivation/reoxygenation in organotypic hippocampal slice cultures. Steroids 2016; 113:71-7. [PMID: 27389922 DOI: 10.1016/j.steroids.2016.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/22/2016] [Accepted: 06/08/2016] [Indexed: 10/21/2022]
Abstract
In this study, protective actions of the sex steroid hormones, progesterone, testosterone, and 17β-estradiol, against oxygen-glucose deprivation (OGD)/reoxygenation-induced neuronal cell death were examined using rat organotypic hippocampal slice cultures. Progesterone, testosterone, and 17β-estradiol significantly attenuated neuronal cell death elicited by OGD/reoxygenation. While the neuroprotection conferred by progesterone was not affected by SU-10603, an inhibitor of cytochrome P45017α, finasteride, a 5α-reductase inhibitor that blocks the conversion of progesterone to allopregnanolone, partially reversed the neuroprotection induced by progesterone. The progesterone metabolite, allopregnanolone attenuated neuronal injury induced by OGD/reoxygenation. Pretreatment with letrozole, a cytochrome P450 aromatase inhibitor or 4-hydroxyphenyl-1-naphthol, a 17β-hydroxysteroid dehydrogenase 2 inhibitor showed no effect on testosterone-mediated neuroprotection, while finasteride completely abolished the protective action of testosterone. Treatment with 5α-dihydrotestosterone significantly suppressed neuronal injury. Pretreatment with mifepristone, a progesterone receptor antagonist and hydroxyflutamid, an androgen receptor antagonist significantly diminished the neuroprotective effects of progesterone and testosterone, respectively. ICI182,780, an estrogen receptor antagonist, showed no effect on neuroprotection mediated by 17β-estradiol. Pretreatment with actinomycin D or cycloheximide clearly abolished the neuroprotective effects of progesterone and testosterone, while actinomycin D and cycloheximide did not show any effect on neuroprotection mediated by 17β-estradiol. Taken together, progesterone protects neurons via progesterone receptor-dependent genomic pathway, and allopregnanolone is involved in progesterone-mediated neuroprotection. Testosterone and its metabolite 5α-dihydrotestosterone protect neurons via the genomic pathway of the androgen receptor. Metabolism of sex steroid hormones in the brain might complicate their protective actions in the brain.
Collapse
Affiliation(s)
- Yasuhiro Ishihara
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan.
| | - Noriko Fujitani
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Hikaru Sakurai
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Takuya Takemoto
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Nami Ikeda-Ishihara
- Division of Gene Research, Natural Science Center for Basic Research and Development, Hiroshima University, Higashi-Hiroshima 739-8527, Japan
| | - Kanami Mori-Yasumoto
- Laboratory of Pharmacognosy and Natural Products Chemistry, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Tatsuo Nehira
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Atsuhiko Ishida
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| | - Takeshi Yamazaki
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima 739-8521, Japan
| |
Collapse
|
30
|
Deng Y, Xiong D, Yin C, Liu B, Shi J, Gong Q. Icariside II protects against cerebral ischemia–reperfusion injury in rats via nuclear factor-κB inhibition and peroxisome proliferator-activated receptor up-regulation. Neurochem Int 2016; 96:56-61. [DOI: 10.1016/j.neuint.2016.02.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/18/2016] [Accepted: 02/25/2016] [Indexed: 01/18/2023]
|
31
|
Wang T, Duan S, Wang H, Sun S, Han B, Fu F. Neurological function following cerebral ischemia/reperfusion is improved by the Ruyi Zhenbao pill in a rats. Biomed Rep 2016; 4:161-166. [PMID: 26893831 PMCID: PMC4734183 DOI: 10.3892/br.2016.568] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 12/16/2015] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate the effect and underlying mechanisms of the Ruyi Zhenbao pill on neurological function following cerebral ischemia/reperfusion in rats. Male Sprague-Dawley rats underwent middle cerebral artery occlusion following reperfusion. The rats received intragastrically either sodium carboxymethyl cellulose (control and model groups) or Ruyi Zhenbao pill at doses of 0.2, 0.4 or 0.8 g/kg. Neurological function was assessed by cylinder, adhesive and beam-walking tests after 14-day Ruyi Zhenbao pill treatment. Neurogenesis and angiogenesis were detected using immunofluorescence staining. The expression levels of brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and vascular endothelial growth factor (VEGF) were determined by enzyme-linked immunosorbent assays. Treatment with 0.4 and 0.8 g/kg Ruyi Zhenbao for 14 days significantly improved neurological function, and increased the number of von Willebrand Factor- and neuronal nuclear antigen-positive cells in the ischemic hemisphere of rats. Ruyi Zhenbao pill treatment also significantly enhanced the expression levels of BDNF, NGF and VEGF in the ischemic hemisphere. The results demonstrated that the Ruyi Zhenbao pill improved neurological function following ischemia in rats. The mechanisms of the Ruyi Zhenbao pill are associated with increasing the expression levels of BDNF, NGF and VEGF, and subsequently promoting neurogenesis and angiogenesis in the ischemic zone.
