1
|
Zhang LY, Zhang SY, Wen R, Zhang TN, Yang N. Role of histone deacetylases and their inhibitors in neurological diseases. Pharmacol Res 2024; 208:107410. [PMID: 39276955 DOI: 10.1016/j.phrs.2024.107410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Histone deacetylases (HDACs) are zinc-dependent deacetylases that remove acetyl groups from lysine residues of histones or form protein complexes with other proteins for transcriptional repression, changing chromatin structure tightness, and inhibiting gene expression. Recent in vivo and in vitro studies have amply demonstrated the critical role of HDACs in the cell biology of the nervous system during both physiological and pathological processes and have provided new insights into the conduct of research on neurological disease targets. In addition, in vitro and in vivo studies on HDAC inhibitors show promise for the treatment of various diseases. This review summarizes the regulatory mechanisms of HDAC and the important role of its downstream targets in nervous system diseases, and summarizes the therapeutic mechanisms and efficacy of HDAC inhibitors in various nervous system diseases. Additionally, the current pharmacological situation, problems, and developmental prospects of HDAC inhibitors are described. A better understanding of the pathogenic mechanisms of HDACs in the nervous system may reveal new targets for therapeutic interventions in diseases and help to relieve healthcare pressure through preventive measures.
Collapse
Affiliation(s)
- Li-Ying Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Sen-Yu Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ri Wen
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Tie-Ning Zhang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Ni Yang
- Department of Pediatrics, PICU, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
2
|
Raval M, Mishra S, Tiwari AK. Epigenetic regulons in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:185-247. [DOI: 10.1016/bs.pmbts.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
3
|
Yildirim F, Foddis M, Blumenau S, Müller S, Kajetan B, Holtgrewe M, Kola V, Beule D, Sassi C. Shared and oppositely regulated transcriptomic signatures in Huntington's disease and brain ischemia confirm known and unveil novel potential neuroprotective genes. Neurobiol Aging 2021; 104:122.e1-122.e17. [PMID: 33875290 DOI: 10.1016/j.neurobiolaging.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 11/20/2022]
Abstract
Huntington's disease and subcortical vascular dementia display similar dementing features, shaped by different degrees of striatal atrophy, deep white matter degeneration and tau pathology. To investigate the hypothesis that Huntington's disease transcriptomic hallmarks may provide a window into potential protective genes upregulated during brain acute and subacute ischemia, we compared RNA sequencing signatures in the most affected brain areas of 2 widely used experimental mouse models: Huntington's disease, (R6/2, striatum and cortex and Q175, hippocampus) and brain ischemia-subcortical vascular dementia (BCCAS, striatum, cortex and hippocampus). We identified a cluster of 55 shared genes significantly differentially regulated in both models and we screened these in 2 different mouse models of Alzheimer's disease, and 96 early-onset familial and apparently sporadic small vessel ischemic disease patients. Our data support the prevalent role of transcriptional regulation upon genetic coding variability of known neuroprotective genes (Egr2, Fos, Ptgs2, Itga5, Cdkn1a, Gsn, Npas4, Btg2, Cebpb) and provide a list of potential additional ones likely implicated in different dementing disorders and worth further investigation.
Collapse
Affiliation(s)
- Ferah Yildirim
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanne Müller
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bentele Kajetan
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Manuel Holtgrewe
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Vasilis Kola
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
4
|
Hecklau K, Mueller S, Koch SP, Mehkary MH, Kilic B, Harms C, Boehm-Sturm P, Yildirim F. The Effects of Selective Inhibition of Histone Deacetylase 1 and 3 in Huntington's Disease Mice. Front Mol Neurosci 2021; 14:616886. [PMID: 33679321 PMCID: PMC7925995 DOI: 10.3389/fnmol.2021.616886] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/27/2021] [Indexed: 01/15/2023] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease characterized by a late clinical onset of psychiatric, cognitive, and motor symptoms. Transcriptional dysregulation is an early and central disease mechanism which is accompanied by epigenetic alterations in HD. Previous studies demonstrated that targeting transcriptional changes by inhibition of histone deacetylases (HDACs), especially the class I HDACs, provides therapeutic effects. Yet, their exact mechanisms of action and the features of HD pathology, on which these inhibitors act remain to be elucidated. Here, using transcriptional profiling, we found that selective inhibition of HDAC1 and HDAC3 by RGFP109 alleviated transcriptional dysregulation of a number of genes, including the transcription factor genes Neurod2 and Nr4a2, and gene sets and programs, especially those that are associated to insulin-like growth factor pathway, in the striatum of R6/1 mice. RGFP109 treatment led to a modest improvement of the motor skill learning and coordination deficit on the RotaRod test, while it did not alter the locomotor and anxiety-like phenotypes in R6/1 animals. We also found, by volumetric MRI, a widespread brain atrophy in the R6/1 mice at the symptomatic disease stage, on which RGFP109 showed no significant effects. Collectively, our combined work suggests that specific HDAC1 and HDAC3 inhibition may offer benefits for alleviating the motor phenotypic deficits and transcriptional dysregulation in HD.
Collapse
Affiliation(s)
- Katharina Hecklau
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Mueller
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité – Universitätsmedizin Berlin, Berlin, Germany
- Charité Core Facility 7T Experimental MRIs, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Paul Koch
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité – Universitätsmedizin Berlin, Berlin, Germany
- Charité Core Facility 7T Experimental MRIs, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Mustafa Hussain Mehkary
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Busra Kilic
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité – Universitätsmedizin Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité – Universitätsmedizin Berlin, Berlin, Germany
- Charité Core Facility 7T Experimental MRIs, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ferah Yildirim
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
Formisano L, Laudati G, Guida N, Mascolo L, Serani A, Cuomo O, Cantile M, Boscia F, Molinaro P, Anzilotti S, Pizzorusso V, Di Renzo G, Pignataro G, Annunziato L. HDAC4 and HDAC5 form a complex with DREAM that epigenetically down-regulates NCX3 gene and its pharmacological inhibition reduces neuronal stroke damage. J Cereb Blood Flow Metab 2020; 40:2081-2097. [PMID: 31696766 PMCID: PMC7786841 DOI: 10.1177/0271678x19884742] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The histone deacetylases (HDACs)-dependent mechanisms regulating gene transcription of the Na+/Ca+ exchanger isoform 3 (ncx3) after stroke are still unknown. Overexpression or knocking-down of HDAC4/HDAC5 down-regulates or increases, respectively, NCX3 mRNA and protein. Likewise, MC1568 (class IIa HDACs inhibitor), but not MS-275 (class I HDACs inhibitor) increased NCX3 promoter activity, gene and protein expression. Furthermore, HDAC4 and HDAC5 physically interacted with the transcription factor downstream regulatory element antagonist modulator (DREAM). As MC1568, DREAM knocking-down prevented HDAC4 and HDAC5 recruitment to the ncx3 promoter. Importantly, DREAM, HDAC4, and HDAC5 recruitment to the ncx3 gene was increased in the temporoparietal cortex of rats subjected to transient middle cerebral artery occlusion (tMCAO), with a consequent histone-deacetylation of ncx3 promoter. Conversely, the tMCAO-induced NCX3 reduction was prevented by intracerebroventricular injection of siDREAM, siHDAC4, and siHDAC5. Notably, MC1568 prevented oxygen glucose deprivation plus reoxygenation and tMCAO-induced neuronal damage, whereas its neuroprotective effect was abolished by ncx3 knockdown. Collectively, we found that: (1) DREAM/HDAC4/HDAC5 complex epigenetically down-regulates ncx3 gene transcription after stroke, and (2) pharmacological inhibition of class IIa HDACs reduces stroke-induced neurodetrimental effects.
Collapse
Affiliation(s)
- Luigi Formisano
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Giusy Laudati
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | | | - Luigi Mascolo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Angelo Serani
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Maria Cantile
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Pasquale Molinaro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | | | - Vincenzo Pizzorusso
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Gianfranco Di Renzo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, "Federico II" University of Naples, Naples, Italy
| | | |
Collapse
|
6
|
Cores Á, Piquero M, Villacampa M, León R, Menéndez JC. NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases. Biomolecules 2020; 10:E904. [PMID: 32545924 PMCID: PMC7356958 DOI: 10.3390/biom10060904] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/03/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
NRF2 acts by controlling gene expression, being the master regulator of the Phase II antioxidant response, and also being key to the control of neuroinflammation. NRF2 activity is regulated at several levels, including protein degradation by the proteasome, transcription, and post-transcription. The purpose of this review is to offer a concise and critical overview of the main mechanisms of NRF2 regulation and their actual or potential use as targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Marta Piquero
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Mercedes Villacampa
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Rafael León
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - J. Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| |
Collapse
|
7
|
Romoli M, Mazzocchetti P, D'Alonzo R, Siliquini S, Rinaldi VE, Verrotti A, Calabresi P, Costa C. Valproic Acid and Epilepsy: From Molecular Mechanisms to Clinical Evidences. Curr Neuropharmacol 2020; 17:926-946. [PMID: 30592252 PMCID: PMC7052829 DOI: 10.2174/1570159x17666181227165722] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/03/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
After more than a century from its discovery, valproic acid (VPA) still represents one of the most efficient antiepi-leptic drugs (AEDs). Pre and post-synaptic effects of VPA depend on a very broad spectrum of actions, including the regu-lation of ionic currents and the facilitation of GABAergic over glutamatergic transmission. As a result, VPA indirectly mod-ulates neurotransmitter release and strengthens the threshold for seizure activity. However, even though participating to the anticonvulsant action, such mechanisms seem to have minor impact on epileptogenesis. Nonetheless, VPA has been reported to exert anti-epileptogenic effects. Epigenetic mechanisms, including histone deacetylases (HDACs), BDNF and GDNF modulation are pivotal to orientate neurons toward a neuroprotective status and promote dendritic spines organization. From such broad spectrum of actions comes constantly enlarging indications for VPA. It represents a drug of choice in child and adult with epilepsy, with either general or focal seizures, and is a consistent and safe IV option in generalized convulsive sta-tus epilepticus. Moreover, since VPA modulates DNA transcription through HDACs, recent evidences point to its use as an anti-nociceptive in migraine prophylaxis, and, even more interestingly, as a positive modulator of chemotherapy in cancer treatment. Furthermore, VPA-induced neuroprotection is under investigation for benefit in stroke and traumatic brain injury. Hence, VPA has still got its place in epilepsy, and yet deserves attention for its use far beyond neurological diseases. In this review, we aim to highlight, with a translational intent, the molecular basis and the clinical indications of VPA.
