1
|
Guo Y, Li J, Liu X, Ding H, Zhang W. Potential therapeutic targets for ischemic stroke in pre-clinical studies: Epigenetic-modifying enzymes DNMT/TET and HAT/HDAC. Front Pharmacol 2025; 16:1571276. [PMID: 40356977 PMCID: PMC12066669 DOI: 10.3389/fphar.2025.1571276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and disability worldwide, driven by genetic predispositions and environmental interactions, with epigenetics playing a pivotal role in mediating these processes. Specific modifying enzymes that regulate epigenetic changes have emerged as promising targets for IS treatment. DNA methyltransferases (DNMTs), ten-eleven translocation (TET) dioxygenases, histone acetyltransferases (HATs), and histone deacetylases (HDACs) are central to epigenetic regulation. These enzymes maintain a dynamic balance between DNA methylation/demethylation and histone acetylation/deacetylation, which critically influences gene expression and neuronal survival in IS. This review is based on both in vivo and in vitro experimental studies, exploring the roles of DNMT/TET and HAT/HDAC in IS, evaluating their potential as therapeutic targets, and discussing the use of natural compounds as modulators of these enzymes to develop novel treatment strategies.
Collapse
Affiliation(s)
- Yurou Guo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaodan Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Huang Ding
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Wei Zhang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| |
Collapse
|
2
|
Aksan B, Mauceri D. Beyond vessels: unraveling the impact of VEGFs on neuronal functions and structure. J Biomed Sci 2025; 32:33. [PMID: 40050849 PMCID: PMC11884128 DOI: 10.1186/s12929-025-01128-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025] Open
Abstract
Neurons rely on the bloodstream for essential nutrients and oxygen, which is facilitated by an intricate coupling of the neuronal and vascular systems. Central to this neurovascular interaction is the vascular endothelial growth factor (VEGF) family, a group of secreted growth factors traditionally known for their roles in promoting endothelial cell proliferation, migration, and survival in the cardiovascular and lymphatic systems. However, emerging evidence shows that VEGFs also play indispensable roles in the nervous system, extending beyond their canonical angiogenic and lymphangiogenic functions. Over the past two decades, VEGFs have been found to exert direct effects on neurons, influencing key aspects of neuronal function independently of their actions on vascular cells. In particular, it has become increasingly evident that VEGFs also play crucial functions in the development, regulation, and maintenance of neuronal morphology. Understanding the roles of VEGFs in neuronal development is of high scientific and clinical interest because of the significance of precise neuronal morphology for neural connectivity and network function, as well as the association of morphological abnormalities with neurological and neurodegenerative disorders. This review begins with an overview of the VEGF family members, their structural characteristics, receptors, and established roles in vasculature. However, it then highlights and focuses on the exciting variety of neuronal functions of VEGFs, especially their crucial role in the development, regulation, and maintenance of neuronal morphology.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Institute of Anatomy and Cell Biology, Dept. Molecular and Cellular Neuroscience, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
3
|
Safaei Z, Thompson GL. Histone deacetylase 4 and 5 translocation elicited by microsecond pulsed electric field exposure is mediated by kinase activity. Front Bioeng Biotechnol 2022; 10:1047851. [PMID: 36466344 PMCID: PMC9713944 DOI: 10.3389/fbioe.2022.1047851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/04/2022] [Indexed: 10/17/2023] Open
Abstract
Electroporation-based technologies using microsecond pulsed electric field (µsPEF) exposures are established as laboratory and clinical tools that permeabilize cell membranes. We demonstrate a µsPEF bioeffect on nucleocytoplasmic import and export of enzymes that regulate genetic expression, histone deacetylases (HDAC) -4 and -5. Their μsPEF-induced nucleocytoplasmic transport depends on presence and absence of extracellular calcium ions (Ca2+) for both MCF7 and CHO-K1 cells. Exposure to 1, 10, 30 and 50 consecutive square wave pulses at 1 Hz and of 100 µs duration with 1.45 kV/cm magnitude leads to translocation of endogenous HDAC4 and HDAC5. We posit that by eliciting a rise in intracellular Ca2+ concentration, a signaling pathway involving kinases, such as Ca2+/CaM-dependent protein kinase II (CaMKII), is activated. This cascade causes nuclear export and import of HDAC4 and HDAC5. The potential of µsPEF exposures to control nucleocytoplasmic transport unlocks future opportunities in epigenetic modification.
Collapse
Affiliation(s)
| | - Gary L. Thompson
- Department of Chemical Engineering, Rowan University, Glassboro, NJ, United States
| |
Collapse
|
4
|
Srivastava A, Banerjee J, Dubey V, Tripathi M, Chandra PS, Sharma MC, Lalwani S, Siraj F, Doddamani R, Dixit AB. Role of Altered Expression, Activity and Sub-cellular Distribution of Various Histone Deacetylases (HDACs) in Mesial Temporal Lobe Epilepsy with Hippocampal Sclerosis. Cell Mol Neurobiol 2022; 42:1049-1064. [PMID: 33258018 PMCID: PMC11441253 DOI: 10.1007/s10571-020-00994-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDACs) have been described to have both neurotoxic and neuroprotective roles, and partly, depend on its sub-cellular distribution. HDAC inhibitors have a long history of use in the treatment of various neurological disorders including epilepsy. Key role of HDACs in GABAergic neurotransmission, synaptogenesis, synaptic plasticity and memory formation was demonstrated whereas very less is known about their role in drug-resistant epilepsy pathologies. The present study was aimed to investigate the changes in the expression of HDACs, activity and its sub-cellular distribution in mesial temporal lobe epilepsy with hippocampal sclerosis (MTLE-HS) patients. For this study, surgically resected hippocampal tissue specimens of 28 MTLE-HS patients and 20 hippocampus from post-mortem cases were obtained. Real-time PCR was done to analyse the mRNA expression. HDAC activity and the protein levels of HDACs in cytoplasm as well as nucleus were measured spectrophotometrically. Further, sub-cellular localization of HDACs was characterized by immunofluorescence. Significant upregulation of HDAC1, HDAC2, HDAC4, HDAC5, HDAC6, HDAC10 and HDAC11 mRNA were observed in MTLE-HS. Alterations in the mRNA expression of glutamate and gamma-aminobutyric acid (GABA) receptor subunits have been also demonstrated. We observed significant increase of HDAC activity and nuclear level of HDAC1, HDAC2, HDAC5 and HDAC11 in the hippocampal samples obtained from patients with MTLE-HS. Moreover, we found altered cytoplasmic level of HDAC4, HDAC6 and HDAC10 in the hippocampal sample obtained from patients with MTLE-HS. Alterations in the level of HDACs could potentially be part of a dynamic transcription regulation associated with MTLE-HS. Changes in cytoplasmic level of HDAC4, 6 and 10 suggest that cytoplasmic substrates may play a crucial role in the pathophysiology of MTLE-HS. Knowledge regarding expression pattern and sub-cellular distribution of HDACs may help to devise specific HDACi therapy for epilepsy.