Collapse
Affiliation(s)
- Tian Wang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Sijin Duan
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Haiping Wang
- Shandong ARURA Pharmaceutical R&D Co., Ltd., Jinan, Shandong 250101, P.R. China
| | - Shan Sun
- Shandong ARURA Pharmaceutical R&D Co., Ltd., Jinan, Shandong 250101, P.R. China
| | - Bing Han
- Department of Food Safety, School of Life Science, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Fenghua Fu
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| |
Collapse
|
32
|
Xiong D, Deng Y, Huang B, Yin C, Liu B, Shi J, Gong Q. Icariin attenuates cerebral ischemia-reperfusion injury through inhibition of inflammatory response mediated by NF-κB, PPARα and PPARγ in rats. Int Immunopharmacol 2015; 30:157-162. [PMID: 26679678 DOI: 10.1016/j.intimp.2015.11.035] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 11/22/2015] [Accepted: 11/30/2015] [Indexed: 11/19/2022]
Abstract
Icariin (ICA), an active flavonoid extracted from Chinese medicinal herb Epimedii, has been reported to exhibit many pharmacological effects including alleviating brain injury. However, little is known about the protection of ICA on ischemic stroke. Hence, this study was designed to investigate the neuroprotective effect of ICA and explore its underlying mechanisms on ischemic stroke induced by cerebral ischemia-reperfusion (I/R) injury in rats. The animals were pretreated with ICA at doses of 10, 30mg/kg twice per day for 3 consecutive days followed by cerebral I/R injury induced by middle cerebral artery occlusion (MCAO) for 2h and reperfusion for 24h. Neurological function and infarct volume were observed at 24h after reperfusion, the protein expression levels of interleukin-1β (IL-1β), transforming growth factor-β1 (TGF-β1), PPARα and PPARγ, inhibitory κB-α (IκB-α) degradation and nuclear factor κB (NF-κB) p65 phosphorylation were detected by Western blot, respectively. It was found that pretreatment with ICA could decrease neurological deficit score, diminish the infarct volume, and reduce the protein levels of IL-1β and TGF-β1. Moreover, ICA suppressed IκB-α degradation and NF-κB activation induced by I/R. Furthermore, the present study also showed that ICA up-regulated PPARα and PPARγ protein levels. These findings suggest that ICA has neuroprotective effect on ischemic stroke in rats through inhibition of inflammatory responses mediated by NF-κB and PPARα and PPARγ.
