1
|
Zhu Y, Li X, Lei X, Tang L, Wen D, Zeng B, Zhang X, Huang Z, Guo Z. The potential mechanism and clinical application value of remote ischemic conditioning in stroke. Neural Regen Res 2025; 20:1613-1627. [PMID: 38845225 DOI: 10.4103/nrr.nrr-d-23-01800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/19/2024] [Indexed: 08/07/2024] Open
Abstract
Some studies have confirmed the neuroprotective effect of remote ischemic conditioning against stroke. Although numerous animal researches have shown that the neuroprotective effect of remote ischemic conditioning may be related to neuroinflammation, cellular immunity, apoptosis, and autophagy, the exact underlying molecular mechanisms are unclear. This review summarizes the current status of different types of remote ischemic conditioning methods in animal and clinical studies and analyzes their commonalities and differences in neuroprotective mechanisms and signaling pathways. Remote ischemic conditioning has emerged as a potential therapeutic approach for improving stroke-induced brain injury owing to its simplicity, non-invasiveness, safety, and patient tolerability. Different forms of remote ischemic conditioning exhibit distinct intervention patterns, timing, and application range. Mechanistically, remote ischemic conditioning can exert neuroprotective effects by activating the Notch1/phosphatidylinositol 3-kinase/Akt signaling pathway, improving cerebral perfusion, suppressing neuroinflammation, inhibiting cell apoptosis, activating autophagy, and promoting neural regeneration. While remote ischemic conditioning has shown potential in improving stroke outcomes, its full clinical translation has not yet been achieved.
Collapse
Affiliation(s)
- Yajun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Huang J, Shen Q, Wang Z, Ni S, Sun F, Hua Y, Huang J. The influence of the NRG1/ERBB4 signaling pathway on pulmonary artery endothelial cells. Pulm Circ 2024; 14:e12439. [PMID: 39411231 PMCID: PMC11475022 DOI: 10.1002/pul2.12439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
This study aimed to examine the influence of the Neuregulin-1 (NRG1)/ERBB4 signaling pathway on the function of human pulmonary artery endothelial cells (HPAECs) and investigate the underlying mechanisms. Enzyme-linked immunosorbent assay indicated that ERBB4 levels in the serum of patients with pulmonary embolism (PE) were significantly higher than those of healthy controls (p < 0.05). In cellular studies, thrombin stimulation for 6 h led to a significant decrease in cell viability and overexpression of ERBB4 compared to control (p < 0.05). In the NRG1 group, apoptosis of HPAECs was reduced (p < 0.05), accompanied by a decrease in ERBB4 expression and an increase in p-ERBB4, phosphorylated serine/threonine kinase proteins (Akt) (p-Akt), and p-phosphoinositide 3-kinase (PI3K) expression (p < 0.05). In the AG1478 group, there was a significant increase in HPAEC apoptosis and a significant decrease in p-ERBB4 and ERBB4 expression compared to the Con group (p < 0.05). In the AG1478 + NRG1 group, there was an increase in the apoptosis rate and a significant decrease in the expression of p-ERBB4, ERBB4, p-Akt, and phosphorylated PI3K compared to the NRG1 group (p < 0.05). In animal studies, the PE group showed an increase in the expression of ERBB4 and p-ERBB4 compared to the Con group (p < 0.05). NRG1 treatment led to a significant reduction in embolism severity with decreased ERBB4 expression and increased p-ERBB4 expression (p < 0.05). Gene set enrichment analysis identified five pathways that were significantly associated with high ERBB4 expression, including CHOLESTEROL HOMEOSTASIS, OXIDATIVE PHOSPHORYLATION, and FATTY ACID METABOLISM (p < 0.05). Therefore, NRG1 inhibits apoptosis of HPAECs, accompanied by a decrease in ERBB4 and an increase in p-ERBB4. NRG1 inhibition in HPAECs apoptosis can be partially reversed by inhibiting ERBB4 expression with AG1478. ERBB4 has the potential to be a novel biological marker of PE.
Collapse
Affiliation(s)
- Jin‐Bo Huang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Qin Shen
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Zhi‐Qi Wang
- Jiangnan University Affiliated Wuxi Fifth People's HospitalWuxiJiangsuChina
| | - Song‐Shi Ni
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Fei Sun
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Yun Hua
- Department of Pulmonary and Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Jian‐An Huang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
3
|
Neuroprotective effects of donepezil against Aβ25-35-induced neurotoxicity. Eur J Med Res 2022; 27:219. [PMID: 36307893 PMCID: PMC9617393 DOI: 10.1186/s40001-022-00862-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 10/20/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to investigate the neuroprotective effect of donepezil against β-amyloid25-35 (Aβ25-35)-induced neurotoxicity and the possible mechanism. Methods PC12 cells were conventionally cultured. Serial concentrations of Aβ25-35 and donepezil (0, 0.5, 1, 5, 10, 20 and 50 μmol/L) were added to the PC12 cells, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) staining was performed to detect the effects of these treatments on PC 12 viability. The PC 12 cells were pretreated with 1, 5, 10, 20 or 50 μmol/L donepezil two hours before 20 μmol/L Aβ25-35 was added to pretreatment groups A, B, C, D and E. Normal control group I and the 20 μmol/L Aβ25-35-treated group were selected. An MTT assay was used to detect PC12 cell viability, and the level of lactate dehydrogenase (LDH) was determined. PC12 cells were pretreated with 10 μmol/L GF109203X (a protein kinase C [PKC] antagonist) 30 min before 10 μmol/L donepezil was added to pretreatment group F, and normal control group II, the 10 μmol/L GF109203X-treated group and the 10 μmol/L donepezil-treated group were chosen. The expression of phosphorylation-PKC (P-PKC) and its major substrate phosphorylated myristoylated alanine-rich protein C kinase substrate (P-MARCKS) was measured by Western blotting. The effects of donepezil on the subcellular distribution of the PKCα and PKCε isoforms were detected by immunofluorescence staining. Results Treatment with Aβ25-35 (5, 10, 20 or 50 μmol/L) for 24 h significantly (P < 0.05) decreased PC 12 cell viability in a dose-dependent manner. Compared with the PC12 cells in the control group, those in the 20 μmol/L Aβ25-35-treated group exhibited lower viability but higher LDH release. Compared with the 20 μmol/L Aβ25–35-treated group, pretreatment groups B, C, D and E exhibited significantly (P < 0.05) increased cell viability but significantly (P < 0.05) decreased LDH release. Western blotting demonstrated that compared with control, 10 μmol/L donepezil promoted PKC and MARCKS phosphorylation and that the expression of P-PKC and P-MARCKS in pretreatment group F was significantly (P < 0.05) lower than that in the donepezil-treated group. Immunofluorescence staining revealed that the PKCα and PKCε isoforms were located mainly in the cytoplasm of PC12 control cells, whereas donepezil increased the expression of the PKCα and PKCε isoforms in the membrane fraction. The Western blot results showed that donepezil altered the subcellular distribution of the PKCα and PKCε isoforms by decreasing their expression in the cytosolic fraction but increasing their expression in the membrane fraction. Conclusion Donepezil can antagonize Aβ25–350-induced neurotoxicity in PC 12 cells, and PKC activation may account for the neuroprotective effect of donepezil.
Collapse
|
4
|
Ghozy S, Reda A, Varney J, Elhawary AS, Shah J, Murry K, Sobeeh MG, Nayak SS, Azzam AY, Brinjikji W, Kadirvel R, Kallmes DF. Neuroprotection in Acute Ischemic Stroke: A Battle Against the Biology of Nature. Front Neurol 2022; 13:870141. [PMID: 35711268 PMCID: PMC9195142 DOI: 10.3389/fneur.2022.870141] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Stroke is the second most common cause of global death following coronary artery disease. Time is crucial in managing stroke to reduce the rapidly progressing insult of the ischemic penumbra and the serious neurologic deficits that might follow it. Strokes are mainly either hemorrhagic or ischemic, with ischemic being the most common of all types of strokes. Thrombolytic therapy with recombinant tissue plasminogen activator and endovascular thrombectomy are the main types of management of acute ischemic stroke (AIS). In addition, there is a vital need for neuroprotection in the setting of AIS. Neuroprotective agents are important to investigate as they may reduce mortality, lessen disability, and improve quality of life after AIS. In our review, we will discuss the main types of management and the different modalities of neuroprotection, their mechanisms of action, and evidence of their effectiveness after ischemic stroke.