Collapse
Affiliation(s)
- Arpna Srivastava
- Centre of Excellence for Epilepsy, AIIMS, New Delhi, India
- Department of Neurosurgery, AIIMS, New Delhi, India
| | - Jyotirmoy Banerjee
- Centre of Excellence for Epilepsy, AIIMS, New Delhi, India
- Department of Biophysics, AIIMS, New Delhi, India
| | - Vivek Dubey
- Department of Biophysics, AIIMS, New Delhi, India
| | - Manjari Tripathi
- Centre of Excellence for Epilepsy, AIIMS, New Delhi, India
- Department of Neurology, AIIMS, New Delhi, India
| | - P Sarat Chandra
- Centre of Excellence for Epilepsy, AIIMS, New Delhi, India
- Department of Neurosurgery, AIIMS, New Delhi, India
| | - M C Sharma
- Department of Pathology, AIIMS, New Delhi, India
| | - Sanjeev Lalwani
- Department of Forensic Medicine and Toxicology, AIIMS, New Delhi, India
| | - Fouzia Siraj
- National Institute of Pathology, New Delhi, India
| | | | - Aparna Banerjee Dixit
- Centre of Excellence for Epilepsy, AIIMS, New Delhi, India.
- Dr B R Ambedkar Centre for Biomedical Research, University of Delhi, New Delhi, India.
| |
Collapse
|
5
|
Litke C, Hagenston AM, Kenkel AK, Paldy E, Lu J, Kuner R, Mauceri D. Organic anion transporter 1 is an HDAC4-regulated mediator of nociceptive hypersensitivity in mice. Nat Commun 2022; 13:875. [PMID: 35169129 PMCID: PMC8847565 DOI: 10.1038/s41467-022-28357-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 01/17/2022] [Indexed: 01/26/2023] Open
Abstract
Persistent pain is sustained by maladaptive changes in gene transcription resulting in altered function of the relevant circuits; therapies are still unsatisfactory. The epigenetic mechanisms and affected genes linking nociceptive activity to transcriptional changes and pathological sensitivity are unclear. Here, we found that, among several histone deacetylases (HDACs), synaptic activity specifically affects HDAC4 in murine spinal cord dorsal horn neurons. Noxious stimuli that induce long-lasting inflammatory hypersensitivity cause nuclear export and inactivation of HDAC4. The development of inflammation-associated mechanical hypersensitivity, but neither acute nor basal sensitivity, is impaired by the expression of a constitutively nuclear localized HDAC4 mutant. Next generation RNA-sequencing revealed an HDAC4-regulated gene program comprising mediators of sensitization including the organic anion transporter OAT1, known for its renal transport function. Using pharmacological and molecular tools to modulate OAT1 activity or expression, we causally link OAT1 to persistent inflammatory hypersensitivity in mice. Thus, HDAC4 is a key epigenetic regulator that translates nociceptive activity into sensitization by regulating OAT1, which is a potential target for pain-relieving therapies. Chronic pain is sustained by alterations in gene transcription. Here, the authors show that increased expression of Organic Anionic Transporter 1 in the spinal cord is epigenetically controlled and key to hypersensitivity in pathological pain.
Collapse
Affiliation(s)
- Christian Litke
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Anna M Hagenston
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Ann-Kristin Kenkel
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Eszter Paldy
- Institute of Pharmacology, Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Jianning Lu
- Institute of Pharmacology, Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Rohini Kuner
- Institute of Pharmacology, Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Center for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
| |
Collapse
|
6
|
Paeoniflorin ameliorates ischemic injury in rat brain via inhibiting cytochrome c/caspase3/HDAC4 pathway. Acta Pharmacol Sin 2022; 43:273-284. [PMID: 33976387 PMCID: PMC8791966 DOI: 10.1038/s41401-021-00671-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/24/2021] [Indexed: 02/03/2023]
Abstract
Paeoniflorin (PF), a bioactive monoterpene glucoside, has shown a variety of pharmacological effects such as anti-inflammation and autophagy modulation etc. In this study, we investigated whether and how PF exerted a protective effect against ischemic brain injury in vivo and in vitro. Primary rat cortical neurons underwent oxygen/glucose deprivation/reperfusion (OGD/R) for 90 min. We showed that after OGD/R, a short fragment of histone deacetylase 4 (HDAC4) produced by caspase3-mediated degradation was markedly accumulated in the nucleus and the activity of caspase3 was increased. Treatment with PF (100 nM, 1 μM) significantly improved the viability of cortical neurons after OGD/R. Furthermore, PF treatment could maintain HDAC4 intrinsic subcellular localization and reduce the caspase3 activity without changing the HDAC4 at the transcriptional level. PF treatment significantly reduced OGD/R-caused inhibition of transcriptional factor MEF2 expression and increased the expression of downstream proteins such as GDNF, BDNF, and Bcl-xl, thus exerting a great anti-apoptosis effect as revealed by TUNEL staining. The beneficial effects of PF were almost canceled in HDAC4 (D289E)-transfected PC12 cells after OGD/R. In addition, PF treatment reduced the caspase9 activity, rescued the release of cytochrome c from mitochondria, and maintained the integrity of mitochondria membrane. We conducted in vivo experiments in 90-min-middle cerebral artery occlusion (MCAO) rat model. The rats were administered PF (20, 40 mg/kg, ip, 3 times at the reperfusion, 24 h and 48 h after the surgery). We showed that PF administration dose-dependently reduced infarction area, improved neurological symptoms, and maintained HDAC4 localization in rats after MCAO. These results demonstrate that PF is effective in protecting against ischemic brain injury and inhibit apoptosis through inhibiting the cytochrome c/caspase3/HDAC4 pathway.
Collapse
|
7
|
Demyanenko S, Dzreyan V, Sharifulina S. Histone Deacetylases and Their Isoform-Specific Inhibitors in Ischemic Stroke. Biomedicines 2021; 9:biomedicines9101445. [PMID: 34680562 PMCID: PMC8533589 DOI: 10.3390/biomedicines9101445] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 01/01/2023] Open
Abstract
Cerebral ischemia is the second leading cause of death in the world and multimodal stroke therapy is needed. The ischemic stroke generally reduces the gene expression due to suppression of acetylation of histones H3 and H4. Histone deacetylases inhibitors have been shown to be effective in protecting the brain from ischemic damage. Histone deacetylases inhibitors induce neurogenesis and angiogenesis in damaged brain areas promoting functional recovery after cerebral ischemia. However, the role of different histone deacetylases isoforms in the survival and death of brain cells after stroke is still controversial. This review aims to analyze the data on the neuroprotective activity of nonspecific and selective histone deacetylase inhibitors in ischemic stroke.