Collapse
Affiliation(s)
- Michele Romoli
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Petra Mazzocchetti
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Renato D'Alonzo
- Pediatric Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | | | - Victoria Elisa Rinaldi
- Pediatric Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| | - Alberto Verrotti
- Department of Pediatrics, University of L'Aquila - San Salvatore Hospital, L'Aquila, Italy
| | - Paolo Calabresi
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy.,IRCCS "Santa Lucia", Rome, Italy
| | - Cinzia Costa
- Neurology Clinic, University of Perugia - S. Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
8
|
Benavente-Fernandez I, Ramos-Rodriguez JJ, Infante-Garcia C, Jimenez-Gomez G, Lechuga-Sancho A, Lubian-Lopez S, Garcia-Alloza M. Altered plasma-type gelsolin and amyloid-β in neonates with hypoxic-ischaemic encephalopathy under therapeutic hypothermia. J Cereb Blood Flow Metab 2019; 39:1349-1354. [PMID: 29466895 PMCID: PMC6668521 DOI: 10.1177/0271678x18757419] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/06/2018] [Accepted: 01/12/2018] [Indexed: 01/08/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a severe neonatal complication responsible for ∼23% of all neonatal deaths. Also, 30-70% of these patients will suffer lifetime disabilities, including learning impairment, epilepsy or cerebral palsy. However, biomarkers for HIE screening, or monitoring disease progression are limited. Herein, we sought to evaluate the clinical usefulness of plasma-type gelsolin (pGSN) and amyloid-beta (Aβ) 40 and 42 as prognostic biomarkers for HIE. pGSN has been previously suggested as a feasible marker in other brain injuries and amyloid-beta 40 and 42 are classically assessed in neurodegenerative diseases. However, to our knowledge, they have not been previously assessed in HIE patients. We have analyzed plasma pGSN and Aβ 40 and 42 levels in 55 newborns (16 controls, 16 mild and 23 moderate-severe HIE) at birth, during 72 h of therapeutic hypothermia, a gold-standard treatment for HIE, and 24 h after hypothermia. Aβ levels were lower in HIE patients, and pGSN levels were progressively reduced in mild and moderate-severe HIE patients. The fact that pGSN reductions could predict the severity of HIE and significantly correlated with the time to undergo hypothermia supports the prognostic value of plasmatic pGSN. Further studies are warranted to investigate the role of pGSN in neonatal HIE.
Collapse
Affiliation(s)
| | - Juan J Ramos-Rodriguez
- Division of Physiology, School of Medicine, Instituto de Investigación Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
- Salus Infirmorum, Universidad de Cadiz, Cádiz, Spain
| | - Carmen Infante-Garcia
- Division of Physiology, School of Medicine, Instituto de Investigación Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| | | | - Alfonso Lechuga-Sancho
- Department of Pediatrics, Neonatology Unit, Puerta del Mar University Hospital, Cadiz, Spain
- Department of Mother and Child Health and Radiology, Universidad de Cadiz, Cadiz, Spain
| | - Simon Lubian-Lopez
- Department of Pediatrics, Neonatology Unit, Puerta del Mar University Hospital, Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Instituto de Investigación Biomedica de Cadiz (INIBICA), Universidad de Cadiz, Cadiz, Spain
| |
Collapse
|
9
|
Yusoff SI, Roman M, Lai FY, Eagle-Hemming B, Murphy GJ, Kumar T, Wozniak M. Systematic review and meta-analysis of experimental studies evaluating the organ protective effects of histone deacetylase inhibitors. Transl Res 2019; 205:1-16. [PMID: 30528323 PMCID: PMC6386580 DOI: 10.1016/j.trsl.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 01/07/2023]
Abstract
The clinical efficacy of organ protection interventions are limited by the redundancy of cellular activation mechanisms. Interventions that target epigenetic mechanisms overcome this by eliciting genome wide changes in transcription and signaling. We aimed to review preclinical studies evaluating the organ protection effects of histone deacetylase inhibitors (HDACi) with a view to informing the design of early phase clinical trials. A systematic literature search was performed. Methodological quality was assessed against prespecified criteria. The primary outcome was mortality, with secondary outcomes assessing mechanisms. Prespecified analyses evaluated the effects of likely moderators on heterogeneity. The analysis included 101 experimental studies in rodents (n = 92) and swine (n = 9), exposed to diverse injuries, including: ischemia (n = 72), infection (n = 7), and trauma (n = 22). There were a total of 448 comparisons due to the evaluation of multiple independent interventions within single studies. Sodium valproate (VPA) was the most commonly evaluated HDACi (50 studies, 203 comparisons). All of the studies were judged to have significant methodological limitations. HDACi reduced mortality in experimental models of organ injury (risk ratio = 0.52, 95% confidence interval 0.40-0.68, p < 0.001) without heterogeneity. HDACi administration resulted in myocardial, brain and kidney protection across diverse species and injuries that was attributable to increases in prosurvival cell signaling, and reductions in inflammation and programmed cell death. Heterogeneity in the analyses of secondary outcomes was explained by differences in species, type of injury, HDACi class (Class I better), drug (trichostatin better), and time of administration (at least 6 hours prior to injury better). These findings highlight a potential novel application for HDACi in clinical settings characterized by acute organ injury.
Collapse
Affiliation(s)
- Syabira I Yusoff
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK.
| | - Marius Roman
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Florence Y Lai
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Bryony Eagle-Hemming
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Gavin J Murphy
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Tracy Kumar
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| | - Marcin Wozniak
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Unit in Cardiovascular Medicine, University of Leicester, Clinical Sciences Wing, Glenfield Hospital, Leicester, UK
| |
Collapse
|
10
|
Interplay between NAD + and acetyl‑CoA metabolism in ischemia-induced mitochondrial pathophysiology. Biochim Biophys Acta Mol Basis Dis 2018; 1865:2060-2067. [PMID: 30261291 DOI: 10.1016/j.bbadis.2018.09.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 12/11/2022]
Abstract
Brain injury caused by ischemic insult due to significant reduction or interruption in cerebral blood flow leads to disruption of practically all cellular metabolic pathways. This triggers a complex stress response followed by overstimulation of downstream enzymatic pathways due to massive activation of post-translational modifications (PTM). Mitochondria are one of the most sensitive organelle to ischemic conditions. They become dysfunctional due to extensive fragmentation, inhibition of acetyl‑CoA production, and increased activity of NAD+ consuming enzymes. These pathologic conditions ultimately lead to inhibition of oxidative phosphorylation and mitochondrial ATP production. Both acetyl‑CoA and NAD+ are essential intermediates in cellular bioenergetics metabolism and also serve as substrates for post-translational modifications such as acetylation and ADP‑ribosylation. In this review we discuss ischemia/reperfusion-induced changes in NAD+ and acetyl‑CoA metabolism, how these affect relevant PTMs, and therapeutic approaches that restore the physiological levels of these metabolites leading to promising neuroprotection.
Collapse
|
11
|
Plasma Gelsolin: Indicator of Inflammation and Its Potential as a Diagnostic Tool and Therapeutic Target. Int J Mol Sci 2018; 19:ijms19092516. [PMID: 30149613 PMCID: PMC6164782 DOI: 10.3390/ijms19092516] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/14/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022] Open
Abstract
Gelsolin, an actin-depolymerizing protein expressed both in extracellular fluids and in the cytoplasm of a majority of human cells, has been recently implicated in a variety of both physiological and pathological processes. Its extracellular isoform, called plasma gelsolin (pGSN), is present in blood, cerebrospinal fluid, milk, urine, and other extracellular fluids. This isoform has been recognized as a potential biomarker of inflammatory-associated medical conditions, allowing for the prediction of illness severity, recovery, efficacy of treatment, and clinical outcome. A compelling number of animal studies also demonstrate a broad spectrum of beneficial effects mediated by gelsolin, suggesting therapeutic utility for extracellular recombinant gelsolin. In the review, we summarize the current data related to the potential of pGSN as an inflammatory predictor and therapeutic target, discuss gelsolin-mediated mechanisms of action, and highlight recent progress in the clinical use of pGSN.
Collapse
|
12
|
Ng GYQ, Yun-An L, Sobey CG, Dheen T, Fann DYW, Arumugam TV. Epigenetic regulation of inflammation in stroke. Ther Adv Neurol Disord 2018; 11:1756286418771815. [PMID: 29774056 PMCID: PMC5949939 DOI: 10.1177/1756286418771815] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 03/29/2018] [Indexed: 12/30/2022] Open
Abstract
Despite extensive research, treatments for clinical stroke are still limited only to the administration of tissue plasminogen activator and the recent introduction of mechanical thrombectomy, which can be used in only a limited proportion of patients due to time constraints. A plethora of inflammatory events occur during stroke, arising in part due to the body's immune response to brain injury. Neuroinflammation contributes significantly to neuronal cell death and the development of functional impairment and death in stroke patients. Therefore, elucidating the molecular and cellular mechanisms underlying inflammatory damage following stroke injury will be essential for the development of useful therapies. Research findings increasingly point to the likelihood that epigenetic mechanisms play a role in the pathophysiology of stroke. Epigenetics involves the differential regulation of gene expression, including those involved in brain inflammation and remodelling after stroke. Hence, it is conceivable that epigenetic mechanisms may contribute to differential interindividual vulnerability and injury responses to cerebral ischaemia. In this review, we summarize recent findings on the emerging role of epigenetics in the regulation of neuroinflammation in stroke. We also discuss potential epigenetic targets that may be assessed for the development of stroke therapies.