Collapse
Affiliation(s)
- Sherief Ghozy
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States.,Nuffield Department of Primary Care Health Sciences and Department for Continuing Education (EBHC Program), Oxford University, Oxford, United Kingdom
| | - Abdullah Reda
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Joseph Varney
- School of Medicine, American University of the Caribbean, Philipsburg, Sint Maarten
| | | | - Jaffer Shah
- Medical Research Center, Kateb University, Kabul, Afghanistan
| | | | - Mohamed Gomaa Sobeeh
- Faculty of Physical Therapy, Sinai University, Cairo, Egypt.,Faculty of Physical Therapy, Cairo University, Giza, Egypt
| | - Sandeep S Nayak
- Department of Internal Medicine, NYC Health + Hospitals/Metropolitan, New York, NY, United States
| | - Ahmed Y Azzam
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Waleed Brinjikji
- Department of Neurosurgery, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - David F Kallmes
- Department of Neuroradiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
5
|
Sheng R, Chen JL, Qin ZH. Cerebral conditioning: Mechanisms and potential clinical implications. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
6
|
Liu J, Gu Y, Guo M, Ji X. Neuroprotective effects and mechanisms of ischemic/hypoxic preconditioning on neurological diseases. CNS Neurosci Ther 2021; 27:869-882. [PMID: 34237192 PMCID: PMC8265941 DOI: 10.1111/cns.13642] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/20/2022] Open
Abstract
As the organ with the highest demand for oxygen, the brain has a poor tolerance to ischemia and hypoxia. Despite severe ischemia/hypoxia induces the occurrence and development of various central nervous system (CNS) diseases, sublethal insult may induce strong protection against subsequent fatal injuries by improving tolerance. Searching for potential measures to improve brain ischemic/hypoxic is of great significance for treatment of ischemia/hypoxia related CNS diseases. Ischemic/hypoxic preconditioning (I/HPC) refers to the approach to give the body a short period of mild ischemic/hypoxic stimulus which can significantly improve the body's tolerance to subsequent more severe ischemia/hypoxia event. It has been extensively studied and been considered as an effective therapeutic strategy in CNS diseases. Its protective mechanisms involved multiple processes, such as activation of hypoxia signaling pathways, anti-inflammation, antioxidant stress, and autophagy induction, etc. As a strategy to induce endogenous neuroprotection, I/HPC has attracted extensive attention and become one of the research frontiers and hotspots in the field of neurotherapy. In this review, we discuss the basic and clinical research progress of I/HPC on CNS diseases, and summarize its mechanisms. Furthermore, we highlight the limitations and challenges of their translation from basic research to clinical application.
Collapse
Affiliation(s)
- Jia Liu
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Yakun Gu
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Mengyuan Guo
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China
| | - Xunming Ji
- Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Capital Medical University, Beijing, China.,Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Boltze J, Aronowski JA, Badaut J, Buckwalter MS, Caleo M, Chopp M, Dave KR, Didwischus N, Dijkhuizen RM, Doeppner TR, Dreier JP, Fouad K, Gelderblom M, Gertz K, Golubczyk D, Gregson BA, Hamel E, Hanley DF, Härtig W, Hummel FC, Ikhsan M, Janowski M, Jolkkonen J, Karuppagounder SS, Keep RF, Koerte IK, Kokaia Z, Li P, Liu F, Lizasoain I, Ludewig P, Metz GAS, Montagne A, Obenaus A, Palumbo A, Pearl M, Perez-Pinzon M, Planas AM, Plesnila N, Raval AP, Rueger MA, Sansing LH, Sohrabji F, Stagg CJ, Stetler RA, Stowe AM, Sun D, Taguchi A, Tanter M, Vay SU, Vemuganti R, Vivien D, Walczak P, Wang J, Xiong Y, Zille M. New Mechanistic Insights, Novel Treatment Paradigms, and Clinical Progress in Cerebrovascular Diseases. Front Aging Neurosci 2021; 13:623751. [PMID: 33584250 PMCID: PMC7876251 DOI: 10.3389/fnagi.2021.623751] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
The past decade has brought tremendous progress in diagnostic and therapeutic options for cerebrovascular diseases as exemplified by the advent of thrombectomy in ischemic stroke, benefitting a steeply increasing number of stroke patients and potentially paving the way for a renaissance of neuroprotectants. Progress in basic science has been equally impressive. Based on a deeper understanding of pathomechanisms underlying cerebrovascular diseases, new therapeutic targets have been identified and novel treatment strategies such as pre- and post-conditioning methods were developed. Moreover, translationally relevant aspects are increasingly recognized in basic science studies, which is believed to increase their predictive value and the relevance of obtained findings for clinical application.This review reports key results from some of the most remarkable and encouraging achievements in neurovascular research that have been reported at the 10th International Symposium on Neuroprotection and Neurorepair. Basic science topics discussed herein focus on aspects such as neuroinflammation, extracellular vesicles, and the role of sex and age on stroke recovery. Translational reports highlighted endovascular techniques and targeted delivery methods, neurorehabilitation, advanced functional testing approaches for experimental studies, pre-and post-conditioning approaches as well as novel imaging and treatment strategies. Beyond ischemic stroke, particular emphasis was given on activities in the fields of traumatic brain injury and cerebral hemorrhage in which promising preclinical and clinical results have been reported. Although the number of neutral outcomes in clinical trials is still remarkably high when targeting cerebrovascular diseases, we begin to evidence stepwise but continuous progress towards novel treatment options. Advances in preclinical and translational research as reported herein are believed to have formed a solid foundation for this progress.
Collapse
Affiliation(s)
- Johannes Boltze
- School of Life Sciences, University of Warwick, Warwick, United Kingdom
| | - Jaroslaw A. Aronowski
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jerome Badaut
- NRS UMR 5287, INCIA, Brain Molecular Imaging Team, University of Bordeaux, Bordeaux cedex, France
| | - Marion S. Buckwalter
- Departments of Neurology and Neurological Sciences, and Neurosurgery, Wu Tsai Neurosciences Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Mateo Caleo
- Neuroscience Institute, National Research Council, Pisa, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nadine Didwischus
- School of Life Sciences, University of Warwick, Warwick, United Kingdom
| | - Rick M. Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Thorsten R. Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens P. Dreier
- Department of Neurology, Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Karim Fouad
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta, Edmonton, AB, Canada
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karen Gertz
- Department of Neurology, Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Barbara A. Gregson
- Neurosurgical Trials Group, Institute of Neuroscience, The University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Daniel F. Hanley
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, United States
| | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Friedhelm C. Hummel
- Clinical Neuroengineering, Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology Valais, Clinique Romande de Réadaptation, Sion, Switzerland
- Clinical Neuroscience, University of Geneva Medical School, Geneva, Switzerland
| | - Maulana Ikhsan
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, United States
| | - Jukka Jolkkonen
- Department of Neurology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Saravanan S. Karuppagounder
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Inga K. Koerte
- Psychiatric Neuroimaging Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Child and Adolescent Psychiatry, Psychosomatic, and Psychotherapy, Ludwig Maximilians University, Munich, Germany
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Madrid, Spain
| | - Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerlinde A. S. Metz
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Alex Palumbo
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| | - Monica Pearl
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Miguel Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anna M. Planas
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Àrea de Neurociències, Barcelona, Spain
- Department d’Isquèmia Cerebral I Neurodegeneració, Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich University Hospital, Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maria A. Rueger
- Faculty of Medicine and University Hospital, Department of Neurology, University of Cologne, Cologne, Germany
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Farida Sohrabji
- Women’s Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX, United States
| | - Charlotte J. Stagg
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, United Kingdom
| | - R. Anne Stetler
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ann M. Stowe
- Department of Neurology and Neurotherapeutics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Dandan Sun
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, PA, United States
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Mickael Tanter
- Institute of Physics for Medicine Paris, INSERM U1273, ESPCI Paris, CNRS FRE 2031, PSL University, Paris, France
| | - Sabine U. Vay
- Faculty of Medicine and University Hospital, Department of Neurology, University of Cologne, Cologne, Germany
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, United States
| | - Denis Vivien
- UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Normandy University, Caen, France
- CHU Caen, Clinical Research Department, CHU de Caen Côte de Nacre, Caen, France
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, United States
| | - Jian Wang
- Department of Human Anatomy, College of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, United States
| | - Marietta Zille
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
8
|
NRG1-ErbB4 signaling promotes functional recovery in a murine model of traumatic brain injury via regulation of GABA release. Exp Brain Res 2019; 237:3351-3362. [PMID: 31720762 DOI: 10.1007/s00221-019-05680-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/09/2019] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is a serious health problem in the world. However, little is known about the pathogenesis and molecular mechanisms of TBI. Here, we show that TBI activates neuregulin 1 (NRG1)-ErbB4 signaling, with an increased expression of NRG1 and ErbB4 in the traumatic region. Specifically knocking out ErbB4 in parvalbumin-positive (PV+) interneurons exacerbates motor function deficits in mice after TBI. Consistently, PV-ErbB4-/- mice showed larger necrotic area and more edema when compared with PV-ErbB4+/+ mice. Replenishment of NRG1 through intranasal application of the recombinant protein in PV-ErbB4+/+ mice enhanced neurological function. Moreover, using an in vitro neuronal culture system, we found that NRG1-ErbB4 signaling protects neurons from glutamate-induced death, and such protective effects could be diminished by GABA receptor antagonist. These results indicate that NRG-ErbB4 signaling protects cortical neurons from TBI-induced damage, and such effect is probably mediated by promoting GABA activity. Taken together, these findings unveil a previously unappreciated role for NRG1-ErB4 signaling in preventing neuronal cell death during functional recovery after TBI.