Collapse
|
8
|
Abstract
Neuroepigenetics, a new branch of epigenetics, plays an important role in the regulation of gene expression. Neuroepigenetics is associated with holistic neuronal function and helps in formation and maintenance of memory and learning processes. This includes neurodevelopment and neurodegenerative defects in which histone modification enzymes appear to play a crucial role. These modifications, carried out by acetyltransferases and deacetylases, regulate biologic and cellular processes such as apoptosis and autophagy, inflammatory response, mitochondrial dysfunction, cell-cycle progression and oxidative stress. Alterations in acetylation status of histone as well as non-histone substrates lead to transcriptional deregulation. Histone deacetylase decreases acetylation status and causes transcriptional repression of regulatory genes involved in neural plasticity, synaptogenesis, synaptic and neural plasticity, cognition and memory, and neural differentiation. Transcriptional deactivation in the brain results in development of neurodevelopmental and neurodegenerative disorders. Mounting evidence implicates histone deacetylase inhibitors as potential therapeutic targets to combat neurologic disorders. Recent studies have targeted naturally-occurring biomolecules and micro-RNAs to improve cognitive defects and memory. Multi-target drug ligands targeting HDAC have been developed and used in cell-culture and animal-models of neurologic disorders to ameliorate synaptic and cognitive dysfunction. Herein, we focus on the implications of histone deacetylase enzymes in neuropathology, their regulation of brain function and plausible involvement in the pathogenesis of neurologic defects.
Collapse
|
9
|
Demyanenko S, Sharifulina S. The Role of Post-Translational Acetylation and Deacetylation of Signaling Proteins and Transcription Factors after Cerebral Ischemia: Facts and Hypotheses. Int J Mol Sci 2021; 22:ijms22157947. [PMID: 34360712 PMCID: PMC8348732 DOI: 10.3390/ijms22157947] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Histone deacetylase (HDAC) and histone acetyltransferase (HAT) regulate transcription and the most important functions of cells by acetylating/deacetylating histones and non-histone proteins. These proteins are involved in cell survival and death, replication, DNA repair, the cell cycle, and cell responses to stress and aging. HDAC/HAT balance in cells affects gene expression and cell signaling. There are very few studies on the effects of stroke on non-histone protein acetylation/deacetylation in brain cells. HDAC inhibitors have been shown to be effective in protecting the brain from ischemic damage. However, the role of different HDAC isoforms in the survival and death of brain cells after stroke is still controversial. HAT/HDAC activity depends on the acetylation site and the acetylation/deacetylation of the main proteins (c-Myc, E2F1, p53, ERK1/2, Akt) considered in this review, that are involved in the regulation of cell fate decisions. Our review aims to analyze the possible role of the acetylation/deacetylation of transcription factors and signaling proteins involved in the regulation of survival and death in cerebral ischemia.
Collapse
Affiliation(s)
- Svetlana Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, pr. Stachki 194/1, 344090 Rostov-on-Don, Russia
| | - Svetlana Sharifulina
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, pr. Stachki 194/1, 344090 Rostov-on-Don, Russia
- Neuroscience Center HiLife, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, 00014 Helsinki, Finland
| |
Collapse
|
10
|
Klingl YE, Pakravan D, Van Den Bosch L. Opportunities for histone deacetylase inhibition in amyotrophic lateral sclerosis. Br J Pharmacol 2021; 178:1353-1372. [PMID: 32726472 PMCID: PMC9327724 DOI: 10.1111/bph.15217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. ALS patients suffer from a progressive loss of motor neurons, leading to respiratory failure within 3 to 5 years after diagnosis. Available therapies only slow down the disease progression moderately or extend the lifespan by a few months. Epigenetic hallmarks have been linked to the disease, creating an avenue for potential therapeutic approaches. Interference with one class of epigenetic enzymes, histone deacetylases, has been shown to affect neurodegeneration in many preclinical models. Consequently, it is crucial to improve our understanding about histone deacetylases and their inhibitors in (pre)clinical models of ALS. We conclude that selective inhibitors with high tolerability and safety and sufficient blood-brain barrier permeability will be needed to interfere with both epigenetic and non-epigenetic targets of these enzymes. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Yvonne E. Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| | - Donya Pakravan
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| |
Collapse
|
11
|
Zhong L, Yan J, Li H, Meng L. HDAC9 Silencing Exerts Neuroprotection Against Ischemic Brain Injury via miR-20a-Dependent Downregulation of NeuroD1. Front Cell Neurosci 2021; 14:544285. [PMID: 33584204 PMCID: PMC7873949 DOI: 10.3389/fncel.2020.544285] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022] Open
Abstract
Cerebral stroke is an acute cerebrovascular disease that is a leading cause of death and disability worldwide. Stroke includes ischemic stroke and hemorrhagic strokes, of which the incidence of ischemic stroke accounts for 60–70% of the total number of strokes. Existing preclinical evidence suggests that inhibitors of histone deacetylases (HDACs) are a promising therapeutic intervention for stroke. In this study, the purpose was to investigate the possible effect of HDAC9 on ischemic brain injury, with the underlying mechanism related to microRNA-20a (miR-20a)/neurogenic differentiation 1 (NeuroD1) explored. The expression of HDAC9 was first detected in the constructed middle cerebral artery occlusion (MCAO)-provoked mouse model and oxygen-glucose deprivation (OGD)-induced cell model. Next, primary neuronal apoptosis, expression of apoptosis-related factors (Bax, cleaved caspase3 and bcl-2), LDH leakage rate, as well as the release of inflammatory factors (TNF-α, IL-1β, and IL-6) were evaluated by assays of TUNEL, Western blot, and ELISA. The relationships among HDAC9, miR-20a, and NeuroD1 were validated by in silico analysis and ChIP assay. HDAC9 was highly-expressed in MCAO mice and OGD-stimulated cells. Silencing of HDAC9 inhibited neuronal apoptosis and inflammatory factor release in vitro. HDAC9 downregulated miR-20a by enriching in its promoter region, while silencing of HDCA9 promoted miR-20a expression. miR-20a targeted Neurod1 and down-regulated its expression. Silencing of HDAC9 diminished OGD-induced neuronal apoptosis and inflammatory factor release in vitro as well as ischemic brain injury in vivo by regulating the miR-20a/NeuroD1 signaling. Overall, our study revealed that HDAC9 silencing could retard ischemic brain injury through the miR-20a/Neurod1 signaling.