Collapse
Affiliation(s)
- Deqing Xiong
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Yuanyuan Deng
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Bin Huang
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Caixia Yin
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Bo Liu
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Jingshan Shi
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Qihai Gong
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
33
|
Pedata F, Dettori I, Coppi E, Melani A, Fusco I, Corradetti R, Pugliese AM. Purinergic signalling in brain ischemia. Neuropharmacology 2015; 104:105-30. [PMID: 26581499 DOI: 10.1016/j.neuropharm.2015.11.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022]
Abstract
Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia a primary damage due to the early massive increase of extracellular glutamate is followed by activation of resident immune cells, i.e microglia, and production or activation of inflammation mediators. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. Extracellular concentrations of ATP and adenosine in the brain increase dramatically during ischemia in concentrations able to stimulate their respective specific P2 and P1 receptors. Both ATP P2 and adenosine P1 receptor subtypes exert important roles in ischemia. Although adenosine exerts a clear neuroprotective effect through A1 receptors during ischemia, the use of selective A1 agonists is hampered by undesirable peripheral effects. Evidence up to now in literature indicate that A2A receptor antagonists provide protection centrally by reducing excitotoxicity, while agonists at A2A (and possibly also A2B) and A3 receptors provide protection by controlling massive infiltration and neuroinflammation in the hours and days after brain ischemia. Among P2X receptors most evidence indicate that P2X7 receptor contribute to the damage induced by the ischemic insult due to intracellular Ca(2+) loading in central cells and facilitation of glutamate release. Antagonism of P2X7 receptors might represent a new treatment to attenuate brain damage and to promote proliferation and maturation of brain immature resident cells that can promote tissue repair following cerebral ischemia. Among P2Y receptors, antagonists of P2Y12 receptors are of value because of their antiplatelet activity and possibly because of additional anti-inflammatory effects. Moreover strategies that modify adenosine or ATP concentrations at injury sites might be of value to limit damage after ischemia. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy.
| | - Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Elisabetta Coppi
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alessia Melani
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Irene Fusco
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Renato Corradetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| |
Collapse
|
34
|
A Novel Ligustrazine Derivative T-VA Prevents Neurotoxicity in Differentiated PC12 Cells and Protects the Brain against Ischemia Injury in MCAO Rats. Int J Mol Sci 2015; 16:21759-74. [PMID: 26370988 PMCID: PMC4613278 DOI: 10.3390/ijms160921759] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/24/2015] [Accepted: 08/31/2015] [Indexed: 01/01/2023] Open
Abstract
Broad-spectrum drugs appear to be more promising for the treatment of acute ischemic stroke. In our previous work, a new ligustrazine derivative (3,5,6-trimethylpyrazin-2-yl) methyl 3-methoxy-4-[(3,5,6-trimethylpyrazin-2-yl)methoxy]benzoate (T-VA) showed neuroprotective effect on injured PC12 cells (EC50 = 4.249 µM). In the current study, we show that this beneficial effect was due to the modulation of nuclear transcription factor-κB/p65 (NF-κB/p65) and cyclooxygenase-2 (COX-2) expressions. We also show that T-VA exhibited neuroprotective effect in a rat model of ischemic stroke with concomitant improvement of motor functions. We propose that the protective effect observed in vivo is owing to increased vascular endothelial growth factor (VEGF) expression, decreased oxidative stress, and up-regulation of Ca2+–Mg2+ ATP enzyme activity. Altogether, our results warrant further studies on the utility of T-VA for the potential treatment of ischemic brain injuries, such as stroke.
Collapse
|
35
|
Chronic Brain Inflammation: The Neurochemical Basis for Drugs to Reduce Inflammation. Neurochem Res 2015; 41:523-33. [PMID: 26177578 DOI: 10.1007/s11064-015-1661-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/29/2015] [Accepted: 07/01/2015] [Indexed: 12/11/2022]
Abstract
It is now recognised that the brain and the peripheral immune system have bidirectional communication in both health and neuronal diseases. Brain inflammation results after both acute injury and also with the appearance of mutated proteins or endogenous neurotoxic metabolites associated with slow neurodegenerative diseases such as Alzheimer's and Parkinson's diseases and some psychiatric disorders. Microglia play a key role in brain inflammation by the release of pro-inflammatory cytokines and with ageing, microglia exhibit 'priming' leading to increased basal release of the pro-inflammatory cytokines. Neurochemical targets to reduce or slow chronic brain inflammation include cyclooxygenase enzymes, Nrf2 transcription factor, angiotensin AT1 receptors and sigma-1 receptors. Development of more selective drugs to act at these targets is occurring but large scale clinical trials to validate the drugs will take significant time.