Collapse
Affiliation(s)
- Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore
| | - Lim Yun-An
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore
| | - Christopher G. Sobey
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia
| | - Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - David Yang-Wei Fann
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore
| | - Thiruma V. Arumugam
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Medical Drive, MD9, Singapore School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea Neurobiology/Ageing Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
13
|
Jhelum P, Karisetty BC, Kumar A, Chakravarty S. Implications of Epigenetic Mechanisms and their Targets in Cerebral Ischemia Models. Curr Neuropharmacol 2018; 15:815-830. [PMID: 27964703 PMCID: PMC5652028 DOI: 10.2174/1570159x14666161213143907] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/07/2016] [Accepted: 12/09/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Understanding the complexities associated with the ischemic condition and identifying therapeutic targets in ischemia is a continued challenge in stroke biology. Emerging evidence reveals the potential involvement of epigenetic mechanisms in the incident and outcome of stroke, suggesting novel therapeutic options of targeting different molecules related to epigenetic regulation. OBJECTIVE This review summarizes our current understanding of ischemic pathophysiology, describes various in vivo and in vitro models of ischemia, and examines epigenetic modifications associated with the ischemic condition. METHOD We focus on microRNAs, DNA methylation, and histone modifying enzymes, and present how epigenetic studies are revealing novel drug target candidates in stroke. CONCLUSION Finally, we discuss emerging approaches for the prevention and treatment of stroke and post-stroke effects using pharmacological interventions with a wide therapeutic window.
Collapse
Affiliation(s)
- Priya Jhelum
- Chemical Biology, CSIR, Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Bhanu C Karisetty
- Chemical Biology, CSIR, Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Arvind Kumar
- CSIR, Centre for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad 500007, India
| | - Sumana Chakravarty
- Chemical Biology, CSIR-Indian Institute of Chemical Technology (IICT), Tarnaka, Hyderabad-500007, India
| |
Collapse
|
14
|
Klimova N, Long A, Kristian T. Significance of Mitochondrial Protein Post-translational Modifications in Pathophysiology of Brain Injury. Transl Stroke Res 2017; 9:223-237. [DOI: 10.1007/s12975-017-0569-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 01/13/2023]
|
15
|
The emerging field of epigenetics in neurodegeneration and neuroprotection. Nat Rev Neurosci 2017; 18:347-361. [PMID: 28515491 DOI: 10.1038/nrn.2017.46] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic mechanisms - including DNA methylation, histone post-translational modifications and changes in nucleosome positioning - regulate gene expression, cellular differentiation and development in almost all tissues, including the brain. In adulthood, changes in the epigenome are crucial for higher cognitive functions such as learning and memory. Striking new evidence implicates the dysregulation of epigenetic mechanisms in neurodegenerative disorders and diseases. Although these disorders differ in their underlying causes and pathophysiologies, many involve the dysregulation of restrictive element 1-silencing transcription factor (REST), which acts via epigenetic mechanisms to regulate gene expression. Although not somatically heritable, epigenetic modifications in neurons are dynamic and reversible, which makes them good targets for therapeutic intervention.
Collapse
|
16
|
Jaworska J, Ziemka-Nalecz M, Sypecka J, Zalewska T. The potential neuroprotective role of a histone deacetylase inhibitor, sodium butyrate, after neonatal hypoxia-ischemia. J Neuroinflammation 2017; 14:34. [PMID: 28187734 PMCID: PMC5303300 DOI: 10.1186/s12974-017-0807-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 02/01/2017] [Indexed: 02/07/2023] Open
Abstract
Background Histone deacetylase inhibitor (HDACi), sodium butyrate (SB), has been shown to be neuroprotective in adult brain injury models. Potential explanation for the inhibitor action involves among others reduced inflammation. We therefore anticipated that SB will provide a suitable option for brain injury in immature animals. The aim of our study was to test the hypothesis that one of the mechanisms of protection afforded by SB after neonatal hypoxia-ischemia is associated with anti-inflammatory action. We examined the effect of SB on the production of inflammatory factors including analysis of the microglial and astrocytic cell response. We also examined the effect of SB on molecular mediators that are crucial for inducing cerebral damage after ischemia (transcription factors, HSP70, as well as pro- and anti-apoptotic proteins). Methods Seven-day-old rat pups were subjected to unilateral carotid artery ligation followed by 60 min of hypoxia (7.6% O2). SB (300 mg/kg) was administered in a 5-day regime with the first injection given immediately after hypoxic exposure. The damage of the ipsilateral hemisphere was evaluated by hematoxylin-eosin staining (HE) 6 days after the insult. Samples were collected at 24 and 48 h and 6 days. Effects of SB on hypoxia-ischemia (HI)-induced inflammation (cytokines and chemokine) were assessed by Luminex assay and immunohistochemistry. Expression of molecular mediators (NFκB, p53, HSP70, COX-2, pro- and anti-apoptotic factors Bax, Bcl-2, caspase-3) were assayed by Western blot analysis. Results SB treatment-reduced brain damage, as assessed by HE staining, suppressed the production of inflammatory markers—IL-1β, chemokine CXCL10, and blocked ischemia-elicited upregulation of COX-2 in the damaged ipsilateral hemisphere. Furthermore, administration of SB promoted the conversion of microglia phenotype from inflammatory M1 to anti-inflammatory M2. None of the investigated molecular mediators that are known to be affected by HDACis in adults were modified after SB administration. Conclusions Administration of SB is neuroprotective in neonatal hypoxia-ischemia injury. This neuroprotective activity prevented the delayed rise in chemokine CXCL10, IL-1β, and COX-2 in the ipsilateral hemisphere. SB appears to exert a beneficial effect via suppression of HI-induced cerebral inflammation.
Collapse
Affiliation(s)
- Joanna Jaworska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 A. Pawinskiego Street, 02-106, Warsaw, Poland.
| |
Collapse
|
17
|
Qiu X, Xiao X, Li N, Li Y. Histone deacetylases inhibitors (HDACis) as novel therapeutic application in various clinical diseases. Prog Neuropsychopharmacol Biol Psychiatry 2017; 72:60-72. [PMID: 27614213 DOI: 10.1016/j.pnpbp.2016.09.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/31/2016] [Accepted: 09/05/2016] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that histone hypoacetylation which is partly mediated by histone deacetylase (HDAC), plays a causative role in the etiology of various clinical disorders such as cancer and central nervous diseases. HDAC inhibitors (HDACis) are natural or synthetic small molecules that can inhibit the activities of HDACs and restore or increase the level of histone acetylation, thus may represent the potential approach to treating a number of clinical disorders. This manuscript reviewed the progress of the most recent experimental application of HDACis as novel potential drugs or agents in a large number of clinical disorders including various brain disorders including neurodegenerative and neurodevelopmental cognitive disorders and psychiatric diseases like depression, anxiety, fear and schizophrenia, and cancer, endometriosis and cell reprogramming in somatic cell nuclear transfer in human and animal models of disease, and concluded that HDACis as potential novel therapeutic agents could be used alone or in adjunct to other pharmacological agents in various clinical diseases.
Collapse
Affiliation(s)
- Xiaoyan Qiu
- School of Animal Science & Technology, Southwest University, Chong Qing 400715, PR China
| | - Xiong Xiao
- School of Animal Science & Technology, Southwest University, Chong Qing 400715, PR China
| | - Nan Li
- School of Animal Science & Technology, Southwest University, Chong Qing 400715, PR China
| | - Yuemin Li
- School of Animal Science & Technology, Southwest University, Chong Qing 400715, PR China.
| |
Collapse
|
18
|
Yang X, Wu Q, Zhang L, Feng L. Inhibition of Histone Deacetylase 3 (HDAC3) Mediates Ischemic Preconditioning and Protects Cortical Neurons against Ischemia in Rats. Front Mol Neurosci 2016; 9:131. [PMID: 27965534 PMCID: PMC5124709 DOI: 10.3389/fnmol.2016.00131] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/11/2016] [Indexed: 01/22/2023] Open
Abstract
Brain ischemic preconditioning (PC) provides vital insights into the endogenous protection against stroke. Genomic and epigenetic responses to PC condition the brain into a state of ischemic tolerance. Notably, PC induces the elevation of histone acetylation, consistent with evidence that histone deacetylase (HDAC) inhibitors protect the brain from ischemic injury. However, less is known about the specific roles of HDACs in this process. HDAC3 has been implicated in several neurodegenerative conditions. Deletion of HDAC3 confers protection against neurotoxicity and neuronal injury. Here, we hypothesized that inhibition of HDAC3 may contribute to the neuronal survival elicited by PC. To address this notion, PC and transient middle cerebral artery occlusion (MCAO) were conducted in Sprague-Dawley rats. Additionally, primary cultured cortical neurons were used to identify the modulators and effectors of HDAC3 involved in PC. We found that nuclear localization of HDAC3 was significantly reduced following PC in vivo and in vitro. Treatment with the HDAC3-specific inhibitor, RGFP966, mimicked the neuroprotective effects of PC 24 h and 7 days after MCAO, causing a reduced infarct volume and less Fluoro-Jade C staining. Improved functional outcomes were observed in the neurological score and rotarod test. We further showed that attenuated recruitment of HDAC3 to promoter regions following PC potentiates transcriptional initiation of genes including Hspa1a, Bcl2l1, and Prdx2, which may underlie the mechanism of protection. In addition, PC-activated calpains were implicated in the cleavage of HDAC3. Pretreatment with calpeptin blockaded the attenuated nuclear distribution of HDAC3 and the protective effect of PC in vivo. Collectively, these results demonstrate that the inhibition of HDAC3 preconditions the brain against ischemic insults, indicating a new approach to evoke endogenous protection against stroke.