Collapse
Affiliation(s)
- Liangjun Zhong
- Department of Neurosurgery, Pingyin County People's Hospital, Jinan, China
| | - Jinxiang Yan
- Department of Neurosurgery, Ningyang No. 1 People's Hospital, Tai'an, China
| | - Haitao Li
- Department of Neurology, Qihe County People's Hospital, Dezhou, China
| | - Lei Meng
- Department of Neurosurgery, Shandong Provincial Hospital, Jinan, China
| |
Collapse
|
12
|
Lv H, Li Y, Cheng Q, Chen J, Chen W. Neuroprotective Effects Against Cerebral Ischemic Injury Exerted by Dexmedetomidine via the HDAC5/NPAS4/MDM2/PSD-95 Axis. Mol Neurobiol 2021; 58:1990-2004. [PMID: 33411316 DOI: 10.1007/s12035-020-02223-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/19/2020] [Indexed: 01/16/2023]
Abstract
Numerous evidences have highlighted the efficient role of dexmedetomidine (DEX) in multi-organ protection. In the present study, the neuroprotective role of DEX on cerebral ischemic injury and the underlining signaling mechanisms were explored. In order to simulate cerebral ischemic injury, we performed middle cerebral artery occlusion in mice and oxygen-glucose deprivation in neurons. Immunohistochemistry, Western blot analysis, and RT-qPCR were used to examine expression of HDAC5, NPAS4, MDM2, and PSD-95 in hippocampus tissues of MCAO mice and OGD-treated neurons. MCAO mice received treatment with DEX and sh-PSD-95, followed by neurological function evaluation, behavioral test, infarct volume detection by TTC staining, and apoptosis by TUNEL staining. Additionally, gain- and loss-of-function approaches were conducted in OGD-treated neuron after DEX treatment. Cell viability and apoptosis were assessed with the application of CCK-8 and flow cytometry. The interaction between MDM2 and PSD-95 was evaluated using Co-IP assay, followed by ubiquitination of PSD-95 detection. As per the results, HDAC5 and MDM2 were abundantly expressed, while NPAS4 and PSD-95 were poorly expressed in hippocampus tissues of MCAO mice and OGD-treated neurons. DEX elevated viability, and reduced LDH leakage rate and apoptosis rate of OGD-treated neurons, which was reversed following the overexpression of HDAC5. Moreover, HDAC5 augmented MDM2 expression via NPAS4 inhibition. MDM2 induced PSD-95 ubiquitination and degradation. In MCAO mice, DEX improved neurological function and behaviors and decreased infarct volume and apoptosis, which was negated as a result of PSD-95 silencing. DEX plays a neuroprotective role against cerebral ischemic injury by disrupting MDM2-induced PSD-95 ubiquitination and degradation via HDAC5 and NPAS4.
Collapse
Affiliation(s)
- Hu Lv
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Ying Li
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Qian Cheng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China
| | - Jiawei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.
| | - Wei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, No. 270, Dong'an Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
13
|
Lin YH, Yao MC, Wu HY, Dong J, Ni HY, Kou XL, Chang L, Luo CX, Zhu DY. HDAC2 (Histone deacetylase 2): A critical factor in environmental enrichment-mediated stroke recovery. J Neurochem 2020; 155:679-696. [PMID: 32415988 DOI: 10.1111/jnc.15043] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/09/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022]
Abstract
Environmental enrichment (EE) is a generally accepted strategy to promote stroke recovery and its beneficial effect is positively correlated with neuroplasticity. However, the mechanisms underlying it remain elusive. Histone deacetylase 2 (HDAC2), a negative regulator of neuroplasticity, is up-regulated after stroke. Thus, we hypothesized that HDAC2 may participate in EE-mediated stroke recovery. In this study, focal stroke was induced by photothrombosis in male mice exposing to EE or standard housing (SH) conditions. Recombinant virus vectors, including Ad-HDAC2-Flag, AAV-CAG-EGFP-Cre, LV-shHDAC2, or their controls were microinjected into the motor cortex at 3 days before stroke. Grid-walking and cylinder tasks were conducted to assess motor function. Western blot and immunostaining were used to uncover the mechanisms underlying EE-mediated stroke recovery. We found that EE exposure reversed stroke-induced HDAC2 up-regulation, implicating HDAC2 in EE-mediated functional recovery. Importantly, EE-dependent stroke recovery was counteracted by over-expressing HDAC2, and HDAC2 knockdown promoted functional recovery from stroke to the similar extent as EE exposure. Moreover, the knockdown of HDAC2 epigenetically enhanced expressions of neurotrophins and neuroplasticity-related proteins, with similar effects as EE, and consequently, whole brain and corticospinal tract (CST) rewiring. Together, our findings indicate that HDAC2 is critical for EE-dependent functional restoration. Precisely targeting HDAC2 may mimic EE and serve as a novel therapeutic strategy for stroke recovery.
Collapse
Affiliation(s)
- Yu-Hui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Meng-Cheng Yao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Hai-Yin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jian Dong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Huan-Yu Ni
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Xiao-Lin Kou
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Chun-Xia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China
| | - Dong-Ya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China.,Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Luo P, Fu X, Chang M, Zhang L, Guo L. Cerebral ischemia-reperfusion causes a down regulation of HCN1 expression via enhancing the nuclear NRSF-HDAC4 gathering that contributes to neuron damage. Brain Res Bull 2020; 156:50-57. [PMID: 31923455 DOI: 10.1016/j.brainresbull.2020.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/21/2019] [Accepted: 01/02/2020] [Indexed: 11/30/2022]
Abstract
Cerebral ischemia-reperfusion (I/R) can trigger neuronal death through several biologically plausible pathways, but its underlying neurobiological mechanisms remain unclear. In this study, we tested whether hyperpolarization-activated cyclic nucleotide-gated channel 1 (HCN1) is altered in I/R that contributes to neuron damage and further clarified the mechanisms underlying this process. Cerebral I/R injury was induced by middle cerebral artery occlusion (MCAO) surgery followed by reperfusion in rats or simulated by oxygen-glucose deprivation/reoxygenation (OGD/R) in cultured cell. After reperfusion, the mRNA and protein levels of HCN1 were tested by RT-PCR and Western blot (WB). The histone deacetylases 4 (HDAC4) shuttling and the nuclear neuron-restrictive silencer factor (NRSF) expression were evaluated by WB and immunohistochemistry. Our data showed that I/R caused a strong decrease of HCN1 subunit in both hippocampus and cortex of rat. Additionally, the nuclear expression of HDAC4 and NRSF were significantly increased. In vitro OGD/R model, the gathering of HDAC4 and NRSF to nuclei was further confirmed. Valproic acid (VPA), a HDAC4 inhibitor, could reverse the decreased HCN1 and protect neuron damage from OGD/R injury. Collectively, these results demonstrated that I/R cause a decrease of HCN1 expression via enhancing nuclear HDAC4-NRSF gathering and might contribute to neuron damage.