Collapse
|
36
|
Cao ZQ, Quan W, Hou SX, Guo C, Ma SB, Zhang W, Li X. The natural therapeutic magnesium lithospermate B potently provides neuroprotective effects on cerebral ischemia/reperfusion injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2015; 162:191-198. [PMID: 25560670 DOI: 10.1016/j.jep.2014.12.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 12/08/2014] [Accepted: 12/23/2014] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salvia miltiorrhiza, a perennial plant in the genus Salvia and popularly known as "Danshen", is highly valued for its roots in traditional Chinese medicines (TCMs). It has widely used for the treatment of cerebrovascular and cardiovascular diseases in China. Recently, the cerebral protection of magnesium lithospermate B (MLB), a working extract from Salvia miltiorrhiza, has received more attention. Here, we investigated the therapeutic effects of MLB on cerebral ischemia/reperfusion (CI/R) injury using the middle cerebral artery occlusion (MCAO) model in rats. MATERIALS AND METHODS Male Sprague-Dawley rats were subjected to CI/R using a thread to occlude the right middle cerebral artery. After 2h of cerebral ischemia, the middle cerebral artery was reperfused for 24 h. Rats were injected with different doses of MLB (15, 30 and 60 mg/kg). Infarct zones, neurological deficit scores, brain water content, glutamate levels and protein expressions were evaluated after 24h of reperfusion. RESULTS We found that MLB treatment of rats exposed to focal CI/R decreased neurological deficit scores, brain water content, glutamate levels and cerebral infarct zones. We also demonstrated that MLB can inhibit CI/R injury-induced activation of caspase-3, a marker of apoptosis. This protection by MLB against CI/R injury was accompanied by an upregulation of p-Akt in the ischemic hemisphere. Furthermore, the MLB-induced protection was prevented by treatment with a PI3K inhibitor (LY-294002). CONCLUSIONS The data in the present study suggest a potential protective role of MLB against CI/R injury in rats. The salient finding of the present study is that this protective effect of MLB is likely mediated through an Akt-dependent pathway.
Collapse
Affiliation(s)
- Zhi-qiang Cao
- Department of Urinary Urology, General Hospital of Shenyang Military Region, Shenyang 110840, China
| | - Wei Quan
- Department of Pharmacy, Xi'an Mental Health Center, Institute of Mental Health, Xi'an Medical University, Xi'an 710061, China
| | - Shuang-xing Hou
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shan-bo Ma
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Zhang
- Department of Medical Statistics, School of Preventive Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xin Li
- Department of Urinary Urology, General Hospital of Shenyang Military Region, Shenyang 110840, China.
| |
Collapse
|
37
|
Lehmann J, Härtig W, Seidel A, Füldner C, Hobohm C, Grosche J, Krueger M, Michalski D. Inflammatory cell recruitment after experimental thromboembolic stroke in rats. Neuroscience 2014; 279:139-54. [DOI: 10.1016/j.neuroscience.2014.08.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/28/2014] [Accepted: 08/17/2014] [Indexed: 10/24/2022]
|
38
|
Deng J, Lei C, Chen Y, Fang Z, Yang Q, Zhang H, Cai M, Shi L, Dong H, Xiong L. Neuroprotective gases – Fantasy or reality for clinical use? Prog Neurobiol 2014; 115:210-45. [DOI: 10.1016/j.pneurobio.2014.01.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/03/2014] [Accepted: 01/03/2014] [Indexed: 12/17/2022]
|
39
|
Yasuda N, Ishii T, Oyama D, Fukuta T, Agato Y, Sato A, Shimizu K, Asai T, Asakawa T, Kan T, Yamada S, Ohizumi Y, Oku N. Neuroprotective effect of nobiletin on cerebral ischemia–reperfusion injury in transient middle cerebral artery-occluded rats. Brain Res 2014; 1559:46-54. [DOI: 10.1016/j.brainres.2014.02.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 01/28/2014] [Accepted: 02/03/2014] [Indexed: 12/20/2022]
|
40
|
Bartels K, Karhausen J, Clambey ET, Grenz A, Eltzschig HK. Perioperative organ injury. Anesthesiology 2014; 119:1474-89. [PMID: 24126264 DOI: 10.1097/aln.0000000000000022] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the fact that a surgical procedure may have been performed for the appropriate indication and in a technically perfect manner, patients are threatened by perioperative organ injury. For example, stroke, myocardial infarction, acute respiratory distress syndrome, acute kidney injury, or acute gut injury are among the most common causes for morbidity and mortality in surgical patients. In the current review, the authors discuss the pathogenesis of perioperative organ injury, and provide select examples for novel treatment concepts that have emerged over the past decade. Indeed, the authors are of the opinion that research to provide mechanistic insight into acute organ injury and identification of novel therapeutic approaches for the prevention or treatment of perioperative organ injury represent the most important opportunity to improve outcomes of anesthesia and surgery.