Collapse
|
9
|
Pastore D, Pacifici F, Dave KR, Palmirotta R, Bellia A, Pasquantonio G, Guadagni F, Donadel G, Di Daniele N, Abete P, Lauro D, Rundek T, Perez-Pinzon MA, Della-Morte D. Age-Dependent Levels of Protein Kinase Cs in Brain: Reduction of Endogenous Mechanisms of Neuroprotection. Int J Mol Sci 2019; 20:E3544. [PMID: 31331067 PMCID: PMC6678180 DOI: 10.3390/ijms20143544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are among the leading causes of mortality and disability worldwide. However, current therapeutic approaches have failed to reach significant results in their prevention and cure. Protein Kinase Cs (PKCs) are kinases involved in the pathophysiology of neurodegenerative diseases, such as Alzheimer's Disease (AD) and cerebral ischemia. Specifically ε, δ, and γPKC are associated with the endogenous mechanism of protection referred to as ischemic preconditioning (IPC). Existing modulators of PKCs, in particular of εPKC, such as ψεReceptor for Activated C-Kinase (ψεRACK) and Resveratrol, have been proposed as a potential therapeutic strategy for cerebrovascular and cognitive diseases. PKCs change in expression during aging, which likely suggests their association with IPC-induced reduction against ischemia and increase of neuronal loss occurring in senescent brain. This review describes the link between PKCs and cerebrovascular and cognitive disorders, and proposes PKCs modulators as innovative candidates for their treatment. We report original data showing εPKC reduction in levels and activity in the hippocampus of old compared to young rats and a reduction in the levels of δPKC and γPKC in old hippocampus, without a change in their activity. These data, integrated with other findings discussed in this review, demonstrate that PKCs modulators may have potential to restore age-related reduction of endogenous mechanisms of protection against neurodegeneration.
Collapse
Affiliation(s)
- Donatella Pastore
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Francesca Pacifici
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Kunjan R Dave
- Department of Neurology, The Evelyn McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Alfonso Bellia
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Policlinico Tor Vergata Foundation, University Hospital, 00133 Rome, Italy
| | - Guido Pasquantonio
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Fiorella Guadagni
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Giulia Donadel
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Policlinico Tor Vergata Foundation, University Hospital, 00133 Rome, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, University of Naples, Federico II, 80138 Naples, Italy
| | - Davide Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Policlinico Tor Vergata Foundation, University Hospital, 00133 Rome, Italy
| | - Tatjana Rundek
- Department of Neurology, The Evelyn McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology, The Evelyn McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
- Department of Neurology, The Evelyn McKnight Brain Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
10
|
Khoury N, Xu J, Stegelmann SD, Jackson CW, Koronowski KB, Dave KR, Young JI, Perez-Pinzon MA. Resveratrol Preconditioning Induces Genomic and Metabolic Adaptations within the Long-Term Window of Cerebral Ischemic Tolerance Leading to Bioenergetic Efficiency. Mol Neurobiol 2018; 56:4549-4565. [PMID: 30343466 DOI: 10.1007/s12035-018-1380-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/04/2018] [Indexed: 01/23/2023]
Abstract
Neuroprotective agents administered post-cerebral ischemia have failed so far in the clinic to promote significant recovery. Thus, numerous efforts were redirected toward prophylactic approaches such as preconditioning as an alternative therapeutic strategy. Our laboratory has revealed a novel long-term window of cerebral ischemic tolerance mediated by resveratrol preconditioning (RPC) that lasts for 2 weeks in mice. To identify its mediators, we conducted an RNA-seq experiment on the cortex of mice 2 weeks post-RPC, which revealed 136 differentially expressed genes. The majority of genes (116/136) were downregulated upon RPC and clustered into biological processes involved in transcription, synaptic signaling, and neurotransmission. The downregulation in these processes was reminiscent of metabolic depression, an adaptation used by hibernating animals to survive severe ischemic states by downregulating energy-consuming pathways. Thus, to assess metabolism, we used a neuronal-astrocytic co-culture model and measured the cellular respiration rate at the long-term window post-RPC. Remarkably, we observed an increase in glycolysis and mitochondrial respiration efficiency upon RPC. We also observed an increase in the expression of genes involved in pyruvate uptake, TCA cycle, and oxidative phosphorylation, all of which indicated an increased reliance on energy-producing pathways. We then revealed that these nuclear and mitochondrial adaptations, which reduce the reliance on energy-consuming pathways and increase the reliance on energy-producing pathways, are epigenetically coupled through acetyl-CoA metabolism and ultimately increase baseline ATP levels. This increase in ATP would then allow the brain, a highly metabolic organ, to endure prolonged durations of energy deprivation encountered during cerebral ischemia.
Collapse
Affiliation(s)
- Nathalie Khoury
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Jing Xu
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Samuel D Stegelmann
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA
| | - Charles W Jackson
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Kevin B Koronowski
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Juan I Young
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,John P. Hussman Institute for Human Genomics, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Human Genetics, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA. .,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA. .,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
11
|
Landucci E, Llorente IL, Anuncibay-Soto B, Pellegrini-Giampietro DE, Fernández-López A. Bicuculline Reverts the Neuroprotective Effects of Meloxicam in an Oxygen and Glucose Deprivation (OGD) Model of Organotypic Hippocampal Slice Cultures. Neuroscience 2018; 386:68-78. [PMID: 29949743 DOI: 10.1016/j.neuroscience.2018.06.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/25/2018] [Accepted: 06/18/2018] [Indexed: 01/17/2023]
Abstract
We previously demonstrated that the non-steroidal anti-inflammatory agent meloxicam has neuroprotective effects in an oxygen and glucose deprivation model (OGD) of rat organotypic hippocampal slice cultures. We wondered if GABAergic transmission changed the neuroprotective effects of meloxicam and if meloxicam was able to modulate endoplasmic reticulum stress (ER stress) in this model. Mortality was measured using propidium iodide. Western blot assays were performed to measure levels of cleaved and non-cleaved caspase-3 to quantify apoptosis, while levels of GRP78, GRP94 and phosphorylated eIF2α were used to detect unfolded protein response (UPR). Transcript levels of GRP78, GRP94 and GABAergic receptor α, β, and γ subunits were measured by real-time quantitative polymerase chain reaction (qPCR). In the present study, we show that the presence of meloxicam in a 30 min OGD assay, followed by 24 h of normoxic conditions, presented an antiapoptotic effect. The simultaneous presence of the GABAA receptor antagonist, bicuculline, in combination with meloxicam blocked the neuroprotective effect provided by the latter. However, in light of its effects on caspase 3 and PARP, bicuculline did not seem to promote the apoptotic pathway. Our results also showed that meloxicam modified the unfolded protein response (UPR), as well as the transcriptional response of different genes, including the GABAA receptor, alpha1, beta3 and gamma2 subunits. We concluded that meloxicam has a neuroprotective anti-apoptotic action, is able to enhance the UPR independently of the systemic anti-inflammatory response and its neuroprotective effect can be inhibited by blocking GABAA receptors.
Collapse
Affiliation(s)
- Elisa Landucci
- Sezione di Farmacologia Clinica e Oncologia, Dipartimento di Scienze della Salute, Università di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Irene L Llorente
- Neurology Department, David Geffen School of Medicine, University of California, Los Angeles, USA.
| | - Berta Anuncibay-Soto
- Área de Biología Celular, Instituto de Biomedicina, Universidad de León, 24071 León, Spain; Neural Therapies SL, Edificio Institutos de Investigación, Local B14, Universidad de León, 24071 León, Spain.
| | - Domenico E Pellegrini-Giampietro
- Sezione di Farmacologia Clinica e Oncologia, Dipartimento di Scienze della Salute, Università di Firenze, Viale Pieraccini 6, 50139 Firenze, Italy.
| | | |
Collapse
|
12
|
Ramos-Araque ME, Rodriguez C, Vecino R, Cortijo Garcia E, de Lera Alfonso M, Sanchez Barba M, Colàs-Campàs L, Purroy F, Arenillas JF, Almeida A, Delgado-Esteban M. The Neuronal Ischemic Tolerance Is Conditioned by the Tp53 Arg72Pro Polymorphism. Transl Stroke Res 2018; 10:204-215. [PMID: 29687302 PMCID: PMC6421278 DOI: 10.1007/s12975-018-0631-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/03/2018] [Accepted: 04/06/2018] [Indexed: 01/04/2023]
Abstract
Cerebral preconditioning (PC) confers endogenous brain protection after stroke. Ischemic stroke patients with a prior transient ischemic attack (TIA) may potentially be in a preconditioned state. Although PC has been associated with the activation of pro-survival signals, the mechanism by which preconditioning confers neuroprotection is not yet fully clarified. Recently, we have described that PC-mediated neuroprotection against ischemic insult is promoted by p53 destabilization, which is mediated by its main regulator MDM2. Moreover, we have previously described that the human Tp53 Arg72Pro single nucleotide polymorphism (SNP) controls susceptibility to ischemia-induced neuronal apoptosis and governs the functional outcome of patients after stroke. Here, we studied the contribution of the human Tp53 Arg72Pro SNP on PC-induced neuroprotection after ischemia. Our results showed that cortical neurons expressing the Pro72-p53 variant exhibited higher PC-mediated neuroprotection as compared with Arg72-p53 neurons. PC prevented ischemia-induced nuclear and cytosolic p53 stabilization in Pro72-p53 neurons. However, PC failed to prevent mitochondrial p53 stabilization, which occurs in Arg72-p53 neurons after ischemia. Furthermore, PC promoted neuroprotection against ischemia by controlling the p53/active caspase-3 pathway in Pro72-p53, but not in Arg72-p53 neurons. Finally, we found that good prognosis associated to TIA within 1 month prior to ischemic stroke was restricted to patients harboring the Pro72 allele. Our findings demonstrate that the Tp53 Arg72Pro SNP controls PC-promoted neuroprotection against a subsequent ischemic insult by modulating mitochondrial p53 stabilization and then modulates TIA-induced ischemic tolerance.