Collapse
Affiliation(s)
- Xiaoyu Yang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai, China
| | - Qimei Wu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai, China
| | - Lei Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai, China
| | - Linyin Feng
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai, China
| |
Collapse
|
19
|
Ganai SA, Ramadoss M, Mahadevan V. Histone Deacetylase (HDAC) Inhibitors - emerging roles in neuronal memory, learning, synaptic plasticity and neural regeneration. Curr Neuropharmacol 2016; 14:55-71. [PMID: 26487502 PMCID: PMC4787286 DOI: 10.2174/1570159x13666151021111609] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 08/23/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022] Open
Abstract
Epigenetic regulation of neuronal signalling through histone acetylation dictates transcription programs that govern neuronal memory, plasticity and learning paradigms. Histone Acetyl Transferases (HATs) and Histone Deacetylases (HDACs) are antagonistic enzymes that regulate gene expression through acetylation and deacetylation of histone proteins around which DNA is wrapped inside a eukaryotic cell nucleus. The epigenetic control of HDACs and the cellular imbalance between HATs and HDACs dictate disease states and have been implicated in muscular dystrophy, loss of memory, neurodegeneration and autistic disorders. Altering gene expression profiles through inhibition of HDACs is now emerging as a powerful technique in therapy. This review presents evolving applications of HDAC inhibitors as potential drugs in neurological research and therapy. Mechanisms that govern their
expression profiles in neuronal signalling, plasticity and learning will be covered. Promising and exciting possibilities of HDAC inhibitors in memory formation, fear conditioning, ischemic stroke and neural regeneration have been detailed.
Collapse
Affiliation(s)
| | | | - Vijayalakshmi Mahadevan
- School of Chemical & Biotechnology SASTRA University Tirumalaisamudram, Thanjavur - 613 401 India.
| |
Collapse
|
20
|
Kaminska B, Mota M, Pizzi M. Signal transduction and epigenetic mechanisms in the control of microglia activation during neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2016; 1862:339-51. [DOI: 10.1016/j.bbadis.2015.10.026] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/12/2015] [Accepted: 10/28/2015] [Indexed: 12/21/2022]
|
21
|
Glutamate and ATP at the Interface Between Signaling and Metabolism in Astroglia: Examples from Pathology. Neurochem Res 2016; 42:19-34. [PMID: 26915104 DOI: 10.1007/s11064-016-1848-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 01/21/2016] [Accepted: 01/22/2016] [Indexed: 12/17/2022]
Abstract
Glutamate is the main excitatory transmitter in the brain, while ATP represents the most important energy currency in any living cell. Yet, these chemicals play an important role in both processes, enabling them with dual-acting functions in metabolic and intercellular signaling pathways. Glutamate can fuel ATP production, while ATP can act as a transmitter in intercellular signaling. We discuss the interface between glutamate and ATP in signaling and metabolism of astrocytes. Not only do glutamate and ATP cross each other's paths in physiology of the brain, but they also do so in its pathology. We present the fabric of this process in (patho)physiology through the discussion of synthesis and metabolism of ATP and glutamate in astrocytes as well as by providing a general description of astroglial receptors for these molecules along with the downstream signaling pathways that may be activated. It is astroglial receptors for these dual-acting molecules that could hold a key for medical intervention in pathological conditions. We focus on two examples disclosing the role of activation of astroglial ATP and glutamate receptors in pathology of two kinds of brain tissue, gray matter and white matter, respectively. Interventions at the interface of metabolism and signaling show promise for translational medicine.
Collapse
|
22
|
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: 4.9] [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
|
23
|
Kirmes I, Szczurek A, Prakash K, Charapitsa I, Heiser C, Musheev M, Schock F, Fornalczyk K, Ma D, Birk U, Cremer C, Reid G. A transient ischemic environment induces reversible compaction of chromatin. Genome Biol 2015; 16:246. [PMID: 26541514 PMCID: PMC4635527 DOI: 10.1186/s13059-015-0802-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/09/2015] [Indexed: 11/10/2022] Open
Abstract
Background Cells detect and adapt to hypoxic and nutritional stress through immediate transcriptional, translational and metabolic responses. The environmental effects of ischemia on chromatin nanostructure were investigated using single molecule localization microscopy of DNA binding dyes and of acetylated histones, by the sensitivity of chromatin to digestion with DNAseI, and by fluorescence recovery after photobleaching (FRAP) of core and linker histones. Results Short-term oxygen and nutrient deprivation of the cardiomyocyte cell line HL-1 induces a previously undescribed chromatin architecture, consisting of large, chromatin-sparse voids interspersed between DNA-dense hollow helicoid structures 40–700 nm in dimension. The chromatin compaction is reversible, and upon restitution of normoxia and nutrients, chromatin transiently adopts a more open structure than in untreated cells. The compacted state of chromatin reduces transcription, while the open chromatin structure induced upon recovery provokes a transitory increase in transcription. Digestion of chromatin with DNAseI confirms that oxygen and nutrient deprivation induces compaction of chromatin. Chromatin compaction is associated with depletion of ATP and redistribution of the polyamine pool into the nucleus. FRAP demonstrates that core histones are not displaced from compacted chromatin; however, the mobility of linker histone H1 is considerably reduced, to an extent that far exceeds the difference in histone H1 mobility between heterochromatin and euchromatin. Conclusions These studies exemplify the dynamic capacity of chromatin architecture to physically respond to environmental conditions, directly link cellular energy status to chromatin compaction and provide insight into the effect ischemia has on the nuclear architecture of cells. Electronic supplementary material The online version of this article (doi:10.1186/s13059-015-0802-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ina Kirmes
- Institute for Molecular Biology, 55128, Mainz, Germany
| | | | - Kirti Prakash
- Institute for Molecular Biology, 55128, Mainz, Germany.,Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120, Heidelberg, Germany
| | | | | | | | | | - Karolina Fornalczyk
- Institute for Molecular Biology, 55128, Mainz, Germany.,Department of Molecular Biophysics, University of Łódź, Łódź, Poland
| | - Dongyu Ma
- Institute for Molecular Biology, 55128, Mainz, Germany.,Centre for Biomedicine and Medical Technology Mannheim (CBTM), University of Heidelberg, 68167, Mannheim, Germany
| | - Udo Birk
- Institute for Molecular Biology, 55128, Mainz, Germany
| | - Christoph Cremer
- Institute for Molecular Biology, 55128, Mainz, Germany. .,Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120, Heidelberg, Germany.
| | - George Reid
- Institute for Molecular Biology, 55128, Mainz, Germany.
| |
Collapse
|
24
|
Picascia A, Grimaldi V, Iannone C, Soricelli A, Napoli C. Innate and adaptive immune response in stroke: Focus on epigenetic regulation. J Neuroimmunol 2015; 289:111-20. [PMID: 26616880 DOI: 10.1016/j.jneuroim.2015.10.013] [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: 06/24/2015] [Revised: 10/17/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
Abstract
Inflammation and immune response play a pivotal role in the pathophysiology of ischemic stroke giving their contribution to tissue damage and repair. Emerging evidence supports the involvement of epigenetic mechanisms such as methylation, histone modification and miRNAs in the pathogenesis of stroke. Interestingly, epigenetics can influence the molecular events involved in ischemic injury by controlling the switch from pro- to anti-inflammatory response, however, this is still a field to be fully explored. The knowledge of epigenetic processes could to allow for the discovery of more sensitive and specific biomarkers for risk, onset, and progression of disease as well as further novel tools to be used in both primary prevention and therapy of stroke. Indeed, studies performed in vitro and in small animal models seem to suggest a neuroprotective role of HDAC inhibitors (e.g. valproic acid) and antagomir (e.g. anti-miR-181a) in ischemic condition by modulation of both immune and inflammatory pathways. Thus, the clinical implications of altered epigenetic mechanisms for the prevention of stroke are very promising but clinical prospective studies and translational approaches are still warranted.
Collapse
Affiliation(s)
- Antonietta Picascia
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy
| | - Vincenzo Grimaldi
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy.
| | - Carmela Iannone
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy
| | - Andrea Soricelli
- IRCCS Research Institute SDN, Naples, Italy; Department of Studies of Institutions and Territorial Systems, University of Naples Parthenope, Naples, Italy
| | - Claudio Napoli
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Department of Internal Medicine and Specialistics, Azienda Ospedaliera Universitaria (AOU), Second University of Naples, Naples, Italy; IRCCS Research Institute SDN, Naples, Italy
| |
Collapse
|
25
|
Schweizer S, Harms C, Lerch H, Flynn J, Hecht J, Yildirim F, Meisel A, Märschenz S. Inhibition of histone methyltransferases SUV39H1 and G9a leads to neuroprotection in an in vitro model of cerebral ischemia. J Cereb Blood Flow Metab 2015; 35:1640-7. [PMID: 25966950 PMCID: PMC4640311 DOI: 10.1038/jcbfm.2015.99] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 01/04/2023]
Abstract
Cerebral ischemia induces a complex transcriptional response with global changes in gene expression. It is essentially regulated by transcription factors as well as epigenetic players. While it is well known that the inhibition of transcriptionally repressive histone deacetylases leads to neuroprotection, the role of histone methyltransferases in the postischemic transcriptional response remains elusive. We investigated the effects of inhibition of the repressive H3K9 histone methyltransferases SUV39H1 and G9a on neuronal survival, H3K9 promoter signatures and gene expression. Their inhibition either with the specific blocker chaetocin or by use of RNA interference promoted neuronal survival in oxygen glucose deprivation (OGD). Brain-derived neurotrophic factor (BDNF) was upregulated and BDNF promoter regions showed an increase in histone marks characteristic for active transcription. The BDNF blockade with K252a abrogated the protective effect of chaetocin treatment. In conclusion, inhibition of histone methyltransferases SUV39H1 and G9a confers neuroprotection in a model of hypoxic metabolic stress, which is at least in part mediated by BDNF.