Collapse
Affiliation(s)
- Karsten Bartels
- * Fellow in Critical Care Medicine and Cardiothoracic Anesthesiology, † Assistant Professor of Anesthesiology, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina. ‡ Assistant Professor of Anesthesiology, § Associate Professor of Anesthesiology, ‖ Professor of Anesthesiology, Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado
| | | | | | | | | |
Collapse
|
41
|
Malignant Cerebral Edema After Large Anterior Circulation Infarction: A Review. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:275. [DOI: 10.1007/s11936-013-0275-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
42
|
Synthesis and protective effect of new ligustrazine-benzoic acid derivatives against CoCl2-induced neurotoxicity in differentiated PC12 cells. Molecules 2013; 18:13027-42. [PMID: 24145795 PMCID: PMC6270565 DOI: 10.3390/molecules181013027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 09/30/2013] [Accepted: 10/09/2013] [Indexed: 11/17/2022] Open
Abstract
A series of novel ligustrazine-benzoic acid derivatives were synthesized and evaluated for their protective effect against cobalt chloride-induced neurotoxicity in differentiated PC12 cells. Combining hematoxylin and eosin staining, we found compound that (3,5,6-trimethylpyrazin-2-yl)methyl 3-methoxy-4-[(3,5,6-trimethylpyrazin-2-yl)methoxy]benzoate (4a) displayed promising protective effect on the proliferation of the injured PC12 cells (EC50 = 4.249 µM). Structure-activity relationships are briefly discussed.
Collapse
|
43
|
He X, Sandhu HK, Yang Y, Hua F, Belser N, Kim DH, Xia Y. Neuroprotection against hypoxia/ischemia: δ-opioid receptor-mediated cellular/molecular events. Cell Mol Life Sci 2013; 70:2291-303. [PMID: 23014992 PMCID: PMC11113157 DOI: 10.1007/s00018-012-1167-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 09/08/2012] [Accepted: 09/10/2012] [Indexed: 12/24/2022]
Abstract
Hypoxic/ischemic injury remains the most dreaded cause of neurological disability and mortality. Despite the humbling experiences due to lack of promising therapy, our understanding of the complex cascades underlying the neuronal insult has led to advances in basic science research. One of the most noteworthy has been the effect of opioid receptors, especially the delta-opioid receptor (DOR), on hypoxic/ischemic neurons. Our recent studies, and those of others worldwide, present strong evidence that sheds light on DOR-mediated neuroprotection in the brain, especially in the cortex. The mechanisms of DOR neuroprotection are broadly categorized as: (1) stabilization of the ionic homeostasis, (2) inhibition of excitatory transmitter release, (3) attenuation of disrupted neuronal transmission, (4) increase in antioxidant capacity, (5) regulation of intracellular pathways-inhibition of apoptotic signals and activation of pro-survival signaling, (6) regulation of specific gene and protein expression, and (7) up-regulation of endogenous opioid release and/or DOR expression. Depending upon the severity and duration of hypoxic/ischemic insult, the release of endogenous opioids and DOR expression are regulated in response to the stress, and DOR signaling acts at multiple levels to confer neuronal tolerance to harmful insult. The phenomenon of DOR neuroprotection offers a potential clue for a promising target that may have significant clinical implications in our quest for neurotherapeutics.