Collapse
Affiliation(s)
- Maria E Ramos-Araque
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Calle Zacarías González 2, 37007, Salamanca, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | - Cristina Rodriguez
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Calle Zacarías González 2, 37007, Salamanca, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | - Rebeca Vecino
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Calle Zacarías González 2, 37007, Salamanca, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | - Elisa Cortijo Garcia
- Stroke Unit, Department of Neurology, University Hospital of Valladolid, University of Valladolid, Valladolid, Spain
| | - Mercedes de Lera Alfonso
- Stroke Unit, Department of Neurology, University Hospital of Valladolid, University of Valladolid, Valladolid, Spain
| | - Mercedes Sanchez Barba
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Calle Zacarías González 2, 37007, Salamanca, Spain
- Department of Statistics, University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | | | - Francisco Purroy
- Clinical Neurosciences Group, IRBLleida. UdL, Lleida, Spain
- Stroke Unit, University Hospital Arnau de Vilanova, Lleida, Spain
| | - Juan F Arenillas
- Stroke Unit, Department of Neurology, University Hospital of Valladolid, University of Valladolid, Valladolid, Spain
- Neurovascular Research Laboratory (i3), Instituto de Biología y Genética Molecular, Universidad de Valladolid, CSIC, Valladolid, Spain
| | - Angeles Almeida
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Calle Zacarías González 2, 37007, Salamanca, Spain
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | - Maria Delgado-Esteban
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Calle Zacarías González 2, 37007, Salamanca, Spain.
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain.
| |
Collapse
|
13
|
Stradecki-Cohan HM, Cohan CH, Raval AP, Dave KR, Reginensi D, Gittens RA, Youbi M, Perez-Pinzon MA. Cognitive Deficits after Cerebral Ischemia and Underlying Dysfunctional Plasticity: Potential Targets for Recovery of Cognition. J Alzheimers Dis 2018; 60:S87-S105. [PMID: 28453486 DOI: 10.3233/jad-170057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cerebral ischemia affects millions of people worldwide and survivors suffer from long-term functional and cognitive deficits. While stroke and cardiac arrest are typically considered when discussing ischemic brain injuries, there is much evidence that smaller ischemic insults underlie neurodegenerative diseases, including Alzheimer's disease. The "regenerative" capacity of the brain relies on several aspects of plasticity that are crucial for normal functioning; less affected brain areas may take over function previously performed by irreversibly damaged tissue. To harness the endogenous plasticity mechanisms of the brain to provide recovery of cognitive function, we must first understand how these mechanisms are altered after damage, such as cerebral ischemia. In this review, we discuss the long-term cognitive changes that result after cerebral ischemia and how ischemia alters several plasticity processes. We conclude with a discussion of how current and prospective therapies may restore brain plasticity and allow for recovery of cognitive function, which may be applicable to several disorders that have a disruption of cognitive processing, including traumatic brain injury and Alzheimer's disease.
Collapse
Affiliation(s)
- Holly M Stradecki-Cohan
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Charles H Cohan
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA
| | - Ami P Raval
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA
| | - Kunjan R Dave
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Diego Reginensi
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Rolando A Gittens
- Centro de Neurociencias, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), City of Knowledge, Panama, Republic of Panama
| | - Mehdi Youbi
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology Cerebral Vascular Disease Research Laboratories, Miami, FL, USA.,Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
14
|
Lee RHC, Lee MHH, Wu CYC, Couto e Silva A, Possoit HE, Hsieh TH, Minagar A, Lin HW. Cerebral ischemia and neuroregeneration. Neural Regen Res 2018; 13:373-385. [PMID: 29623912 PMCID: PMC5900490 DOI: 10.4103/1673-5374.228711] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Cerebral ischemia is one of the leading causes of morbidity and mortality worldwide. Although stroke (a form of cerebral ischemia)-related costs are expected to reach 240.67 billion dollars by 2030, options for treatment against cerebral ischemia/stroke are limited. All therapies except anti-thrombolytics (i.e., tissue plasminogen activator) and hypothermia have failed to reduce neuronal injury, neurological deficits, and mortality rates following cerebral ischemia, which suggests that development of novel therapies against stroke/cerebral ischemia are urgently needed. Here, we discuss the possible mechanism(s) underlying cerebral ischemia-induced brain injury, as well as current and future novel therapies (i.e., growth factors, nicotinamide adenine dinucleotide, melatonin, resveratrol, protein kinase C isozymes, pifithrin, hypothermia, fatty acids, sympathoplegic drugs, and stem cells) as it relates to cerebral ischemia.
Collapse
Affiliation(s)
- Reggie H. C. Lee
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Michelle H. H. Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, China
| | - Celeste Y. C. Wu
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Alexandre Couto e Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Harlee E. Possoit
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Tsung-Han Hsieh
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Alireza Minagar
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Science Center, Shreveport, LA, USA
- Center for Brain Health, Louisiana State University Health Science Center, Shreveport, LA, USA
- Department of Cellular Biology and Anatomy, Louisiana State University Health Science Center, Shreveport, LA, USA
- Cardiovascular and Metabolomics Research Center, Hualien Tzu Chi Hospital, Hualien, Taiwan, China
| |
Collapse
|
15
|
The Hypoxia Mimetic Protocatechuic Acid Ethyl Ester Inhibits Synaptic Signaling and Plasticity in the Rat Hippocampus. Neuroscience 2018; 369:168-182. [DOI: 10.1016/j.neuroscience.2017.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/27/2017] [Accepted: 11/04/2017] [Indexed: 01/01/2023]
|
16
|
Cohan CH, Stradecki-Cohan HM, Morris-Blanco KC, Khoury N, Koronowski KB, Youbi M, Wright CB, Perez-Pinzon MA. Protein kinase C epsilon delays latency until anoxic depolarization through arc expression and GluR2 internalization. J Cereb Blood Flow Metab 2017; 37:3774-3788. [PMID: 28585865 PMCID: PMC5718329 DOI: 10.1177/0271678x17712178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Global cerebral ischemia is a debilitating injury that damages the CA1 region of the hippocampus, an area important for learning and memory. Protein kinase C epsilon (PKCɛ) activation is a critical component of many neuroprotective treatments. The ability of PKCɛ activation to regulate AMPA receptors (AMPARs) remains unexplored despite the role of AMPARs in excitotoxicity after brain ischemia. We determined that PKCɛ activation increased expression of a protein linked to learning and memory, activity-regulated cytoskeleton-associated protein (arc). Also, arc is necessary for neuroprotection and confers protection through decreasing AMPAR currents via GluR2 internalization. In vivo, activation of PKCɛ increased arc expression through a BDNF/TrkB pathway, and decreased GluR2 mRNA levels. In hippocampal cultured slices, PKCɛ activation decreased AMPAR current amplitudes in an arc- and GluR2-dependent manner. Additionally, PKCɛ activation triggered an arc- and GluR2 internalization-dependent delay in latency until anoxic depolarization. Inhibiting arc also blocked PKCɛ-mediated neuroprotection against lethal oxygen and glucose deprivation. These data characterize a novel PKCɛ-dependent mechanism that for the first time defines a role for arc and AMPAR internalization in conferring neuroprotection.
Collapse
Affiliation(s)
- Charles H Cohan
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Holly M Stradecki-Cohan
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kahlilia C Morris-Blanco
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Nathalie Khoury
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Kevin B Koronowski
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Mehdi Youbi
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Clinton B Wright
- 2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- 1 Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,2 Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,3 Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.,4 Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
Amantea D, Bagetta G. Excitatory and inhibitory amino acid neurotransmitters in stroke: from neurotoxicity to ischemic tolerance. Curr Opin Pharmacol 2017; 35:111-119. [DOI: 10.1016/j.coph.2017.07.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022]
|
18
|
Capuani B, Pacifici F, Pastore D, Palmirotta R, Donadel G, Arriga R, Bellia A, Di Daniele N, Rogliani P, Abete P, Sbraccia P, Guadagni F, Lauro D, Della-Morte D. The role of epsilon PKC in acute and chronic diseases: Possible pharmacological implications of its modulators. Pharmacol Res 2016; 111:659-667. [PMID: 27461137 DOI: 10.1016/j.phrs.2016.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
|
19
|
Morris-Blanco KC, Dave KR, Saul I, Koronowski KB, Stradecki HM, Perez-Pinzon MA. Protein Kinase C Epsilon Promotes Cerebral Ischemic Tolerance Via Modulation of Mitochondrial Sirt5. Sci Rep 2016; 6:29790. [PMID: 27435822 PMCID: PMC4951704 DOI: 10.1038/srep29790] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/24/2016] [Indexed: 01/21/2023] Open
Abstract
Sirtuin 5 (SIRT5) is a mitochondrial-localized NAD(+)-dependent lysine desuccinylase and a major regulator of the mitochondrial succinylome. We wanted to determine whether SIRT5 is activated by protein kinase C epsilon (PKCε)-mediated increases in mitochondrial Nampt and whether SIRT5 regulates mitochondrial bioenergetics and neuroprotection against cerebral ischemia. In isolated mitochondria from rat cortical cultures, PKCε activation increased SIRT5 levels and desuccinylation activity in a Nampt-dependent manner. PKCε activation did not lead to significant modifications in SIRT3 activity, the major mitochondrial lysine deacetylase. Assessments of mitochondrial bioenergetics in the cortex of wild type (WT) and SIRT5-/- mice revealed that SIRT5 regulates oxygen consumption in the presence of complex I, complex II, and complex IV substrates. To explore the potential role of SIRT5 in PKCε-mediated protection, we compared WT and SIRT5-/- mice by employing both in vitro and in vivo ischemia paradigms. PKCε-mediated decreases in cell death following oxygen-glucose deprivation were abolished in cortical cultures harvested from SIRT5-/- mice. Furthermore, PKCε failed to prevent cortical degeneration following MCAO in SIRT5-/- mice. Collectively this demonstrates that SIRT5 is an important mitochondrial enzyme for protection against metabolic and ischemic stress following PKCε activation in the brain.