Collapse
Affiliation(s)
- Sophie Schweizer
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Heike Lerch
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jennifer Flynn
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jochen Hecht
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ferah Yildirim
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Meisel
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefanie Märschenz
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
26
|
Metabolism and epigenetics in the nervous system: Creating cellular fitness and resistance to neuronal death in neurological conditions via modulation of oxygen-, iron-, and 2-oxoglutarate-dependent dioxygenases. Brain Res 2015; 1628:273-287. [PMID: 26232572 DOI: 10.1016/j.brainres.2015.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 07/11/2015] [Accepted: 07/21/2015] [Indexed: 12/30/2022]
Abstract
Modern definitions of epigenetics incorporate models for transient but biologically important changes in gene expression that are unrelated to DNA code but responsive to environmental changes such as injury-induced stress. In this scheme, changes in oxygen levels (hypoxia) and/or metabolic co-factors (iron deficiency or diminished 2-oxoglutarate levels) are transduced into broad genetic programs that return the cell and the organism to a homeostatic set point. Over the past two decades, exciting studies have identified a superfamily of iron-, oxygen-, and 2-oxoglutarate-dependent dioxygenases that sit in the nucleus as modulators of transcription factor stability, co-activator function, histone demethylases, and DNA demethylases. These studies have provided a concrete molecular scheme for how changes in metabolism observed in a host of neurological conditions, including stroke, traumatic brain injury, and Alzheimer's disease, could be transduced into adaptive gene expression to protect the nervous system. We will discuss these enzymes in this short review, focusing primarily on the ten eleven translocation (TET) DNA demethylases, the jumonji (JmJc) histone demethylases, and the oxygen-sensing prolyl hydroxylase domain enzymes (HIF PHDs). This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
|
27
|
Aune SE, Herr DJ, Kutz CJ, Menick DR. Histone Deacetylases Exert Class-Specific Roles in Conditioning the Brain and Heart Against Acute Ischemic Injury. Front Neurol 2015; 6:145. [PMID: 26175715 PMCID: PMC4485035 DOI: 10.3389/fneur.2015.00145] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/15/2015] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion (IR) injury comprises a significant portion of morbidity and mortality from heart and brain diseases worldwide. This enduring clinical problem has inspired myriad reports in the scientific literature of experimental interventions seeking to elucidate the pathology of IR injury. Elective cardiac surgery presents perhaps the most viable scenario for protecting the heart and brain from IR injury due to the opportunity to condition the organs prior to insult. The physiological parameters for the preconditioning of vital organs prior to insult through mechanical and pharmacological maneuvers have been heavily examined. These investigations have revealed new insights into how preconditioning alters cellular responses to IR injury. However, the promise of preconditioning remains unfulfilled at the clinical level, and research seeking to implicate cell signals essential to this protection continues. Recent discoveries in molecular biology have revealed that gene expression can be controlled through posttranslational modifications, without altering the chemical structure of the genetic code. In this scenario, gene expression is repressed by enzymes that cause chromatin compaction through catalytic removal of acetyl moieties from lysine residues on histones. These enzymes, called histone deacetylases (HDACs), can be inhibited pharmacologically, leading to the de-repression of protective genes. The discovery that HDACs can also alter the function of non-histone proteins through posttranslational deacetylation has expanded the potential impact of HDAC inhibitors for the treatment of human disease. HDAC inhibitors have been applied in a very small number of experimental models of IR. However, the scientific literature contains an increasing number of reports demonstrating that HDACs converge on preconditioning signals in the cell. This review will describe the influence of HDACs on major preconditioning signaling pathways in the heart and brain.
Collapse
Affiliation(s)
- Sverre E Aune
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Daniel J Herr
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Craig J Kutz
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Donald R Menick
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
28
|
Uhlemann R, Gertz K, Boehmerle W, Schwarz T, Nolte C, Freyer D, Kettenmann H, Endres M, Kronenberg G. Actin dynamics shape microglia effector functions. Brain Struct Funct 2015; 221:2717-34. [PMID: 25989853 DOI: 10.1007/s00429-015-1067-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/15/2015] [Indexed: 11/28/2022]
Abstract
Impaired actin filament dynamics have been associated with cellular senescence. Microglia, the resident immune cells of the brain, are emerging as a central pathophysiological player in neurodegeneration. Microglia activation, which ranges on a continuum between classical and alternative, may be of critical importance to brain disease. Using genetic and pharmacological manipulations, we studied the effects of alterations in actin dynamics on microglia effector functions. Disruption of actin dynamics did not affect transcription of genes involved in the LPS-triggered classical inflammatory response. By contrast, in consequence of impaired nuclear translocation of phospho-STAT6, genes involved in IL-4 induced alternative activation were strongly downregulated. Functionally, impaired actin dynamics resulted in reduced NO secretion and reduced release of TNFalpha and IL-6 from LPS-stimulated microglia and of IGF-1 from IL-4 stimulated microglia. However, pathological stabilization of the actin cytoskeleton increased LPS-induced release of IL-1beta and IL-18, which belong to an unconventional secretory pathway. Reduced NO release was associated with decreased cytoplasmic iNOS protein expression and decreased intracellular arginine uptake. Furthermore, disruption of actin dynamics resulted in reduced microglia migration, proliferation and phagocytosis. Finally, baseline and ATP-induced [Ca(2+)]int levels were significantly increased in microglia lacking gelsolin, a key actin-severing protein. Together, the dynamic state of the actin cytoskeleton profoundly and distinctly affects microglia behaviours. Disruption of actin dynamics attenuates M2 polarization by inhibiting transcription of alternative activation genes. In classical activation, the role of actin remodelling is complex, does not relate to gene transcription and shows a major divergence between cytokines following conventional and unconventional secretion.
Collapse
Affiliation(s)
- Ria Uhlemann
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Karen Gertz
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.,Klinik und Poliklinik für Neurologie and Center for Stroke Research Berlin (CSB), Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Wolfgang Boehmerle
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Tobias Schwarz
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christiane Nolte
- Cellular Neuroscience, Max-Delbruck-Center for Molecular Medicine, 13092, Berlin-Buch, Germany
| | - Dorette Freyer
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neuroscience, Max-Delbruck-Center for Molecular Medicine, 13092, Berlin-Buch, Germany
| | - Matthias Endres
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Klinik und Poliklinik für Neurologie and Center for Stroke Research Berlin (CSB), Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Excellence Cluster NeuroCure, 10117, Berlin, Germany. .,German Center for Cardiovascular Research (DZHK), 13347, Berlin, Germany.
| | - Golo Kronenberg
- Klinik und Hochschulambulanz für Neurologie, Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Klinik und Poliklinik für Neurologie and Center for Stroke Research Berlin (CSB), Charité Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany. .,Klinik und Poliklinik für Psychiatrie und Psychotherapie, Charité Universitätsmedizin Berlin, 10117, Berlin, Germany.
| |
Collapse
|
29
|
Interplay between histone acetylation/deacetylation and poly(ADP-ribosyl)ation in the development of ischemic tolerance in vitro. Neuropharmacology 2015; 92:125-34. [DOI: 10.1016/j.neuropharm.2015.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/09/2015] [Accepted: 01/10/2015] [Indexed: 02/04/2023]
|
30
|
Edalatmanesh MA, Hosseini M, Ghasemi S, Golestani S, Sadeghnia HR, Mousavi SM, Vafaee F. Valproic acid-mediated inhibition of trimethyltin-induced deficits in memory and learning in the rat does not directly depend on its anti-oxidant properties. Ir J Med Sci 2015; 185:75-84. [DOI: 10.1007/s11845-014-1224-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 11/01/2014] [Indexed: 12/26/2022]
|
31
|
Jeong SG, Cho GW. Trichostatin A modulates intracellular reactive oxygen species through SOD2 and FOXO1 in human bone marrow-mesenchymal stem cells. Cell Biochem Funct 2014; 33:37-43. [PMID: 25515622 DOI: 10.1002/cbf.3084] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/18/2014] [Accepted: 11/07/2014] [Indexed: 12/16/2022]
Abstract
Engraft cells are often exposed to oxidative stress and inflammation; therefore, any factor that can provide the stem cells resistance to these stresses may yield better efficacy in stem cell therapy. Studies indicate that histone deacetylase (HDACs) inhibitors alleviate damage induced by oxidative stress. In this study, we investigated whether regulation of reactive oxygen species (ROS) occurs through the HDAC inhibitor trichostatin A (TSA) in human bone marrow-mesenchymal stem cells (hBM-MSCs). Intracellular ROS levels increased following exposure to hydrogen peroxide (H2 O2 ), and were suppressed by TSA treatment. Levels of the antioxidant enzyme superoxide dismutase 2 (SOD2) increased following treatment with 200 nM TSA and to a lesser level at 1-5 μM TSA. Cell protective effects against oxidative stress were significantly increased in TSA-MSCs after treatment with low doses of TSA (50-500 nM) and decreased with high doses of TSA (5-10 μM). Consistent results were obtained with immunoblot analysis for caspase3. Investigation of Forkhead box O1 (FOXO1), superoxide dismutase 2 (SOD2), and p53 levels to determine intracellular signaling by TSA in oxidative stress-induced MSCs demonstrated that expression of phosphorylated-FOXO1 and phosphorylated-SOD2 decreased in H2 O2 -treated MSCs while levels of p53 increased. These effects were reversed by the treatment of 200 nM TSA. These results suggest that the main function of ROS modulation by TSA is activated through SOD2 and FOXO1. Thus, optimal treatment with TSA may protect hBM-MSCs against oxidative stress.
Collapse
Affiliation(s)
- Sin-Gu Jeong
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Korea; Department of Life Science, BK21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Korea
| | | |
Collapse
|
32
|
Gao Q, Tang J, Chen J, Jiang L, Zhu X, Xu Z. Epigenetic code and potential epigenetic-based therapies against chronic diseases in developmental origins. Drug Discov Today 2014; 19:1744-1750. [PMID: 24880107 DOI: 10.1016/j.drudis.2014.05.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/05/2014] [Accepted: 05/07/2014] [Indexed: 12/14/2022]
Abstract
Accumulated findings have demonstrated that the epigenetic code provides a potential link between prenatal stress and changes in gene expression that could be involved in the developmental programming of various chronic diseases in later life. Meanwhile, based on the fact that epigenetic modifications are reversible and can be manipulated, this provides a unique chance to develop multiple novel epigenetic-based therapeutic strategies against many chronic diseases in early developmental periods. This article will give a short review of recent findings of prenatal insult-induced epigenetic changes in developmental origins of several chronic diseases, and will attempt to provide an overview of the current epigenetic-based strategies applied in the early prevention, diagnosis and possible therapies for human chronic diseases.