Collapse
Affiliation(s)
- Pan Luo
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaopei Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mujun Chang
- Center for Translational Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Zhang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lianjun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Wang Y, Xia Y, Hu K, Zeng M, Zhi C, Lai M, Wu L, Liu S, Zeng S, Huang Z, Ma S, Yuan Z. MKK7 transcription positively or negatively regulated by SP1 and KLF5 depends on HDAC4 activity in glioma. Int J Cancer 2019; 145:2496-2508. [PMID: 30963560 DOI: 10.1002/ijc.32321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/11/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022]
Abstract
JNK activity has been implicated in the malignant proliferation, invasion and drug-resistance of glioma cells (GCs), but the molecular mechanisms underlying JNK activation are currently unknown. Here, we reported that MKK7, not MKK4, directly activates JNK in GCs and exerts oncogenic effects on tumor formation. Notably, MKK7 expression in glioma tissues was closely correlated with the grade of the glioma and JNK/c-Jun activation. Mechanistically, MKK7 transcription critically depends on the complexes formed by HDAC4 and the transcriptional factors SP1 and Krüppel-like factor-5 (KLF5), wherein HDAC4 directly deacetylates both SP1 and KLF5 and synergistically upregulates MKK7 transcription through two SP1 sites located on its promoter. In contrast, the increases in acetylated-SP1 and acetylated-KLF5 after HDAC4 inhibition switched to transcriptionally suppress MKK7. Selective inhibition of HDAC4 by LMK235, siRNAs or blockage of SP1 and KLF5 by the ectopic dominant-negative SP1 greatly reduced the malignant capacity of GCs. Furthermore, suppression of both MKK7 expression and JNK/c-Jun activities was involved in the tumor-growth inhibitory effects induced by LMK235 in U87-xenograft mice. Interestingly, HDAC4 is highly expressed in glioma tissues, and the rate of HDAC4 nuclear import is closely correlated with glioma grade, as well as with MKK7 expression. Collectively, these findings demonstrated that highly expressed MKK7 contributes to JNK/c-Jun signaling-mediated glioma formation. MKK7 transcription, regulated by SP1 and KLF5, critically depends on HDAC4 activity, and inhibition of HDAC4 presents a potential strategy for suppressing the oncogenic roles of MKK7/JNK/c-Jun signaling in GCs.
Collapse
Affiliation(s)
- Yezhong Wang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Yong Xia
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Kunhua Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| | - Minling Zeng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Cheng Zhi
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Miaoling Lai
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liqiang Wu
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Sisi Liu
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Shulian Zeng
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Ziyan Huang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
| | - Shanshan Ma
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| | - Zhongmin Yuan
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Institute of Neurosciences of Guangzhou Medical University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China
| |
Collapse
|
16
|
Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF, Andjelkovic AV. Involvement of Epigenetic Mechanisms and Non-coding RNAs in Blood-Brain Barrier and Neurovascular Unit Injury and Recovery After Stroke. Front Neurosci 2019; 13:864. [PMID: 31543756 PMCID: PMC6732937 DOI: 10.3389/fnins.2019.00864] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cessation of blood flow leads to a complex cascade of pathophysiological events at the blood-vascular-parenchymal interface which evolves over time and space, and results in damage to neural cells and edema formation. Cerebral ischemic injury evokes a profound and deleterious upregulation in inflammation and triggers multiple cell death pathways, but it also induces a series of the events associated with regenerative responses, including vascular remodeling, angiogenesis, and neurogenesis. Emerging evidence suggests that epigenetic reprograming could play a pivotal role in ongoing post-stroke neurovascular unit (NVU) changes and recovery. This review summarizes current knowledge about post-stroke recovery processes at the NVU, as well as epigenetic mechanisms and modifiers (e.g., DNA methylation, histone modifying enzymes and microRNAs) associated with stroke injury, and NVU repair. It also discusses novel drug targets and therapeutic strategies for enhancing post-stroke recovery.
Collapse
Affiliation(s)
- Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chelsea M. Phillips
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
17
|
Demyanenko S, Berezhnaya E, Neginskaya M, Rodkin S, Dzreyan V, Pitinova M. Сlass II histone deacetylases in the post-stroke recovery period-expression, cellular, and subcellular localization-promising targets for neuroprotection. J Cell Biochem 2019; 120:19590-19609. [PMID: 31264264 DOI: 10.1002/jcb.29266] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/12/2019] [Indexed: 12/13/2022]
Abstract
Histone deacetylases (HDAC) inhibitors can protect nerve cells after a stroke, but it is unclear which HDAC isoform is involved in this effect. We studied cellular and intracellular rearrangement of class II HDACs at late periods after photothrombotic infarct (PTI) in the mouse sensorimotor cortex in the tissue surrounding the ischemia core and in the corresponding region of the contralateral hemisphere. We observed a decrease in HDAC4 in cortical neurons and an increase in its nuclear translocation. HDAC6 expression in neurons was also increased. Moreover, HDAC6-positive cells had elevated apoptosis. Tubostatin A (Tub A)-induced decrease in the activity of HDAC6 restored acetylation of α-tubulin during the early poststroke recovery period and reduced apoptosis of nerve cells thus protecting the brain tissue. Selective inhibition of HDAC6 elevated expression of growth-associated protein-43 (GAP43), which remained high up to 14 days after stroke and promoted axogenesis and recovery from the PTI-induced neurological deficit. Selective HDAC6 inhibitor Tub A markedly reduced neuronal death and increased acetylation of α-tubulin and the level of GAP43. Thus, HDAC6 inhibition could be a promising strategy for modulation of brain recovery as it can increase the intensity and reduce the duration of reparation processes in the brain after stroke.