Collapse
Affiliation(s)
- Kahlilia C. Morris-Blanco
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kunjan R. Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Isabel Saul
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kevin B. Koronowski
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Holly M. Stradecki
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Miguel A. Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
20
|
Kantati YT, Kodjo KM, Dogbeavou KS, Vaudry D, Leprince J, Gbeassor M. Ethnopharmacological survey of plant species used in folk medicine against central nervous system disorders in Togo. JOURNAL OF ETHNOPHARMACOLOGY 2016; 181:214-220. [PMID: 26869544 DOI: 10.1016/j.jep.2016.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 02/04/2016] [Accepted: 02/06/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neurological diseases are rising all around the world. In a developing country such as Togo, although plant-based medicines are the only means, still very little is known regarding the nature and efficiency of medicinal plants used by indigenous people to manage central nervous system (CNS) disorders. AIM OF THE STUDY This study, an ethnobotanical survey, aimed to report plant species used in traditional medicine (TM) for the management of various CNS disorders in Togo. MATERIALS AND METHODS 52 traditional actors (TA) including 33 traditional healers (TH) and 19 medicinal plant sellers (MPS) were interviewed, using a questionnaire mentioning informants' general data and uses of medicinal plants. RESULTS The present study reports 44 medicinal plant species distributed into 26 families, mentioning scientific and common local names, plant organs used, preparation method, root of administration and putative applications. CONCLUSION It appears that there is a real knowledge on medicinal plants used for traditional treatment of CNS disorders in Togo and that the local flora abounds of potentially neuroactive plants which could be useful for the discovery of antipsychotic or neuroprotective molecules.
Collapse
Affiliation(s)
- Yendube T Kantati
- Laboratory of Physiology/Pharmacology, University of Lomé, Po Box: 1515 Lomé, Togo.
| | - K Magloire Kodjo
- Laboratory of Physiology/Pharmacology, University of Lomé, Po Box: 1515 Lomé, Togo
| | - Koffi S Dogbeavou
- Laboratory of Physiology/Pharmacology, University of Lomé, Po Box: 1515 Lomé, Togo
| | - David Vaudry
- INSERM U982, Neurotrophic Factors and Neuronal Differentiation Team, University of Rouen, Place E. Blondel, 76821 Mont-Saint-Aignan, France
| | - Jérôme Leprince
- INSERM U982, Neurotrophic Factors and Neuronal Differentiation Team, University of Rouen, Place E. Blondel, 76821 Mont-Saint-Aignan, France
| | - Messanvi Gbeassor
- Laboratory of Physiology/Pharmacology, University of Lomé, Po Box: 1515 Lomé, Togo
| |
Collapse
|
21
|
Abstract
UNLABELLED Ischemic preconditioning (IPC) is a robust neuroprotective phenomenon whereby brief ischemic exposure confers tolerance to a subsequent ischemic challenge. IPC has not been studied selectively in CNS white matter (WM), although stroke frequently involves WM. We determined whether IPC is present in WM and, if so, its mechanism. We delivered a brief in vivo preconditioning ischemic insult (unilateral common carotid artery ligation) to 12- to 14-week-old mice and determined WM ischemic vulnerability [oxygen-glucose deprivation (OGD)] 72 h later, using acutely isolated optic nerves (CNS WM tracts) from the preconditioned (ipsilateral) and control (contralateral) hemispheres. Functional and structural recovery was assessed by quantitative measurement of compound action potentials (CAPs) and immunofluorescent microscopy. Preconditioned mouse optic nerves (MONs) showed better functional recovery after OGD than the non-preconditioned MONs (31 ± 3 vs 17 ± 3% normalized CAP area, p < 0.01). Preconditioned MONs also showed improved axon integrity and reduced oligodendrocyte injury compared with non-preconditioned MONs. Toll-like receptor-4 (TLR4) and type 1 interferon receptor (IFNAR1), key receptors in innate immune response, are implicated in gray matter preconditioning. Strikingly, IPC-mediated WM protection was abolished in both TLR4(-/-) and IFNAR1(-/-) mice. In addition, IPC-mediated protection in WM was also abolished in IFNAR1(fl/fl) LysM(cre), but not in IFNAR1(fl/fl) control, mice. These findings demonstrated for the first time that IPC was robust in WM, the phenomenon being intrinsic to WM itself. Furthermore, WM IPC was dependent on innate immune cell signaling pathways. Finally, these data demonstrated that microglial-specific expression of IFNAR1 plays an indispensable role in WM IPC. SIGNIFICANCE STATEMENT Ischemic preconditioning (IPC) has been studied predominantly in gray matter, but stroke in humans frequently involves white matter (WM) as well. Here we describe a novel, combined in vivo/ex vivo mouse model to determine whether IPC occurs in WM. It does. Using genetically altered mice, we identified two innate immune cell receptors, Toll-like receptor 4 and type 1 interferon receptor (IFNAR1), that are required for IPC-mediated protection in WM. Furthermore, using microglia-targeted IFNAR1 knockdown, we demonstrate that interferon signaling specifically in microglia is essential for this protection. The discovery of IPC as an intrinsic capability of WM is novel and important. This is also the first in vivo demonstration that cell-type-specific expression of an individual gene plays an indispensable role in IPC-mediated protection.
Collapse
|
22
|
Sobol CV, Belostotskaya GB. Product fermented by Lactobacilli induces changes in intracellular calcium dynamics in rat brain neurons. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2016. [DOI: 10.1134/s199074781505013x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
23
|
Guan YF, Wu CY, Fang YY, Zeng YN, Luo ZY, Li SJ, Li XW, Zhu XH, Mei L, Gao TM. Neuregulin 1 protects against ischemic brain injury via ErbB4 receptors by increasing GABAergic transmission. Neuroscience 2015; 307:151-9. [DOI: 10.1016/j.neuroscience.2015.08.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 08/20/2015] [Accepted: 08/20/2015] [Indexed: 12/22/2022]
|
24
|
Wang Y, Reis C, Applegate R, Stier G, Martin R, Zhang JH. Ischemic conditioning-induced endogenous brain protection: Applications pre-, per- or post-stroke. Exp Neurol 2015; 272:26-40. [PMID: 25900056 DOI: 10.1016/j.expneurol.2015.04.009] [Citation(s) in RCA: 318] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 04/06/2015] [Accepted: 04/11/2015] [Indexed: 11/17/2022]
Abstract
In the area of brain injury and neurodegenerative diseases, a plethora of experimental and clinical evidence strongly indicates the promise of therapeutically exploiting the endogenous adaptive system at various levels like triggers, mediators and the end-effectors to stimulate and mobilize intrinsic protective capacities against brain injuries. It is believed that ischemic pre-conditioning and post-conditioning are actually the strongest known interventions to stimulate the innate neuroprotective mechanism to prevent or reverse neurodegenerative diseases including stroke and traumatic brain injury. Recently, studies showed the effectiveness of ischemic per-conditioning in some organs. Therefore the term ischemic conditioning, including all interventions applied pre-, per- and post-ischemia, which spans therapeutic windows in 3 time periods, has recently been broadly accepted by scientific communities. In addition, it is extensively acknowledged that ischemia-mediated protection not only affects the neurons but also all the components of the neurovascular network (consisting of neurons, glial cells, vascular endothelial cells, pericytes, smooth muscle cells, and venule/veins). The concept of cerebroprotection has been widely used in place of neuroprotection. Intensive studies on the cellular signaling pathways involved in ischemic conditioning have improved the mechanistic understanding of tolerance to cerebral ischemia. This has added impetus to exploration for potential pharmacologic mimetics, which could possibly induce and maximize inherent protective capacities. However, most of these studies were performed in rodents, and the efficacy of these mimetics remains to be evaluated in human patients. Several classical signaling pathways involving apoptosis, inflammation, or oxidation have been elaborated in the past decades. Newly characterized mechanisms are emerging with the advances in biotechnology and conceptual renewal. In this review we are going to focus on those recently reported methodological and mechanistic discoveries in the realm of ischemic conditioning. Due to the varied time differences of ischemic conditioning in different animal models and clinical trials, it is important to define optimal timing to achieve the best conditioning induced neuroprotection. This brings not only an opportunity in the treatment of stroke, but challenges as well, as data is just becoming available and the procedures are not yet optimized. The purpose of this review is to shed light on exploiting these ischemic conditioning modalities to protect the cerebrovascular system against diverse injuries and neurodegenerative disorders.