Collapse
Affiliation(s)
- Qinqin Gao
- Institute for Fetology, The First Hospital of Soochow University, Suzhou 215006, China
| | - Jiaqi Tang
- Institute for Fetology, The First Hospital of Soochow University, Suzhou 215006, China
| | - Jie Chen
- Institute for Fetology, The First Hospital of Soochow University, Suzhou 215006, China
| | - Lin Jiang
- Institute for Fetology, The First Hospital of Soochow University, Suzhou 215006, China
| | - Xiaolin Zhu
- Institute for Fetology, The First Hospital of Soochow University, Suzhou 215006, China
| | - Zhice Xu
- Institute for Fetology, The First Hospital of Soochow University, Suzhou 215006, China; Center for Prenatal Biology, Loma Linda University, CA 92350, USA.
| |
Collapse
|
33
|
Ma XH, Gao Q, Jia Z, Zhang ZW. Neuroprotective capabilities of TSA against cerebral ischemia/reperfusion injury via PI3K/Akt signaling pathway in rats. Int J Neurosci 2014; 125:140-6. [DOI: 10.3109/00207454.2014.912217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Zhan Y, Yang YT, You HM, Cao D, Liu CY, Zhou CJ, Wang ZY, Bai SJ, Mu J, Wu B, Zhan QL, Xie P. Plasma-based proteomics reveals lipid metabolic and immunoregulatory dysregulation in post-stroke depression. Eur Psychiatry 2014; 29:307-15. [PMID: 24853294 DOI: 10.1016/j.eurpsy.2014.03.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 03/23/2014] [Accepted: 03/26/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Post-stroke depression (PSD) is the most common psychiatric complication facing stroke survivors and has been associated with increased distress, physical disability, poor rehabilitation, and suicidal ideation. However, the pathophysiological mechanisms underlying PSD remain unknown, and no objective laboratory-based test is available to aid PSD diagnosis or monitor progression. METHODS Here, an isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomic approach was performed to identify differentially expressed proteins in plasma samples obtained from PSD, stroke, and healthy control subjects. RESULTS The significantly differentiated proteins were primarily involved in lipid metabolism and immunoregulation. Six proteins associated with these processes--apolipoprotein A-IV (ApoA-IV), apolipoprotein C-II (ApoC-II), C-reactive protein (CRP), gelsolin, haptoglobin, and leucine-rich alpha-2-glycoprotein (LRG)--were selected for Western blotting validation. ApoA-IV expression was significantly upregulated in PSD as compared to stroke subjects. ApoC-II, LRG, and CRP expression were significantly downregulated in both PSD and HC subjects relative to stroke subjects. Gelsolin and haptoglobin expression were significantly dysregulated across all three groups with the following expression profiles: gelsolin, healthy control>PSD>stroke subjects; haptoglobin, stroke>PSD>healthy control. CONCLUSIONS Early perturbation of lipid metabolism and immunoregulation may be involved in the pathophysiology of PSD. The combination of increased gelsolin levels accompanied by decreased haptoglobin levels shows promise as a plasma-based diagnostic biomarker panel for detecting increased PSD risk in post-stroke patients.
Collapse
Affiliation(s)
- Y Zhan
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Y-T Yang
- Department of Neurology, The First Affiliated Hospital at Chongqing Medical University, 1, Yixue Road, Yuzhong District, Chongqing, 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - H-M You
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - D Cao
- Department of Neurology, The First Affiliated Hospital at Chongqing Medical University, 1, Yixue Road, Yuzhong District, Chongqing, 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - C-Y Liu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - C-J Zhou
- Department of Neurology, The First Affiliated Hospital at Chongqing Medical University, 1, Yixue Road, Yuzhong District, Chongqing, 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Z-Y Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - S-J Bai
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - J Mu
- Department of Neurology, The First Affiliated Hospital at Chongqing Medical University, 1, Yixue Road, Yuzhong District, Chongqing, 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - B Wu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Q-L Zhan
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - P Xie
- Department of Neurology, The First Affiliated Hospital at Chongqing Medical University, 1, Yixue Road, Yuzhong District, Chongqing, 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
35
|
Muñiz J, Romero J, Holubiec M, Barreto G, González J, Saint-Martin M, Blanco E, Carlos Cavicchia J, Castilla R, Capani F. Neuroprotective effects of hypothermia on synaptic actin cytoskeletal changes induced by perinatal asphyxia. Brain Res 2014; 1563:81-90. [DOI: 10.1016/j.brainres.2014.03.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 01/28/2014] [Accepted: 03/17/2014] [Indexed: 10/25/2022]
|
36
|
Yildirim F, Ji S, Kronenberg G, Barco A, Olivares R, Benito E, Dirnagl U, Gertz K, Endres M, Harms C, Meisel A. Histone acetylation and CREB binding protein are required for neuronal resistance against ischemic injury. PLoS One 2014; 9:e95465. [PMID: 24748101 PMCID: PMC3991684 DOI: 10.1371/journal.pone.0095465] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 03/26/2014] [Indexed: 11/19/2022] Open
Abstract
Epigenetic transcriptional regulation by histone acetylation depends on the balance between histone acetyltransferase (HAT) and deacetylase activities (HDAC). Inhibition of HDAC activity provides neuroprotection, indicating that the outcome of cerebral ischemia depends crucially on the acetylation status of histones. In the present study, we characterized the changes in histone acetylation levels in ischemia models of focal cerebral ischemia and identified cAMP-response element binding protein (CREB)–binding protein (CBP) as a crucial factor in the susceptibility of neurons to ischemic stress. Both neuron-specific RNA interference and neurons derived from CBP heterozygous knockout mice showed increased damage after oxygen-glucose deprivation (OGD) in vitro. Furthermore, we demonstrated that ischemic preconditioning by a short (5 min) subthreshold occlusion of the middle cerebral artery (MCA), followed 24 h afterwards by a 30 min occlusion of the MCA, increased histone acetylation levels in vivo. Ischemic preconditioning enhanced CBP recruitment and histone acetylation at the promoter of the neuroprotective gene gelsolin leading to increased gelsolin expression in neurons. Inhibition of CBP's HAT activity attenuated neuronal ischemic preconditioning. Taken together, our findings suggest that the levels of CBP and histone acetylation determine stroke outcome and are crucially associated with the induction of an ischemia-resistant state in neurons.
Collapse
Affiliation(s)
- Ferah Yildirim
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Shengbo Ji
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Golo Kronenberg
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Klinik und Poliklinik für Psychiatrie, Campus Mitte, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Angel Barco
- Instituto de Neurociencias de Alicante (Universidad Miguel Hernandez-Consejo Superior de Investigaciones Cientificas), Campus de Sant Joan, Sant Joan d'Alacant, Alicante, Spain
| | - Roman Olivares
- Instituto de Neurociencias de Alicante (Universidad Miguel Hernandez-Consejo Superior de Investigaciones Cientificas), Campus de Sant Joan, Sant Joan d'Alacant, Alicante, Spain
| | - Eva Benito
- Instituto de Neurociencias de Alicante (Universidad Miguel Hernandez-Consejo Superior de Investigaciones Cientificas), Campus de Sant Joan, Sant Joan d'Alacant, Alicante, Spain
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
- ExcellenceCluster NeuroCure, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Karen Gertz
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
- ExcellenceCluster NeuroCure, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
- ExcellenceCluster NeuroCure, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| | - Andreas Meisel
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB) and Klinik und Hochschulambulanz für Neurologie, Charité–Universitätsmedizin Berlin, Berlin, Germany
- ExcellenceCluster NeuroCure, Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
37
|
Fessler EB, Chibane FL, Wang Z, Chuang DM. Potential roles of HDAC inhibitors in mitigating ischemia-induced brain damage and facilitating endogenous regeneration and recovery. Curr Pharm Des 2014; 19:5105-20. [PMID: 23448466 DOI: 10.2174/1381612811319280009] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/18/2013] [Indexed: 02/06/2023]
Abstract
Ischemic stroke is a leading cause of death and disability worldwide, with few available treatment options. The pathophysiology of cerebral ischemia involves both early phase tissue damage, characterized by neuronal death, inflammation, and blood-brain barrier breakdown, followed by late phase neurovascular recovery. It is becoming clear that any promising treatment strategy must target multiple points in the evolution of ischemic injury to provide substantial therapeutic benefit. Histone deacetylase (HDAC) inhibitors are a class of drugs that increase the acetylation of histone and non-histone proteins to activate transcription, enhance gene expression, and modify the function of target proteins. Acetylation homeostasis is often disrupted in neurological conditions, and accumulating evidence suggests that HDAC inhibitors have robust protective properties in many preclinical models of these disorders, including ischemic stroke. Specifically, HDAC inhibitors such as trichostatin A, valproic acid, sodium butyrate, sodium 4-phenylbutyrate, and suberoylanilide hydroxamic acid have been shown to provide robust protection against excitotoxicity, oxidative stress, ER stress, apoptosis, inflammation, and bloodbrain barrier breakdown. Concurrently, these agents can also promote angiogenesis, neurogenesis and stem cell migration to dramatically reduce infarct volume and improve functional recovery after experimental cerebral ischemia. In the following review, we discuss the mechanisms by which HDAC inhibitors exert these protective effects and provide evidence for their strong potential to ultimately improve stroke outcome in patients.