Collapse
Affiliation(s)
- Xiaozhou He
- The Third Clinical College of Suzhou University, Changzhou, Jiangsu China
| | - Harleen K. Sandhu
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| | - Yilin Yang
- The Third Clinical College of Suzhou University, Changzhou, Jiangsu China
| | - Fei Hua
- The Third Clinical College of Suzhou University, Changzhou, Jiangsu China
| | - Nathalee Belser
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| | - Dong H. Kim
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| | - Ying Xia
- The Vivian L Smith Department of Neurosurgery, The University of Texas Medical School at Houston, Houston, 77030 TX USA
| |
Collapse
|
44
|
Krueger M, Härtig W, Reichenbach A, Bechmann I, Michalski D. Blood-brain barrier breakdown after embolic stroke in rats occurs without ultrastructural evidence for disrupting tight junctions. PLoS One 2013; 8:e56419. [PMID: 23468865 PMCID: PMC3582567 DOI: 10.1371/journal.pone.0056419] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 01/09/2013] [Indexed: 12/27/2022] Open
Abstract
The term blood-brain barrier (BBB) relates to the ability of cerebral vessels to hold back hydrophilic and large molecules from entering the brain, thereby crucially contributing to brain homeostasis. In fact, experimental opening of endothelial tight junctions causes a breakdown of the BBB evidenced as for instance by albumin leakage. This and similar observations led to the conclusion that BBB breakdown is predominantly mediated by damage to tight junction complexes, but evidentiary ultrastructural data are rare. Since functional deficits of the BBB contribute to an increased risk of hemorrhagic transformation and brain edema after stroke, which both critically impact on the clinical outcome, we studied the mechanism of BBB breakdown using an embolic model of focal cerebral ischemia in Wistar rats to closely mimic the essential human pathophysiology. Ischemia-induced BBB breakdown was detected using intravenous injection of FITC-albumin and tight junctions in areas of FITC-albumin extravasation were subsequently studied using fluorescence and electron microscopy. Against our expectation, 25 hours after ischemia induction the morphology of tight junction complexes (identified ultrastructurally and using antibodies against the transcellular proteins occludin and claudin-5) appeared to be regularly maintained in regions where FITC-albumin massively leaked into the neuropil. Furthermore, occludin signals along pan-laminin-labeled vessels in the affected hemisphere equaled the non-affected contralateral side (ratio: 0.966 vs. 0.963; P = 0.500). Additional ultrastructural analyses at 5 and 25 h after ischemia induction clearly indicated FITC-albumin extravasation around vessels with intact tight junctions, while the endothelium exhibited enhanced transendothelial vesicle trafficking and signs of degeneration. Thus, BBB breakdown and leakage of FITC-albumin cannot be correlated with staining patterns for common tight junction proteins alone. Understanding the mechanisms causing functional endothelial alterations and endothelial damage is likely to provide novel protective targets in stroke.
Collapse
Affiliation(s)
- Martin Krueger
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
- * E-mail: (MK); (DM)
| | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Dominik Michalski
- Department of Neurology, University of Leipzig, Leipzig, Germany
- * E-mail: (MK); (DM)
| |
Collapse
|
45
|
Ishii T, Fukuta T, Agato Y, Oyama D, Yasuda N, Shimizu K, Kawaguchi AT, Asai T, Oku N. Nanoparticles accumulate in ischemic core and penumbra region even when cerebral perfusion is reduced. Biochem Biophys Res Commun 2013; 430:1201-5. [DOI: 10.1016/j.bbrc.2012.12.080] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 12/14/2012] [Indexed: 01/12/2023]
|
46
|
Michalski D, Härtig W, Krügel K, Edwards RH, Böddener M, Böhme L, Pannicke T, Reichenbach A, Grosche A. Region-specific expression of vesicular glutamate and GABA transporters under various ischaemic conditions in mouse forebrain and retina. Neuroscience 2012; 231:328-44. [PMID: 23219666 DOI: 10.1016/j.neuroscience.2012.11.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 11/19/2012] [Accepted: 11/25/2012] [Indexed: 10/27/2022]
Abstract
There is accumulating evidence that glutamate and GABA release are key mechanisms of ischaemic events in the CNS. However, data on the expression of involved transporters for these mediators are inconsistent, potentially impeding further neuroprotective approaches. Here, we applied immunofluorescence labelling to characterise the expression pattern of vesicular glutamate (VGLUT) and GABA transporters (VGAT) after acute focal cerebral ischaemia and in two models of retinal ischaemia. Mice were subjected to filament-based focal cerebral ischaemia predominantly involving the middle cerebral artery territory, also leading to retinal ischaemia due to central retinal artery occlusion (CRAO). Alternatively, retinal ischaemia was induced by a transient increase of the intraocular pressure (HIOP). One day after ischaemia onset, diminished immunolabelling of neuronal nuclei and microtubule-associated protein 2-positive structures were found in the ipsilateral neocortex, subcortex and the retina, indicating neuronal degeneration. VGLUT1 expression did not change significantly in ischaemic tissues whereas VGLUT2 was down-regulated in specific areas of the brain. VGLUT3 expression was only slightly down-regulated in the ischaemia-affected neocortex, and was found to form clusters on fibrils of unknown origin in the ischaemic lateral hypothalamus. In contrast, retinae subjected to CRAO or HIOP displayed a rapid loss of VGLUT3-immunoreactivity. The expression of VGAT appears resistant to ischaemia as there was no significant alteration in all the regions analysed. In summary, these data indicate a region- and subtype-specific change of VGLUT expression in the ischaemia-affected CNS, whose consideration might help to generate specific neuroprotective strategies.