Collapse
Affiliation(s)
- Yuechun Wang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, USA; Department of Physiology, Jinan University School of Medicine, Guangzhou, China
| | - Cesar Reis
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Richard Applegate
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Gary Stier
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Robert Martin
- Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, USA; Department of Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA, USA.
| |
Collapse
|
25
|
Neumann JT, Thompson JW, Raval AP, Cohan CH, Koronowski KB, Perez-Pinzon MA. Increased BDNF protein expression after ischemic or PKC epsilon preconditioning promotes electrophysiologic changes that lead to neuroprotection. J Cereb Blood Flow Metab 2015; 35:121-30. [PMID: 25370861 PMCID: PMC4294405 DOI: 10.1038/jcbfm.2014.185] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 01/06/2023]
Abstract
Ischemic preconditioning (IPC) via protein kinase C epsilon (PKCɛ) activation induces neuroprotection against lethal ischemia. Brain-derived neurotrophic factor (BDNF) is a pro-survival signaling molecule that modulates synaptic plasticity and neurogenesis. Interestingly, BDNF mRNA expression increases after IPC. In this study, we investigated whether IPC or pharmacological preconditioning (PKCɛ activation) promoted BDNF-induced neuroprotection, if neuroprotection by IPC or PKCɛ activation altered neuronal excitability, and whether these changes were BDNF-mediated. We used both in vitro (hippocampal organotypic cultures and cortical neuronal-glial cocultures) and in vivo (acute hippocampal slices 48 hours after preconditioning) models of IPC or PKCɛ activation. BDNF protein expression increased 24 to 48 hours after preconditioning, where inhibition of the BDNF Trk receptors abolished neuroprotection against oxygen and glucose deprivation (OGD) in vitro. In addition, there was a significant decrease in neuronal firing frequency and increase in threshold potential 48 hours after preconditioning in vivo, where this threshold modulation was dependent on BDNF activation of Trk receptors in excitatory cortical neurons. In addition, 48 hours after PKCɛ activation in vivo, the onset of anoxic depolarization during OGD was significantly delayed in hippocampal slices. Overall, these results suggest that after IPC or PKCɛ activation, there are BDNF-dependent electrophysiologic modifications that lead to neuroprotection.
Collapse
Affiliation(s)
- Jake T Neumann
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - John W Thompson
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Ami P Raval
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Charles H Cohan
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [4] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Kevin B Koronowski
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- 1] Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [2] Evelyn F. McKnight Brain Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [3] Department of Neurology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA [4] Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
26
|
Della-Morte D, Guadagni F, Palmirotta R, Ferroni P, Testa G, Cacciatore F, Abete P, Rengo F, Perez-Pinzon MA, Sacco RL, Rundek T. Genetics and genomics of ischemic tolerance: focus on cardiac and cerebral ischemic preconditioning. Pharmacogenomics 2013; 13:1741-57. [PMID: 23171338 DOI: 10.2217/pgs.12.157] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A subthreshold ischemic insult applied to an organ such as the heart and/or brain may help to reduce damage caused by subsequent ischemic episodes. This phenomenon is known as ischemic tolerance mediated by ischemic preconditioning (IPC) and represents the most powerful endogenous mechanism against ischemic injury. Various molecular pathways have been implicated in IPC, and several compounds have been proposed as activators or mediators of IPC. Recently, it has been established that the protective phenotype in response to ischemia depends on a coordinated response at the genomic, molecular, cellular and tissue levels by introducing the concept of 'genomic reprogramming' following IPC. In this article, we sought to review the genetic expression profiles found in cardiac and cerebral IPC studies, describe the differences between young and aged organs in IPC-mediated protection, and discuss the potential therapeutic application of IPC and pharmacological preconditioning based on the genomic response.
Collapse
Affiliation(s)
- David Della-Morte
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sun X, Budas GR, Xu L, Barreto GE, Mochly-Rosen D, Giffard RG. Selective activation of protein kinase C∊ in mitochondria is neuroprotective in vitro and reduces focal ischemic brain injury in mice. J Neurosci Res 2013; 91:799-807. [PMID: 23426889 DOI: 10.1002/jnr.23186] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 11/13/2012] [Accepted: 11/13/2012] [Indexed: 01/08/2023]
Abstract
Activation of protein kinase C∊ (PKC∊) confers protection against neuronal ischemia/reperfusion. Activation of PKC∊ leads to its translocation to multiple intracellular sites, so a mitochondria-selective PKC∊ activator was used to test the importance of mitochondrial activation to the neuroprotective effect of PKC∊. PKC∊ can regulate key cytoprotective mitochondrial functions, including electron transport chain activity, reactive oxygen species (ROS) generation, mitochondrial permeability transition, and detoxification of reactive aldehydes. We tested the ability of mitochondria-selective activation of PKC∊ to protect primary brain cell cultures or mice subjected to ischemic stroke. Pretreatment with either general PKC∊ activator peptide, TAT-Ψ∊RACK, or mitochondrial-selective PKC∊ activator, TAT-Ψ∊HSP90, reduced cell death induced by simulated ischemia/reperfusion in neurons, astrocytes, and mixed neuronal cultures. The protective effects of both TAT-Ψ∊RACK and TAT-Ψ∊HSP90 were blocked by the PKC∊ antagonist ∊V1-2 , indicating that protection requires PKC∊ interaction with its anchoring protein, TAT-∊RACK. Further supporting a mitochondrial mechanism for PKC∊, neuroprotection by TAT-Ψ∊HSP90 was associated with a marked delay in mitochondrial membrane depolarization and significantly attenuated ROS generation during ischemia. Importantly, TAT-Ψ∊HSP90 reduced infarct size and reduced neurological deficit in C57/BL6 mice subjected to middle cerebral artery occlusion and 24 hr of reperfusion. Thus selective activation of mitochondrial PKC∊ preserves mitochondrial function in vitro and improves outcome in vivo, suggesting potential therapeutic value clinically when brain ischemia is anticipated, including neurosurgery and cardiac surgery.
Collapse
Affiliation(s)
- Xiaoyun Sun
- Department of Anesthesia, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | | | | | | | | |
Collapse
|
28
|
Ferrari LF, Levine E, Levine JD. Role of a novel nociceptor autocrine mechanism in chronic pain. Eur J Neurosci 2013; 37:1705-13. [PMID: 23379641 DOI: 10.1111/ejn.12145] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/11/2012] [Accepted: 01/07/2013] [Indexed: 12/13/2022]
Abstract
We have previously shown, in the rat, that neuropathic and inflammatory events produce a neuroplastic change in nociceptor function whereby a subsequent exposure to a proinflammatory mediator (e.g. prostaglandin E2 ; PGE2 ) produces markedly prolonged mechanical hyperalgesia. While the initial approximately 30 min of this prolonged PGE2 hyperalgesia remains PKA-dependent, it subsequently switches to become dependent on protein kinase C epsilon (PKCε). In this study we tested the hypothesis that the delayed onset, PKCε-mediated, component of PGE2 hyperalgesia is generated by the active release of a nucleotide from the peripheral terminal of the primed nociceptor and this nucleotide is then metabolized to produce adenosine, which acts on a Gi-coupled A1 adenosine receptor on the nociceptor to generate PKCε-dependent hyperalgesia. We report that inhibitors of ATP-binding cassette transporters, of ecto-5'-phosphodiesterase and ecto-5'nucleotidase (enzymes involved in the metabolism of cyclic nucleotides to adenosine) and of A1 adenosine receptors each eliminated the late, but not the early, phase of PGE2 -induced hyperalgesia in primed animals. A second model of chronic pain induced by transient attenuation of G-protein-coupled receptor kinase 2, in which the prolongation of PGE2 hyperalgesia is not PKCε-dependent, was not attenuated by inhibitors of any of these mechanisms. Based on these results we propose a contribution of an autocrine mechanism, in the peripheral terminal of the nociceptor, in the hyperalgesic priming model of chronic pain.
Collapse
Affiliation(s)
- Luiz F Ferrari
- Division of Neuroscience, Departments of Medicine and Oral Surgery, University of California, San Francisco, CA 94143-0440, USA
| | | | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Ischemic preconditioning (IPC) is gaining attention as a novel neuroprotective therapy and could provide an improved mechanistic understanding of tolerance to cerebral ischemia. The purpose of this article is to review the recent work in the field of IPC and its applications to clinical scenarios. RECENT FINDINGS The cellular signaling pathways that are activated following IPC are now better understood and have enabled investigators to identify several IPC mimetics. Most of these studies were performed in rodents, and efficacy of these mimetics remains to be evaluated in human patients. Additionally, remote ischemic preconditioning (RIPC) may have higher translational value than IPC. Repeated cycles of temporary ischemia in a remote organ can activate protective pathways in the target organ, including the heart and brain. Clinical trials are underway to test the efficacy of RIPC in protecting brain against subarachnoid hemorrhage. SUMMARY IPC, RIPC, and IPC mimetics have the potential to be therapeutic in various clinical scenarios. Further understanding of IPC-induced neuroprotection pathways and utilization of clinically relevant animal models are necessary to increase the translational potential of IPC in the near future.