Collapse
Affiliation(s)
- Emily B Fessler
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, MSC 1363, Bethesda, MD 20892-1363, USA
| | | | | | | |
Collapse
|
38
|
Valor LM, Viosca J, Lopez-Atalaya JP, Barco A. Lysine acetyltransferases CBP and p300 as therapeutic targets in cognitive and neurodegenerative disorders. Curr Pharm Des 2014; 19:5051-64. [PMID: 23448461 PMCID: PMC3722569 DOI: 10.2174/13816128113199990382] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 02/18/2013] [Indexed: 01/27/2023]
Abstract
Neuropsychiatric pathologies, including neurodegenerative diseases and neurodevelopmental syndromes, are frequently associated with dysregulation of various essential cellular mechanisms, such as transcription, mitochondrial respiration and protein degradation. In these complex scenarios, it is difficult to pinpoint the specific molecular dysfunction that initiated the pathology or that led to the fatal cascade of events that ends with the death of the neuron. Among the possible original factors, epigenetic dysregulation has attracted special attention. This review focuses on two highly related epigenetic factors that are directly involved in a number of neurological disorders, the lysine acetyltransferases CREB-binding protein (CBP) and E1A-associated protein p300 (p300). We first comment on the role of chromatin acetylation and the enzymes that control it, particularly CBP and p300, in neuronal plasticity and cognition. Next, we describe the involvement of these proteins in intellectual disability and in different neurodegenerative diseases. Finally, we discuss the potential of ameliorative strategies targeting CBP/p300 for the treatment of these disorders.
Collapse
Affiliation(s)
- Luis M Valor
- Instituto de Neurociencias, Av. Santiago Ramon y Cajal s/n. Sant Joan d'Alacant 03550, Alicante, Spain
| | | | | | | |
Collapse
|
39
|
Trichostatin A increases the levels of plasma gelsolin and amyloid beta-protein in a transgenic mouse model of Alzheimer's disease. Life Sci 2014; 99:31-6. [PMID: 24486299 DOI: 10.1016/j.lfs.2014.01.064] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 12/27/2013] [Accepted: 01/16/2014] [Indexed: 11/20/2022]
Abstract
AIMS Gelsolin (GSN), a multifunctional protein, binds to amyloid beta-protein (Aβ), inhibits its fibrillization, solubilizes preformed Aβ fibrils, and helps in its clearance from the brain. Trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, induces the protein expression of gelsolin. In the present study, we investigated how TSA-treatment of APPswe/PS1δE9 transgenic (Tg) mice of Alzheimer's disease (AD) will affect the plasma levels of gelsolin and Aβ. MAIN METHODS TSA (5mg/kg body weight on alternate days for two months) was intraperitoneally injected to AD Tg mice. Gelsolin was measured by Western blotting and Aβ was measured by enzyme-linked immunosorbent assay. KEY FINDINGS TSA-treatment significantly increased the levels of plasma gelsolin by 1.79-fold as compared with vehicle-treated control mice (p<0.01). The levels of Aβ 1-40 and Aβ 1-42 in the plasma were also higher in TSA-treated mice in comparison with vehicle-treated mice. The treatment of transgenic AD mice with TSA did not affect the body weight in both male and female groups as compared to vehicle-treated animals. A positive correlation was observed between the plasma levels of gelsolin and Aβ 1-40 (r=0.594, p=0.042) or Aβ 1-42 (r=0.616, p=0.033) in AD Tg mice. SIGNIFICANCE These results suggest that TSA increases the levels of plasma gelsolin and Aβ in AD Tg mice, which may have implications in gelsolin-mediated clearance of Aβ.
Collapse
|
40
|
Novitskaya T, McDermott L, Zhang KX, Chiba T, Paueksakon P, Hukriede NA, de Caestecker MP. A PTBA small molecule enhances recovery and reduces postinjury fibrosis after aristolochic acid-induced kidney injury. Am J Physiol Renal Physiol 2013; 306:F496-504. [PMID: 24370591 DOI: 10.1152/ajprenal.00534.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Phenylthiobutanoic acids (PTBAs) are a new class of histone deacetylase (HDAC) inhibitors that accelerate recovery and reduce postinjury fibrosis after ischemia-reperfusion-induced acute kidney injury. However, unlike the more common scenario in which patients present with protracted and less clearly defined onset of renal injury, this model of acute kidney injury gives rise to a clearly defined injury that begins to resolve over a short period of time. In these studies, we show for the first time that treatment with the PTBA analog methyl-4-(phenylthio)butanoate (M4PTB) accelerates recovery and reduces postinjury fibrosis in a progressive model of acute kidney injury and renal fibrosis that occurs after aristolochic acid injection in mice. These effects are apparent when M4PTB treatment is delayed 4 days after the initiating injury and are associated with increased proliferation and decreased G2/M arrest of regenerating renal tubular epithelial cells. In addition, there is reduced peritubular macrophage infiltration and decreased expression of the macrophage chemokines CX3Cl1 and CCL2. Since macrophage infiltration plays a role in promoting kidney injury, and since renal tubular epithelial cells show defective repair and a marked increase in maladaptive G2/M arrest after aristolochic acid injury, these findings suggest M4PTB may be particularly beneficial in reducing injury and enhancing intrinsic cellular repair even when administered days after aristolochic acid ingestion.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Vanderbilt Univ. Medical Center, Dept. of Medicine, Div. of Nephrology, S3223 Medical Center North, 1161 21st Ave. South, Nashville, TN 37232.
| | | | | | | | | | | | | |
Collapse
|
41
|
Thompson JW, Dave KR, Young JI, Perez-Pinzon MA. Ischemic preconditioning alters the epigenetic profile of the brain from ischemic intolerance to ischemic tolerance. Neurotherapeutics 2013; 10:789-97. [PMID: 23868468 PMCID: PMC3805868 DOI: 10.1007/s13311-013-0202-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ischemic preconditioning is an innate neuroprotective mechanism in which a sub-injurious ischemic exposure increases the brain's ability to withstand a subsequent, normally injurious ischemic insult. Part of ischemic preconditioning neuroprotection stems from an epigenetic reprogramming of the brain to a phenotype of ischemic tolerance, which results in a gene expression profile different from that observed in the non-injured and ischemia-injured brains. Such neuroprotective reprograming, activated by ischemic preconditioning, requires specific changes in DNA accessibility coordinated with activation of transcriptional activator and repressor proteins, which allows for expression of specific neuroprotective proteins despite a general repression of gene expression. In this review we examine the effects of injurious ischemia and ischemic preconditioning on the regulation of DNA methylation, histone post-translational modifications, and non-coding RNA expression. There is increasing interest in the role of epigenetics in disease pathobiology, and whether and how pharmacological manipulation of epigenetic processes may allow for ischemic neuroprotection. Therefore, a better understanding of the epigenomic determinants underlying the modulation of gene expression that lead to ischemic tolerance or cell death offers the promise of novel neuroprotective therapies that target global reprograming of genomic activity versus individual cellular signaling pathways.
Collapse
Affiliation(s)
- John W Thompson
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA,
| | | | | | | |
Collapse
|
42
|
Schweizer S, Meisel A, Märschenz S. Epigenetic mechanisms in cerebral ischemia. J Cereb Blood Flow Metab 2013; 33:1335-46. [PMID: 23756691 PMCID: PMC3764391 DOI: 10.1038/jcbfm.2013.93] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 05/14/2013] [Accepted: 05/21/2013] [Indexed: 01/27/2023]
Abstract
Treatment efficacy for ischemic stroke represents a major challenge. Despite fundamental advances in the understanding of stroke etiology, therapeutic options to improve functional recovery remain limited. However, growing knowledge in the field of epigenetics has dramatically changed our understanding of gene regulation in the last few decades. According to the knowledge gained from animal models, the manipulation of epigenetic players emerges as a highly promising possibility to target diverse neurologic pathologies, including ischemia. By altering transcriptional regulation, epigenetic modifiers can exert influence on all known pathways involved in the complex course of ischemic disease development. Beneficial transcriptional effects range from attenuation of cell death, suppression of inflammatory processes, and enhanced blood flow, to the stimulation of repair mechanisms and increased plasticity. Most striking are the results obtained from pharmacological inhibition of histone deacetylation in animal models of stroke. Multiple studies suggest high remedial qualities even upon late administration of histone deacetylase inhibitors (HDACi). In this review, the role of epigenetic mechanisms, including histone modifications as well as DNA methylation, is discussed in the context of known ischemic pathways of damage, protection, and regeneration.
Collapse
Affiliation(s)
- Sophie Schweizer
- Department of Neurology and Experimental Neurology, Center of Stroke Research Berlin, Charité University Medicine, Charitéplatz 1, Berlin, Germany
| | | | | |
Collapse
|
43
|
Sharma B, Sharma PM. Arsenic toxicity induced endothelial dysfunction and dementia: pharmacological interdiction by histone deacetylase and inducible nitric oxide synthase inhibitors. Toxicol Appl Pharmacol 2013; 273:180-8. [PMID: 23921152 DOI: 10.1016/j.taap.2013.07.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 07/18/2013] [Accepted: 07/19/2013] [Indexed: 12/11/2022]
Abstract
Arsenic toxicity has been reported to damage all the major organs including the brain and vasculature. Dementia including Alzheimer's disease (AD) and vascular dementia (VaD) are posing greater risk to the world population as it is now increasing at a faster rate. We have investigated the role of sodium butyrate, a selective histone deacetylase (HDAC) inhibitor and aminoguanidine, a selective inducible nitric oxide synthase (iNOS) inhibitor in pharmacological interdiction of arsenic toxicity induced vascular endothelial dysfunction and dementia in rats. Arsenic toxicity was done by administering arsenic drinking water to rats. Morris water-maze (MWM) test was used for assessment of learning and memory. Endothelial function was assessed using student physiograph. Oxidative stress (aortic superoxide anion, serum and brain thiobarbituric acid reactive species, brain glutathione) and nitric oxide levels (serum nitrite/nitrate) were also measured. Arsenic treated rats have shown impairment of endothelial function, learning and memory, reduction in serum nitrite/nitrate & brain GSH levels along with increase in serum & brain TBARS. Sodium butyrate as well as aminoguanidine significantly convalesce arsenic induced impairment of learning, memory, endothelial function, and alterations in various biochemical parameters. It may be concluded that arsenic induces endothelial dysfunction and dementia, whereas, sodium butyrate, a HDAC inhibitor as well as aminoguanidine, a selective iNOS inhibitor may be considered as potential agents for the management of arsenic induced endothelial dysfunction and dementia.