Collapse
Affiliation(s)
- D Michalski
- Department of Neurology, University of Leipzig, Liebigstr. 20, 04103 Leipzig, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Krol S, Macrez R, Docagne F, Defer G, Laurent S, Rahman M, Hajipour MJ, Kehoe PG, Mahmoudi M. Therapeutic Benefits from Nanoparticles: The Potential Significance of Nanoscience in Diseases with Compromise to the Blood Brain Barrier. Chem Rev 2012; 113:1877-903. [DOI: 10.1021/cr200472g] [Citation(s) in RCA: 158] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Silke Krol
- Fondazione IRCCS Institute of Neurology “Carlo Besta”, Milan, Italy
| | - Richard Macrez
- Inserm U919, University Caen Basse Normandie, Serine Proteases and Pathophysiology of the Neurovascular Unit, GIP CYCERON, F-14074 Caen, France
- Department of Neurology, University Hospital of Caen, Caen, France
| | - Fabian Docagne
- Inserm U919, University Caen Basse Normandie, Serine Proteases and Pathophysiology of the Neurovascular Unit, GIP CYCERON, F-14074 Caen, France
| | - Gilles Defer
- Inserm U919, University Caen Basse Normandie, Serine Proteases and Pathophysiology of the Neurovascular Unit, GIP CYCERON, F-14074 Caen, France
- Department of Neurology, University Hospital of Caen, Caen, France
| | - Sophie Laurent
- Department of General, Organic, and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau, 19, B-7000 Mons, Belgium
| | - Masoud Rahman
- Laboratory of NanoBio Interactions , Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad J. Hajipour
- Laboratory of NanoBio Interactions , Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Patrick G. Kehoe
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, John James Laboratories, Frenchay Hospital, Bristol, U.K
| | - Morteza Mahmoudi
- Laboratory of NanoBio Interactions , Department of Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Current address: School of Chemical Sciences, University of Illinois at Urbana−Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
| |
Collapse
|
48
|
Huang HF, Guo F, Cao YZ, Shi W, Xia Q. Neuroprotection by manganese superoxide dismutase (MnSOD) mimics: antioxidant effect and oxidative stress regulation in acute experimental stroke. CNS Neurosci Ther 2012; 18:811-8. [PMID: 22934841 DOI: 10.1111/j.1755-5949.2012.00380.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 05/27/2012] [Accepted: 06/01/2012] [Indexed: 01/20/2023] Open
Abstract
AIMS Manganese superoxide dismutase (MnSOD), one of the most crucial antioxidant enzymes in the central nervous system, is thought to be one of the major mechanisms by which cells counteract the injuries of reactive oxygen species after cerebral ischemia. In this study, we used a novel synthesized compound (MnTm4PyP) with highly effective superoxide dismutase activity to study the therapeutic potential of MnSOD and the possible underlying mechanisms in cerebral ischemia. METHODS Primary cultured cortical neurons were used to examine the protective effect of the compounds. Mice with middle cerebral artery occlusion were used as ischemic stroke animal model. Animals were pretreated with MnTm4PyP intravenously 30 min before surgery. At 24 h after surgery, neurological behavior and histological function were observed. Infarcted cortex tissues and cultured neurons were collected for investigation of the oxidative stress signaling pathways. RESULTS In vitro studies revealed that MnSOD mimic MnTm4PyP pretreatment significantly increased viability of neurons after injury by H(2) O(2) . Intracellular superoxide radical levels were eliminated. In vivo experiments demonstrated MnTm4PyP pretreatment reduced infarct volume and improved neurological function. The MnSOD mimic alleviated oxidative stress and apoptosis. CONCLUSION MnSOD is an effective therapeutic target in ischemic stroke prevention because of its antioxidant effects and oxidative stress regulation.