Collapse
Affiliation(s)
- Srinivasan V Narayanan
- Department of Neurology, Cerebral Vascular Disease Research Center, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | | |
Collapse
|
30
|
Neumann JT, Cohan CH, Dave KR, Wright CB, Perez-Pinzon MA. Global cerebral ischemia: synaptic and cognitive dysfunction. Curr Drug Targets 2013; 14:20-35. [PMID: 23170794 PMCID: PMC5800514 DOI: 10.2174/138945013804806514] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/27/2012] [Accepted: 11/16/2012] [Indexed: 11/22/2022]
Abstract
Cardiopulmonary arrest is one of the leading causes of death and disability, primarily occurring in the aged population. Numerous global cerebral ischemia animal models induce neuronal damage similar to cardiac arrest. These global cerebral ischemia models range from vessel occlusion to total cessation of cardiac function, both of which have allowed for the investigation of this multifaceted disease and detection of numerous agents that are neuroprotective. Synapses endure a variety of alterations after global cerebral ischemia from the resulting excitotoxicity and have been a major target for neuroprotection; however, neuroprotective agents have proven unsuccessful in clinical trials, as neurological outcomes have not displayed significant improvements in patients. A majority of these neuroprotective agents have specific neuronal targets, where the success of future neuroprotective agents may depend on non-specific targets and numerous cognitive improvements. This review focuses on the different models of global cerebral ischemia, neuronal synaptic alterations, synaptic neuroprotection and behavioral tests that can be used to determine deficits in cognitive function after global cerebral ischemia.
Collapse
Affiliation(s)
- Jake T Neumann
- Cerebral Vascular Disease Research Laboratories, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | | | | | | | | |
Collapse
|
31
|
Ferchmin PA, Pérez D, Castro Alvarez W, Penzo MA, Maldonado HM, Eterovic VA. γ-Aminobutyric acid type A receptor inhibition triggers a nicotinic neuroprotective mechanism. J Neurosci Res 2012; 91:416-25. [PMID: 23280428 DOI: 10.1002/jnr.23155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 08/09/2012] [Accepted: 09/14/2012] [Indexed: 01/09/2023]
Abstract
Nicotinic acetylcholine receptor (nAChR)-mediated neuroprotection has been implicated in the treatment of neurodegenerative disorders such as Alzheimer's and Parkinson's diseases and hypoxic ischemic events as well as other diseases hallmarked by excitotoxic and apoptotic neuronal death. Several modalities of nicotinic neuroprotection have been reported. However, although this process generally involves α4β2 and α7 subtypes, the underlying mechanisms are largely unknown. Interestingly, both activation and inhibition of α7 nAChRs have been reported to be neuroprotective. We have shown that inhibition of α7 nAChRs protects the function of acute hippocampal slices against excitotoxicity in an α4β2-dependent manner. Neuroprotection was assessed as the prevention of the N-methyl-D-aspartate-dependent loss of the area of population spikes (PSs) in the CA1 area of acute hippocampal slices. Our results support a model in which α7 AChRs control the release of γ-aminobutyric acid (GABA). Blocking either α7 or GABA(A) receptors reduces the inhibitory tone on cholinergic terminals, thereby promoting α4β2 activation, which in turn mediates neuroprotection. These results shed light on how α7 nAChR inhibition can be neuroprotective through a mechanism mediated by activation of α4β2 nAChRs.
Collapse
Affiliation(s)
- P A Ferchmin
- School of Medicine, Universidad Central del Caribe, Bayamón, Puerto Rico.
| | | | | | | | | | | |
Collapse
|
32
|
Yokobori S, Mazzeo AT, Hosein K, Gajavelli S, Dietrich WD, Bullock MR. Preconditioning for traumatic brain injury. Transl Stroke Res 2012; 4:25-39. [PMID: 24323189 DOI: 10.1007/s12975-012-0226-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 10/23/2012] [Accepted: 10/29/2012] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI) treatment is now focused on the prevention of primary injury and reduction of secondary injury. However, no single effective treatment is available as yet for the mitigation of traumatic brain damage in humans. Both chemical and environmental stresses applied before injury have been shown to induce consequent protection against post-TBI neuronal death. This concept termed "preconditioning" is achieved by exposure to different pre-injury stressors to achieve the induction of "tolerance" to the effect of the TBI. However, the precise mechanisms underlying this "tolerance" phenomenon are not fully understood in TBI, and therefore even less information is available about possible indications in clinical TBI patients. In this review, we will summarize TBI pathophysiology, and discuss existing animal studies demonstrating the efficacy of preconditioning in diffuse and focal type of TBI. We will also review other non-TBI preconditioning studies, including ischemic, environmental, and chemical preconditioning, which maybe relevant to TBI. To date, no clinical studies exist in this field, and we speculate on possible future clinical situations, in which pre-TBI preconditioning could be considered.
Collapse
Affiliation(s)
- Shoji Yokobori
- Department of Neurosurgery, Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA,
| | | | | | | | | | | |
Collapse
|
33
|
Lymphocyte cell kinase activation mediates neuroprotection during ischemic preconditioning. J Neurosci 2012; 32:7278-86. [PMID: 22623673 DOI: 10.1523/jneurosci.6273-11.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The molecular mechanisms underlying preconditioning (PC), a powerful endogenous neuroprotective phenomenon, remain to be fully elucidated. Once identified, these endogenous mechanisms could be manipulated for therapeutic gain. We investigated whether lymphocyte cell kinase (Lck), a member of the Src kinases family, mediates PC. We used both in vitro primary cortical neurons and in vivo mouse cerebral focal ischemia models of preconditioning, cellular injury, and neuroprotection. Genetically engineered mice deficient in Lck, gene silencing using siRNA, and pharmacological approaches were used. Cortical neurons preconditioned with sublethal exposure to NMDA or oxygen glucose deprivation (OGD) exhibited enhanced Lck kinase activity, and were resistant to injury on subsequent exposure to lethal levels of NMDA or OGD. Lck gene silencing using siRNA abolished tolerance against both stimuli. Lck-/- mice or neurons isolated from Lck-/- mice did not exhibit PC-induced tolerance. An Lck antagonist administered to wild-type mice significantly attenuated the neuroprotective effect of PC in the mouse focal ischemia model. Using pharmacological and gene silencing strategies, we also showed that PKCε is an upstream regulator of Lck, and Fyn is a downstream target of Lck. We have discovered that Lck plays an essential role in PC in both cellular and animal models of stroke. Our data also show that the PKCε-Lck-Fyn axis is a key mediator of PC. These findings provide new opportunities for stroke therapy development.
Collapse
|
34
|
Tauskela JS, Aylsworth A, Hewitt M, Brunette E, Mealing GAR. Preconditioning induces tolerance by suppressing glutamate release in neuron culture ischemia models. J Neurochem 2012; 122:470-81. [DOI: 10.1111/j.1471-4159.2012.07791.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
Fitzgerald CT, Carter LP. Possible role for glutamic acid decarboxylase in fibromyalgia symptoms: A conceptual model for chronic pain. Med Hypotheses 2011; 77:409-15. [DOI: 10.1016/j.mehy.2011.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 05/23/2011] [Accepted: 05/26/2011] [Indexed: 11/28/2022]
|
36
|
Lin HW, Thompson JW, Morris KC, Perez-Pinzon MA. Signal transducers and activators of transcription: STATs-mediated mitochondrial neuroprotection. Antioxid Redox Signal 2011; 14:1853-61. [PMID: 20712401 PMCID: PMC3078497 DOI: 10.1089/ars.2010.3467] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cerebral ischemia is defined as little or no blood flow in cerebral circulation, characterized by low tissue oxygen and glucose levels, which promotes neuronal mitochondria dysfunction leading to cell death. A strategy to counteract cerebral ischemia-induced neuronal cell death is ischemic preconditioning (IPC). IPC results in neuroprotection, which is conferred by a mild ischemic challenge prior to a normally lethal ischemic insult. Although many IPC-induced mechanisms have been described, many cellular and subcellular mechanisms remain undefined. Some reports have suggested key signal transduction pathways of IPC, such as activation of protein kinase C epsilon, mitogen-activated protein kinase, and hypoxia-inducible factors, that are likely involved in IPC-induced mitochondria mediated-neuroprotection. Moreover, recent findings suggest that signal transducers and activators of transcription (STATs), a family of transcription factors involved in many cellular activities, may be intimately involved in IPC-induced ischemic tolerance. In this review, we explore current signal transduction pathways involved in IPC-induced mitochondria mediated-neuroprotection, STAT activation in the mitochondria as it relates to IPC, and functional significance of STATs in cerebral ischemia.
Collapse
Affiliation(s)
- Hung Wen Lin
- Cerebral Vascular Disease Research Center, Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida 33101, USA
| | | | | | | |
Collapse
|
37
|
Morris KC, Lin HW, Thompson JW, Perez-Pinzon MA. Pathways for ischemic cytoprotection: role of sirtuins in caloric restriction, resveratrol, and ischemic preconditioning. J Cereb Blood Flow Metab 2011; 31:1003-19. [PMID: 21224864 PMCID: PMC3070983 DOI: 10.1038/jcbfm.2010.229] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Caloric restriction (CR), resveratrol, and ischemic preconditioning (IPC) have been shown to promote protection against ischemic injury in the heart and brain, as well as in other tissues. The activity of sirtuins, which are enzymes that modulate diverse biologic processes, seems to be vital in the ability of these therapeutic modalities to prevent against cellular dysfunction and death. The protective mechanisms of the yeast Sir2 and the mammalian homolog sirtuin 1 have been extensively studied, but the involvement of other sirtuins in ischemic protection is not yet clear. We examine the roles of mammalian sirtuins in modulating protective pathways against oxidative stress, energy depletion, excitotoxicity, inflammation, DNA damage, and apoptosis. Although many of these sirtuins have not been directly implicated in ischemic protection, they may have unique roles in enhancing function and preventing against stress-mediated cellular damage and death. This review will include in-depth analyses of the roles of CR, resveratrol, and IPC in activating sirtuins and in mediating protection against ischemic damage in the heart and brain.