Collapse
Affiliation(s)
- Svetlana Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Elena Berezhnaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Maria Neginskaya
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Stanislav Rodkin
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Valentina Dzreyan
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| | - Maria Pitinova
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, Russia
| |
Collapse
|
18
|
Demyanenko S, Uzdensky A. Epigenetic Alterations Induced by Photothrombotic Stroke in the Rat Cerebral Cortex: Deacetylation of Histone h3, Upregulation of Histone Deacetylases and Histone Acetyltransferases. Int J Mol Sci 2019; 20:E2882. [PMID: 31200484 PMCID: PMC6627403 DOI: 10.3390/ijms20122882] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/03/2019] [Accepted: 06/09/2019] [Indexed: 12/12/2022] Open
Abstract
Ischemic penumbra that surrounds a stroke-induced infarction core is potentially salvageable; however, mechanisms of its formation are not well known. Covalent modifications of histones control chromatin conformation, gene expression and protein synthesis. To study epigenetic processes in ischemic penumbra, we used photothrombotic stroke (PTS), a stroke model in which laser irradiation of the rat brain cortex photosensitized by Rose Bengal induces local vessel occlusion. Immunoblotting and immunofluorescence microscopy showed decrease in acetylation of lysine 9 in histone H3 in penumbra at 1, 4 or 24 h after PTS. This was associated with upregulation of histone deacetylases HDAC1 and HDAC2, but not HDAC4, which did not localize in the nuclei. HDAC2 was found in cell nuclei, HDAC4 in the cytoplasm and HDAC1 both in nuclei and cytoplasm. Histone acetyltransferases HAT1 and PCAF (p300/CBP associated factor) that acetylated histone H3 synthesis were also upregulated, but lesser and later. PTS increased localization of HDAC2 and HAT1 in astroglia. Thus, the cell fate in PTS-induced penumbra is determined by the balance between opposite tendencies leading either to histone acetylation and stimulation of gene expression, or to deacetylation and suppression of transcriptional processes and protein biosynthesis. These epigenetic proteins may be the potential targets for anti-stroke therapy.
Collapse
Affiliation(s)
- Svetlana Demyanenko
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky ave., Rostov-on-Don 344090, Russia.
| | - Anatoly Uzdensky
- Laboratory of Molecular Neurobiology, Academy of Biology and Biotechnology, Southern Federal University, 194/1 Stachky ave., Rostov-on-Don 344090, Russia.
| |
Collapse
|
19
|
Schlüter A, Aksan B, Fioravanti R, Valente S, Mai A, Mauceri D. Histone Deacetylases Contribute to Excitotoxicity-Triggered Degeneration of Retinal Ganglion Cells In Vivo. Mol Neurobiol 2019; 56:8018-8034. [PMID: 31161423 DOI: 10.1007/s12035-019-01658-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/20/2019] [Indexed: 02/06/2023]
Abstract
Excitotoxicity is known to modulate the nuclear accumulation, and thus activity state, of histone deacetylases (HDACs) in pyramidal neurons. In the retina, deregulation in activity and expression of different HDACs has been linked to pathological conditions such as retinitis pigmentosa, retinal ischemia, glaucoma, and acute optic nerve injury. Up to now, however, the effects of in vivo excitotoxicity on the different HDACs in retinal ganglion cells (RGCs) have not been thoroughly investigated. Here, we injected adult mice intravitreally with N-methyl-D-aspartate (NMDA) as a mean to trigger excitotoxicity-mediated RGC degeneration and we detected time-dependent loss of RGCs at 1 and 7 days after the insult. Further, we characterized the subcellular localization of HDACs belonging to class I (HDAC1, HDAC3), IIa (HDAC4, HDAC5, HDAC7, HDAC9), IIb (HDAC6, HDAC10), and IV (HDAC11) in RGCs. Our analyses revealed a differential pattern of HDACs nuclear distribution in RGCs following excitotoxicity. After 1 day, HDAC3, HDAC5, HDAC6, HDAC7, and HDAC11 showed altered subcellular localization in RGCs while 7 days after the excitotoxic insult, HDAC4 and HDAC9 were the only HDACs displaying changes in their subcellular distribution. Moreover, we found that in vivo selective inhibition of HDAC1/3 or HDAC4/5 via MS-275 (entinostat) or LMK-235, respectively, could prevent ongoing RGC degeneration. In conclusion, our results point towards a role of HDACs in RGC degeneration and identify HDAC1/3 and HDAC4/5 as potential therapeutic targets to treat degenerative retinal diseases.
Collapse
Affiliation(s)
- Annabelle Schlüter
- Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Bahar Aksan
- Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany
| | - Rossella Fioravanti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Daniela Mauceri
- Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Im Neuenheimer Feld 366, 69120, Heidelberg, Germany.
| |
Collapse
|
20
|
HDAC4 in ischemic stroke: mechanisms and therapeutic potential. Clin Epigenetics 2018; 10:117. [PMID: 30208931 PMCID: PMC6136233 DOI: 10.1186/s13148-018-0549-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/28/2018] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of death and disability worldwide, and the majority of the cases are ischemic stroke. However, it still lacks effective treatment except for thrombolytic therapy in an extremely narrow time window. Increased evidence suggests that histone deacetylase 4 (HDAC4) was dysregulated in ischemic stroke, which plays a key role in the pathogenesis of ischemic stroke and post-stroke recovery by affecting neuronal death, angiogenesis, and neurogenesis. Therefore, we aim to review the dysregulation of HDAC4 in ischemic stroke and the role of dysregulated HDAC4 in the pathogenesis of ischemic stroke. Furthermore, the therapeutic potential of modulating HDAC4 in ischemic stroke is discussed.
Collapse
|
21
|
Litke C, Bading H, Mauceri D. Histone deacetylase 4 shapes neuronal morphology via a mechanism involving regulation of expression of vascular endothelial growth factor D. J Biol Chem 2018; 293:8196-8207. [PMID: 29632070 DOI: 10.1074/jbc.ra117.001613] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/05/2018] [Indexed: 12/17/2022] Open
Abstract
Nucleo-cytoplasmic shuttling of class IIa histone deacetylases (i.e HDAC4, -5, -7, and -9) is a synaptic activity- and nuclear calcium-dependent mechanism important for epigenetic regulation of signal-regulated gene expression in hippocampal neurons. HDAC4 in particular has been linked to the regulation of genes important for both synaptic structure and plasticity. Here, using a constitutively nuclear-localized, dominant-active variant of HDAC4 (HDAC4 3SA), we demonstrate that HDAC4 accumulation in the nucleus severely reduces both the length and complexity of dendrites of cultured mature hippocampal neurons, but does not affect the number of dendritic spines. This phenomenon appeared to be specific to HDAC4, as increasing the expression of HDAC3 or HDAC11, belonging to class I and class IV HDACs, respectively, did not alter dendritic architecture. We also show that HDAC4 3SA decreases the expression of vascular endothelial growth factor D (VEGFD), a key protein required for the maintenance of dendritic arbors. The expression of other members of the VEGF family and their receptors was not affected by the nuclear accumulation of HDAC4. VEGFD overexpression or administration of recombinant VEGFD, but not VEGFC, the closest VEGFD homologue, rescued the impaired dendritic architecture caused by the nuclear-localized HDAC4 variant. These results identify HDAC4 as an epigenetic regulator of neuronal morphology that controls dendritic arborization via the expression of VEGFD.