Collapse
Affiliation(s)
- Hai-Feng Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | | | | | | | | |
Collapse
|
49
|
Zeng XN, Xie LL, Liang R, Sun XL, Fan Y, Hu G. AQP4 knockout aggravates ischemia/reperfusion injury in mice. CNS Neurosci Ther 2012; 18:388-94. [PMID: 22533723 DOI: 10.1111/j.1755-5949.2012.00308.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The glial water channel aquaporin-4 (AQP4) has been shown to be involved in a wide range of brain disorders. Although its important role in stroke has already been documented, the underlying mechanism was not clarified yet. Therefore, this study was designed to investigate the impacts of AQP4 deletion in ischemia/reperfusion (I/R). METHODS AND RESULTS Herein we found a higher mortality and more severe neurological deficits in AQP4 knockout (AQP4(-/-)) mice after transient middle cerebral artery occlusion while no difference was observed in water content variation during I/R between two genotypes except a higher basal water content developed in AQP4(-/-) mouse brain, implying the same increment of water content over a higher basal level may provoke an even more elevated intracranial pressure, which might be an important cause of increased mortality in AQP4(-/-) mice. Moreover, AQP4 knockout aggravated I/R injury with enlarged infarct size and a more serious loss of CA1 neurons accompanied by a striking hypertrophy of astrocytes, suggesting an involvement of AQP4 in astrocytic dysfunction. CONCLUSIONS Our findings provide direct evidence that AQP4 plays a crucial role in the pathogenesis of I/R injury, which may confer a new option for stroke treatment.
Collapse
Affiliation(s)
- Xiao-Ning Zeng
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, China
| | | | | | | | | | | |
Collapse
|
50
|
Michalski D, Weise C, Hobohm C, Küppers-Tiedt L, Pelz J, Schneider D, Kacza J, Härtig W. Autonomic reactions and peri-interventional alterations in body weight as potential supplementary outcome parameters for thromboembolic stroke in rats. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2012; 4:7. [PMID: 22510241 PMCID: PMC3398859 DOI: 10.1186/2040-7378-4-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 04/17/2012] [Indexed: 12/02/2022]
Abstract
Background Since several neuroprotectives failed to reproduce promising preclinical results under clinical conditions, efforts emerged to implement clinically relevant endpoints in animal stroke studies. Thereby, insufficient attention was given on autonomic reactions due to experimental stroke, although clinical trials reported on high functional and prognostic impact. This study focused on autonomic consequences and body weight changes in a translational relevant stroke model and investigated interrelations to different outcome measurements. Methods Forty-eight rats underwent thromboembolic middle cerebral artery occlusion (MCAO) while recording heart rate (HR) and mean arterial pressure (MAP). After assessing early functional impairment (Menzies score), animals were assigned to control procedure or potentially neuroprotective treatment with normobaric (NBO) or hyperbaric oxygen (HBO). Four or 24 hours after ischemia onset, functional impairment was re-assessed and FITC-albumin administered intravenously obtaining leakage-related blood–brain barrier (BBB) impairment. Body weight was documented prior to MCAO and 4 or 24 hours after ischemia onset. Results During MCAO, HR was found to increase significantly while MAP decreased. The amount of changes in HR was positively correlated with early functional impairment (P = 0.001): Severely affected animals provided an increase of 15.2 compared to 0.8 beats/minute in rats with low impairment (P = 0.048). Regarding body weight, a decrease of 9.4% within 24 hours after MCAO occurred, but treatment-specific alterations showed no significant correlations with respective functional or BBB impairment. Conclusions Future studies should routinely include autonomic parameters to allow inter-group comparisons and better understanding of autonomic reactions due to experimental stroke. Prospectively, autonomic consequences might represent a useful outcome parameter enhancing the methodological spectrum of preclinical stroke studies.
Collapse
Affiliation(s)
- Dominik Michalski
- Department of Neurology, University of Leipzig, Liebigstr, 20, 04103, Leipzig, Germany.
| | | | | | | | | | | | | | | |
Collapse
|