Collapse
Affiliation(s)
- Kahlilia C Morris
- Department of Neurology, Cerebral Vascular Disease Research Center, University of Miami, Miller School of Medicine, Miami, Florida 33101, USA
| | | | | | | |
Collapse
|
38
|
Wang Q, Li X, Chen Y, Wang F, Yang Q, Chen S, Min Y, Li X, Xiong L. Activation of Epsilon Protein Kinase C-Mediated Anti-Apoptosis Is Involved in Rapid Tolerance Induced by Electroacupuncture Pretreatment Through Cannabinoid Receptor Type 1. Stroke 2011; 42:389-96. [PMID: 21183751 DOI: 10.1161/strokeaha.110.597336] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Qiang Wang
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xuying Li
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yanke Chen
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Feng Wang
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Qianzi Yang
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Shaoyang Chen
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yuyuan Min
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xin Li
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Lize Xiong
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
39
|
Della-Morte D, Raval AP, Dave KR, Lin HW, Perez-Pinzon MA. Post-ischemic activation of protein kinase C ε protects the hippocampus from cerebral ischemic injury via alterations in cerebral blood flow. Neurosci Lett 2011; 487:158-62. [PMID: 20951185 PMCID: PMC3004991 DOI: 10.1016/j.neulet.2010.10.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 10/05/2010] [Accepted: 10/05/2010] [Indexed: 11/17/2022]
Abstract
Protein kinase C (PKC) is a family of serine/threonine-isozymes that are involved in many signaling events in normal and disease states. Previous studies from our lab have demonstrated that ɛPKC plays a pivotal role in neuroprotection induced by ischemic preconditioning. However, the role of ɛPKC during and after brain ischemia is not clearly defined. Therefore, in the present study, we tested the hypothesis that activation of ɛPKC during an ischemic event is neuroprotective. Furthermore, other studies have demonstrated that ɛPKC mediates cerebral ischemic tolerance in the rat brain by decreasing vascular tone. Thus, we also tested the effects of ɛPKC activation during ischemia on cerebral blood flow (CBF). We found that ψɛ-Receptors for Activated C Kinase (RACK), a ɛPKC-selective peptide activator, injected intravenously 30min before induction of global cerebral ischemia conferred neuroprotection in the CA1 region of the rat hippocampus. Moreover, measurements of CBF before, during, and after cerebral ischemia revealed a significant reduction in the reperfusion phase of rats pretreated with ψɛRACK as compared to Tat peptide (vehicle). Our results suggest that ɛPKC can protect the rat brain against ischemic damage by regulating CBF. Thus, ɛPKC may be one of the treatment modalities against ischemic injury.
Collapse
Affiliation(s)
- D Della-Morte
- Department of Neurology, University of Miami, Miami, FL 33101, USA
| | | | | | | | | |
Collapse
|
40
|
Chi OZ, Hunter C, Liu X, Chi Y, Weiss HR. Effects of GABA(A) receptor blockade on regional cerebral blood flow and blood-brain barrier disruption in focal cerebral ischemia. J Neurol Sci 2010; 301:66-70. [PMID: 21094956 DOI: 10.1016/j.jns.2010.10.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 10/14/2010] [Accepted: 10/22/2010] [Indexed: 11/18/2022]
Abstract
In cerebral ischemia, transmission by the inhibitory neurotransmitter, γ-aminobutyric acid (GABA) is altered. This study was performed to determine whether blockade of GABA(A) receptor would affect regional cerebral blood flow (rCBF) and blood-brain barrier (BBB) permeability in a focal ischemic area of the brain. Rats were anesthetized with isoflurane and mechanically ventilated. Fifteen minutes after a permanent middle cerebral artery (MCA) occlusion, one half of the rats were infused with bicuculline 1mg/kg/min iv for 2 min followed by 0.1mg/kg/min iv to the end of the experiment. The other half were infused with normal saline. At one hour after MCA occlusion, rCBF was determined using ¹⁴C-iodoantipyrine and BBB permeability was determined by measuring the transfer coefficient (Ki) of ¹⁴C-α-aminoisobutyric acid. With MCA occlusion, rCBF was decreased in the ischemic cortex (IC) (-70%) in the control rats. In the bicuculline treated rats, the rCBF of the IC was lower (-48%) than the contralateral cortex but higher than the rCBF of the IC of the control rats (+55%). MCA occlusion increased Ki in the IC of the control rats (+72%) and bicuculline administration increased Ki further (+53%) in the IC. Blockade of GABA(A) receptors did not significantly affect rCBF or BBB permeability in the non-ischemic brain regions under isoflurane anesthesia. Our data demonstrated that blockade of GABA(A) receptors increased rCBF and enhanced the BBB disruption in focal cerebral ischemia. Our data suggest that GABA(A) receptors are involved, at least in part, in modulating rCBF and BBB disruption in focal cerebral ischemia.
Collapse
Affiliation(s)
- Oak Z Chi
- Department of Anesthesia, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901-1977, USA.
| | | | | | | | | |
Collapse
|
41
|
Sandu N, Cornelius J, Filis A, Arasho B, Perez-Pinzon M, Schaller B. Ischemic tolerance in stroke treatment. Expert Rev Cardiovasc Ther 2010; 7:1255-61. [PMID: 19814668 DOI: 10.1586/erc.09.90] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although outcome after stroke treatment has significantly improved over the last 30 years, there has been no revolutionary breakthrough. Among different combined approaches, systemic thrombolysis in combination with neuroprotection became a favorite research target. Recent studies suggest that transient ischemic attacks may represent a clinical model of such ischemic tolerance; thus, a new focus on this research has emerged. In this review, we show the parallels between ischemia and neuroprotection and discuss the potential therapeutic options that may be opened by this new molecular knowledge.
Collapse
Affiliation(s)
- Nora Sandu
- Department of Neurosurgery, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
42
|
Cimarosti H, Kantamneni S, Henley JM. Ischaemia differentially regulates GABA(B) receptor subunits in organotypic hippocampal slice cultures. Neuropharmacology 2009; 56:1088-96. [PMID: 19328818 PMCID: PMC3309028 DOI: 10.1016/j.neuropharm.2009.03.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2008] [Revised: 02/19/2009] [Accepted: 03/18/2009] [Indexed: 02/04/2023]
Abstract
Reduced synaptic inhibition due to dysfunction of ionotropic GABA(A) receptors has been proposed as one factor in cerebral ischaemia-induced excitotoxic cell death. However, the participation of the inhibitory metabotropic GABA(B) receptors in these pathological processes has not been extensively investigated. We used oxygen-glucose deprivation (OGD) and NMDA-induced excitotoxicity as models to investigate whether ischaemia-like challenges alter the protein levels of GABA(B1) and GABA(B2) receptor subunits in rat organotypic hippocampal slice cultures. Twenty-four hours after the insult both OGD and NMDA produced a marked decrease in the total levels of GABA(B2) (approximately 75%), while there was no significant change in the levels of GABA(B1) after OGD, but an increase after NMDA treatment (approximately 100%). The GABA(B) receptor agonist baclofen (100 microM) was neuroprotective following OGD or NMDA treatment if added before or during the insult. GABA(B) receptors comprise heterodimers of GABA(B1) and GABA(B2) subunits and our results suggest that the separate subunits are independently regulated in response to extreme neuronal stress. However, because GABA(B2) is required for functional surface expression, down-regulation of this subunit removes an important inhibitory feedback mechanism under pathological conditions.
Collapse
Affiliation(s)
| | | | - Jeremy M. Henley
- Department of Anatomy, MRC Centre for Synaptic Plasticity, School of Medical Sciences, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
43
|
Neuronal plasticity after ischemic preconditioning and TIA-like preconditioning ischemic periods. Acta Neuropathol 2009; 117:511-23. [PMID: 19084975 DOI: 10.1007/s00401-008-0473-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 11/29/2008] [Accepted: 12/06/2008] [Indexed: 01/10/2023]
Abstract
Transient ischemic attacks (TIAs) have recently become the center of attention since they are thought to share some characteristics with experimental ischemic preconditioning (IPC). This phenomenon describes the situation that a brief, per se harmless, cerebral ischemic period renders the brain resistant to a subsequent severe and normally damaging ischemia. Preconditioning (PC) is not restricted to the brain but also occurs in other organs. Furthermore, apart from a short ischemia, the PC event may comprise nearly any noxious stimulus which, however, must not exceed the threshold to tissue damage. In the last two decades, our knowledge concerning the underlying molecular basis of PC has substantially grown and there is hope to potentially imitate the induction of an endogenous neuroprotective state in patients with a high risk of cerebral ischemia. While, at present, there is virtually no neuropathological data on changes after TIAs or TIA-like PC ischemic periods in human brains, the following review will briefly summarize the current knowledge of plastic neuronal changes after PC in animal models, still awaiting their detection in the human brain.
Collapse
|