Collapse
Affiliation(s)
- Christian Litke
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 364 69120 Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 364 69120 Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 364 69120 Heidelberg, Germany.
| |
Collapse
|
22
|
Thomas EA, D'Mello SR. Complex neuroprotective and neurotoxic effects of histone deacetylases. J Neurochem 2018; 145:96-110. [PMID: 29355955 DOI: 10.1111/jnc.14309] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/05/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
By their ability to shatter quality of life for both patients and caregivers, neurodegenerative diseases are the most devastating of human disorders. Unfortunately, there are no effective or long-terms treatments capable of slowing down the relentless loss of neurons in any of these diseases. One impediment is the lack of detailed knowledge of the molecular mechanisms underlying the processes of neurodegeneration. While some neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, are mostly sporadic in nature, driven by both environment and genetic susceptibility, many others, including Huntington's disease, spinocerebellar ataxias, and spinal-bulbar muscular atrophy, are genetically inherited disorders. Surprisingly, given their different roots and etiologies, both sporadic and genetic neurodegenerative disorders have been linked to disease mechanisms involving histone deacetylase (HDAC) proteins, which consists of 18 family members with diverse functions. While most studies have implicated certain HDAC subtypes in promoting neurodegeneration, a substantial body of literature suggests that other HDAC proteins can preserve neuronal viability. Of particular interest, however, is the recent realization that a single HDAC subtype can have both neuroprotective and neurotoxic effects. Diverse mechanisms, beyond transcriptional regulation have been linked to these effects, including deacetylation of non-histone proteins, protein-protein interactions, post-translational modifications of the HDAC proteins themselves and direct interactions with disease proteins. The roles of these HDACs in both sporadic and genetic neurodegenerative diseases will be discussed in the current review.
Collapse
Affiliation(s)
- Elizabeth A Thomas
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - Santosh R D'Mello
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas, USA
| |
Collapse
|
23
|
Interplay between mitochondrial metabolism and oxidative stress in ischemic stroke: An epigenetic connection. Mol Cell Neurosci 2017; 82:176-194. [DOI: 10.1016/j.mcn.2017.05.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 12/18/2022] Open
|
24
|
麦 汉, 姜 涛, 张 爱, 吕 田, 杨 灿, 覃 偲. [Expression of HDAC9 in different brain regions in mice with cerebral ischemic stroke]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:812-816. [PMID: 28669958 PMCID: PMC6744151 DOI: 10.3969/j.issn.1673-4254.2017.06.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To investigate the expression and the subcellular localization of HDAC9 in different brain regions of mice after cerebral ischemic injury and explore the association between HDAC9 and ischemic stroke. METHODS Twenty-one male C57BL/6 mice were randomly divided into sham-operated group (n=9) and operated group (n=12). In the latter group, the mice with Zea-Longa neurological deficit scores of 2 or 3 following middle cerebral artery occlusion (MCAO) were assigned into MCAO group (n=9). Immunofluorescence was performed to investigate the subcellular localization of HDAC9 in the brain tissues on day 3 after MCAO. Western blotting and qRT-PCR were used to analyze the expression of HDAC9 in different regions of the brain. Results Immunofluorescence showed more intense HDAC9 expressions in the brain tissues around the infarct focus, and in the cells surrounding the infarct, HDAC9 expression was obviously increased in the cytoplasm and reduced in the cell nuclei. Compared with the other brain regions, the ipsilesional cortex with MCAO showed more abundant HDAC9 expressions at both the mRNA and protein levels (P<0.05). CONCLUSION HDAC9 may be closely related to cerebral ischemic injury and participate in the pathophysiological process of ischemic stroke.
Collapse
Affiliation(s)
- 汉滔 麦
- 南方医科大学第三附属医院神经内科,广东 广州 510630Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - 涛 姜
- 南方医科大学第三附属医院神经内科,广东 广州 510630Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - 爱武 张
- 中山大学附属第一医院神经内科,广东 广州 510080Department of Neurology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - 田明 吕
- 南方医科大学第三附属医院神经内科,广东 广州 510630Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - 灿洪 杨
- 南方医科大学第三附属医院神经内科,广东 广州 510630Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - 偲偲 覃
- 南方医科大学第三附属医院神经内科,广东 广州 510630Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
25
|
Kassis H, Shehadah A, Chopp M, Zhang ZG. Epigenetics in Stroke Recovery. Genes (Basel) 2017; 8:genes8030089. [PMID: 28264471 PMCID: PMC5368693 DOI: 10.3390/genes8030089] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/02/2017] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Abstract: While the death rate from stroke has continually decreased due to interventions in the hyperacute stage of the disease, long-term disability and institutionalization have become common sequelae in the aftermath of stroke. Therefore, identification of new molecular pathways that could be targeted to improve neurological recovery among survivors of stroke is crucial. Epigenetic mechanisms such as post-translational modifications of histone proteins and microRNAs have recently emerged as key regulators of the enhanced plasticity observed during repair processes after stroke. In this review, we highlight the recent advancements in the evolving field of epigenetics in stroke recovery.
Collapse
Affiliation(s)
- Haifa Kassis
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Amjad Shehadah
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
- Department of Physics, Oakland University, Rochester, MI 48309, USA.
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| |
Collapse
|
26
|
Kassis H, Shehadah A, Li C, Zhang Y, Cui Y, Roberts C, Sadry N, Liu X, Chopp M, Zhang ZG. Class IIa histone deacetylases affect neuronal remodeling and functional outcome after stroke. Neurochem Int 2016; 96:24-31. [PMID: 27103167 DOI: 10.1016/j.neuint.2016.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/11/2016] [Accepted: 04/16/2016] [Indexed: 01/09/2023]
Abstract
We have previously demonstrated that stroke induces nuclear shuttling of class IIa histone deacetylase 4 (HDAC4). Stroke-induced nuclear shuttling of HDAC4 is positively and significantly correlated with improved indices of neuronal remodeling in the peri-infarct cortex. In this study, using a rat model for middle cerebral artery occlusion (MCAO), we tested the effects of selective inhibition of class IIa HDACs on functional recovery and neuronal remodeling when administered 24hr after stroke. Adult male Wistar rats (n = 15-17/group) were subjected to 2 h MCAO and orally gavaged with MC1568 (a selective class IIa HDAC inhibitor), SAHA (a non-selective HDAC inhibitor), or vehicle-control for 7 days starting 24 h after MCAO. A battery of behavioral tests was performed. Lesion volume measurement and immunohistochemistry were performed 28 days after MCAO. We found that stroke increased total HDAC activity in the ipsilateral hemisphere compared to the contralateral hemisphere. Stroke-increased HDAC activity was significantly decreased by the administration of SAHA as well as by MC1568. However, SAHA significantly improved functional outcome compared to vehicle control, whereas selective class IIa inhibition with MC1568 increased mortality and lesion volume and did not improve functional outcome. In addition, MC1568 decreased microtubule associated protein 2 (MAP2, dendrites), phosphorylated neurofilament heavy chain (pNFH, axons) and myelin basic protein (MBP, myelination) immunoreactivity in the peri-infarct cortex. Quantitative RT-PCR of cortical neurons isolated by laser capture microdissection revealed that MC1568, but not SAHA, downregulated CREB and c-fos expression. Additionally, MC1568 decreased the expression of phosphorylated CREB (active) in neurons. Taken together, these findings demonstrate that selective inhibition of class IIa HDACs impairs neuronal remodeling and neurological outcome. Inactivation of CREB and c-fos by MC1568 likely contributes to this detrimental effect.
Collapse
Affiliation(s)
- Haifa Kassis
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Amjad Shehadah
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Chao Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Yisheng Cui
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Cynthia Roberts
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Neema Sadry
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Xianshuang Liu
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| |
Collapse
|
27
|
Yuan H, Denton K, Liu L, Li XJ, Benashski S, McCullough L, Li J. Nuclear translocation of histone deacetylase 4 induces neuronal death in stroke. Neurobiol Dis 2016; 91:182-93. [PMID: 26969532 DOI: 10.1016/j.nbd.2016.03.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/08/2016] [Accepted: 03/07/2016] [Indexed: 10/22/2022] Open
Abstract
Mounting evidence suggests that epigenetic modifications play critical roles in the survival/death of stressed neurons. Chief among these modifications is the deacetylation of histones within the chromatin by histone deacetylases (HDACs). HDAC4 is highly expressed in neurons and is usually trapped in cytosol. However, tightly regulated signal-dependent shuttling of this molecule between cytosol and nucleus occurs. Here, we studied the intracellular trafficking of HDAC4 and regulatory mechanisms during stroke. HDAC4 translocated from the cytosol into the nucleus of neurons in response to stroke induced by middle cerebral artery occlusion (MCAO) in mice. Similar translocation was seen after oxygen-glucose deprivation (OGD) in cultured mouse neurons. Expression of nuclear-restricted HDAC4 increased neuronal death after OGD and worsened infarcts and functional deficits in mice following MCAO; however, expression of cytosolic-restricted HDAC4 did not affect outcome after ischemia. In contrast, HDAC4 knockdown with siRNA improved neuronal survival after OGD. Furthermore, expression of nuclear-restricted HDAC4 reduced the acetylation of histones 3 and 4 as well as the levels of pro-survival downstream molecules after OGD. Finally, genetic deletion of calcium/calmodulin-dependent protein kinase IV (CaMKIV) increased the nuclear accumulation of HDAC4 in MCAO model, while overexpression of CaMKIV reduced the levels of nuclear HDAC4 following OGD. When HDAC4 was inhibited, the neuroprotection provided by CaMKIV overexpression was absent during OGD. Our data demonstrate a detrimental role of the nuclear accumulation of HDAC4 following stroke and identify CaMKIV as a key regulator of neuronal intracellular HDAC4 trafficking during stroke.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Kyle Denton
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Lin Liu
- Department of Neurology, University of Texas Health Science Center, Houston, TX, United States
| | - Xue-Jun Li
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Sharon Benashski
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, United States
| | - Louise McCullough
- Department of Neurology, University of Texas Health Science Center, Houston, TX, United States
| | - Jun Li
- Department of Neurology, University of Texas Health Science Center, Houston, TX, United States.
| |
Collapse
|
28
|
Hu Z, Zhong B, Tan J, Chen C, Lei Q, Zeng L. The Emerging Role of Epigenetics in Cerebral Ischemia. Mol Neurobiol 2016; 54:1887-1905. [PMID: 26894397 DOI: 10.1007/s12035-016-9788-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 02/11/2016] [Indexed: 12/14/2022]
Abstract
Despite great progresses in the treatment and prevention of ischemic stroke, it is still among the leading causes of death and serious long-term disability all over the world, indicating that innovative neural regenerative and neuroprotective agents are urgently needed for the development of therapeutic approaches with greater efficacy for ischemic stroke. More and more evidence suggests that a spectrum of epigenetic processes play an important role in the pathophysiology of cerebral ischemia. In the present review, we first discuss recent developments in epigenetic mechanisms, especially their roles in the pathophysiology of cerebral ischemia. Specifically, we focus on DNA methylation, histone deacetylase, histone methylation, and microRNAs (miRNAs) in the regulation of vascular and neuronal regeneration after cerebral ischemia. Additionally, we highlight epigenetic strategies for ischemic stroke treatments, including the inhibition of histone deacetylase enzyme and DNA methyltransferase activities, and miRNAs. These therapeutic strategies are far from clinic use, but preliminary data indicate that neuroprotective agents targeting these pathways can modulate neural cell regeneration and promote brain repair and functional recovery after cerebral ischemia. A better understanding of how epigenetics influences the process and progress of cerebral ischemia will pave the way for discovering more sensitive and specific biomarkers and new targets and therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Zhiping Hu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Bingwu Zhong
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Department of Traditional Chinese Medicine, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jieqiong Tan
- National Key Laboratory of Medical Genetics, Central South University, Changsha, 410078, Hunan, China
| | - Chunli Chen
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qiang Lei
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Liuwang Zeng
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
29
|
Zhang ZG, Chopp M. Promoting brain remodeling to aid in stroke recovery. Trends Mol Med 2015; 21:543-8. [PMID: 26278490 PMCID: PMC4567429 DOI: 10.1016/j.molmed.2015.07.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/17/2015] [Accepted: 07/17/2015] [Indexed: 12/13/2022]
Abstract
Endogenous brain repair after stroke involves a set of highly interactive processes, such as angiogenesis, neurogenesis, oligodendrogenesis, synaptogenesis, and axonal outgrowth, which together orchestrate neurological recovery. During the past several years, there have been advances in our understanding of miRNAs and histone deacetylases (HDACs) in brain repair processes after stroke. Emerging data indicate the important role of exosomes for intercellular communication in promoting coupled brain remodeling processes. These advances will likely have a major impact on the development of restorative therapies for ischemic brain repair, consequently leading to improvement of neurological function. In this review, we provide an update on our current understanding of cellular and molecular mechanisms of miRNAs, exosomes, and HDACs in brain restorative processes after stroke.
Collapse
Affiliation(s)
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Physics, Oakland University, Rochester, MI, USA
| |
Collapse
|