Collapse
Affiliation(s)
- Bhupesh Sharma
- Department of Pharmacology and Toxicology, School of Pharmacy, Bharat Institute of Technology, Partapur Bypass, Meerut, Uttar Pradesh, India.
| | | |
Collapse
|
44
|
Pan JW, He LN, Xiao F, Shen J, Zhan RY. Plasma gelsolin levels and outcomes after aneurysmal subarachnoid hemorrhage. Crit Care 2013; 17:R149. [PMID: 23880145 PMCID: PMC4057205 DOI: 10.1186/cc12828] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 07/23/2013] [Indexed: 11/28/2022] Open
Abstract
Introduction Lower gelsolin levels have been associated with the severity and poor outcome of critical illness. Nevertheless, their link with clinical outcomes of aneurysmal subarachnoid hemorrhage is unknown. Therefore, we aimed to investigate the relationship between plasma gelsolin levels and clinical outcomes in patients with aneurysmal subarachnoid hemorrhage. Methods A total of 262 consecutive patients and 150 healthy subjects were included. Plasma gelsolin levels were measured by enzyme-linked immunosorbent assay. Mortality and poor long-term outcome (Glasgow Outcome Scale score of 1-3) at 6 months were recorded. Results Plasma gelsolin levels on admission were substantially lower in patients than in healthy controls (66.9 (26.4) mg/L vs. 126.4 (35.4) mg/L, P < 0.001), and negatively associated with World Federation of Neurological Surgeons score (r = -0.554, P < 0.001) and Fisher score (r = -0.538, P < 0.001), and identified as an independent predictor of poor functional outcome (odds ratio, 0.957; 95% confidence interval (CI), 0.933-0.983; P = 0.001) and death (odds ratio, 0.953; 95% CI, 0.917-0.990; P = 0.003) after 6 months. The areas under the ROC curve of gelsolin for functional outcome and mortality were similar to those of World Federation of Neurological Surgeons score and Fisher score (all P > 0.05). Gelsolin improved the predictive values of World Federation of Neurological Surgeons score and Fisher score for functional outcome (both P < 0.05), but not for mortality (both P > 0.05). Conclusions Gelsolin levels are a useful, complementary tool to predict functional outcome and mortality 6 months after aneurysmal subarachnoid hemorrhage.
Collapse
|
45
|
Transglutaminase is a therapeutic target for oxidative stress, excitotoxicity and stroke: a new epigenetic kid on the CNS block. J Cereb Blood Flow Metab 2013; 33:809-18. [PMID: 23571278 PMCID: PMC3677119 DOI: 10.1038/jcbfm.2013.53] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Transglutaminases (TGs) are multifunctional, calcium-dependent enzymes that have been recently implicated in stroke pathophysiology. Classically, these enzymes are thought to participate in cell injury and death in chronic neurodegenerative conditions via their ability to catalyze covalent, nondegradable crosslinks between proteins or to incorporate polyamines into protein substrates. Accumulating lines of inquiry indicate that specific TG isoforms can shuttle into the nucleus when they sense pathologic changes in calcium or oxidative stress, bind to chromatin and thereby transduce these changes into transcriptional repression of genes involved in metabolic or oxidant adaptation. Here, we review the evidence that supports principally a role for one isoform of this family, TG2, in cell injury and death associated with hemorrhagic or ischemic stroke. We also outline an evolving model in which TG2 is a critical mediator between pathologic signaling and epigenetic modifications that lead to gene repression. Accordingly, the salutary effects of TG inhibitors in stroke may derive from their ability to restore homeostasis by removing inappropriate deactivation of adaptive genetic programs by oxidative stress or extrasynaptic glutamate receptor signaling.
Collapse
|
46
|
Epigenetics and the environment: in search of the "toleroasome" vital to execution of ischemic preconditioning. Transl Stroke Res 2013; 4:56-62. [PMID: 24323190 DOI: 10.1007/s12975-012-0235-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 11/29/2012] [Accepted: 11/29/2012] [Indexed: 01/02/2023]
Abstract
Activation and repression of gene expression are key features of ischemic tolerance. Converging lines of inquiry from several groups suggests that epigenetic proteins may transduce sublethal stresses, including bioenergetic or oxidative stress into durable (2-3 days) changes in gene expression that mediate ischemic tolerance. Here we discuss the potential mechanisms by which changes in cell state (e.g., ATP, NAD+, and oxygen) can modify specific targets including polycomb complexes, jumonji domain histone demethylases, and zinc and NAD-dependent histone decetylases and thus trigger an adaptive program. A major unanswered question is whether these proteins work in parallel or convergently as part of a "tolerosome" (tolero is the Latin word for tolerance), a multiprotein complex recruited to promoters or enhancers of specific genes, to mediate preconditioning. Whatever the case may be, epigenetic proteins are fertile targets for the treatment of stroke.
Collapse
|
47
|
Abstract
Many of the best-studied actin regulatory proteins use non-covalent means to modulate the properties of actin. Yet, actin is also susceptible to covalent modifications of its amino acids. Recent work is increasingly revealing that actin processing and its covalent modifications regulate important cellular events. In addition, numerous pathogens express enzymes that specifically use actin as a substrate to regulate their hosts' cells. Actin post-translational alterations have been linked to different normal and disease processes and the effects associated with metabolic and environmental stressors. Herein, we highlight specific co-translational and post-translational modifications of actin and discuss the current understanding of the role that these modifications play in regulating actin.
Collapse
Affiliation(s)
- Jonathan R Terman
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | | |
Collapse
|
48
|
Zhang QH, Li JC, Dong N, Tang LM, Zhu XM, Sheng ZY, Yao YM. Burn injury induces gelsolin expression and cleavage in the brain of mice. Neuroscience 2012; 228:60-72. [PMID: 23079629 DOI: 10.1016/j.neuroscience.2012.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/20/2012] [Accepted: 10/04/2012] [Indexed: 11/26/2022]
Abstract
Gelsolin is an actin filament-severing and capping protein, affecting cellular motility, adhesiveness and apoptosis. Whether it is expressed in the brain of burned mice has not yet been characterized. Mice were subjected to a 15% total body surface area scald injury. Neuropathology was examined by hematoxylin and eosin staining. Cerebral gelsolin mRNA, distribution and cleavage were demonstrated by quantitative polymerase chain reaction (QPCR), immunohistochemistry and Western blot, respectively. Cysteinyl aspartate-specific protease (caspase)-3-positive cells and activity were also measured. Burn injury could induce pathological alterations in the brain including leukocyte infiltration, necrosis, microabscess and gliosis. Compared with sham-injured mice, gelsolin mRNA was up-regulated at 8h post-burn (pb) in a transient manner in the cortex and striatum of burned mice, while it remained at higher levels in the hippocampus up to 72 hpb. Of interest, gelsolin was further cleaved into 42 and 48 kDa (kilo Dalton) fragments as illustrated in the hippocampus at 24 hpb, and was widely expressed in the brain by activated monocyte/macrophages, astrocytes and damaged neurons. In the meantime, caspase-3-positive cells were noted in the striatum of burned mice and its activity peaked at 24 hpb. To clarify inflammation-induced gelsolin expression and cleavage in the brain, rat pheochromocytoma cells were exposed to lipopolysaccharide to show increased gelsolin expression and caspase-3-dependent cleavage. The results suggest that burn-induced cerebral gelsolin expression would be involved in the activation of both the monocytes and astroglial cells, thereby playing a crucial role in the subsequent inflammation-induced neural apoptosis following burn injury.
Collapse
Affiliation(s)
- Qing-Hong Zhang
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing 100048, PR China
| | | | | | | | | | | | | |
Collapse
|
49
|
Hippocampal dendritic spines modifications induced by perinatal asphyxia. Neural Plast 2012; 2012:873532. [PMID: 22645692 PMCID: PMC3356716 DOI: 10.1155/2012/873532] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 11/17/2011] [Accepted: 11/20/2011] [Indexed: 01/16/2023] Open
Abstract
Perinatal asphyxia (PA) affects the synaptic function and morphological organization. In previous works, we have shown neuronal and synaptic changes in rat neostriatum subjected to hypoxia leading to long-term ubi-protein accumulation. Since F-actin is highly concentrated in dendritic spines, modifications in its organization could be related with alterations induced by hypoxia in the central nervous system (CNS). In the present study, we investigate the effects of PA on the actin cytoskeleton of hippocampal postsynaptic densities (PSD) in 4-month-old rats. PSD showed an increment in their thickness and in the level of ubiquitination. Correlative fluorescence-electron microscopy photooxidation showed a decrease in the number of F-actin-stained spines in hippocampal excitatory synapses subjected to PA. Although Western Blot analysis also showed a slight decrease in β-actin in PSD in PA animals, the difference was not significant. Taken together, this data suggests that long-term actin cytoskeleton might have role in PSD alterations which would be a spread phenomenon induced by PA.
Collapse
|
50
|
Xu JF, Liu WG, Dong XQ, Yang SB, Fan J. Change in plasma gelsolin level after traumatic brain injury. J Trauma Acute Care Surg 2012; 72:491-6. [PMID: 22439219 DOI: 10.1097/ta.0b013e318226ec39] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Plasma gelsolin depletion has been associated with poor outcome of critically ill patients. However, there is a paucity of data available on circulating plasma gelsolin concentration in traumatic brain injury (TBI). Thus, we sought to investigate change in plasma gelsolin level after TBI and to evaluate its relation with disease outcome. METHODS Fifty healthy controls and 94 patients with acute severe TBI were included. Plasma samples were obtained on admission and at days 1, 2, 3, 5, and 7. Its concentration was measured by enzyme-linked immunosorbent assay. RESULTS Twenty-six patients (27.7%) died from TBI in a month. After TBI, plasma gelsolin level in patients decreased during the 6-hour period immediately, was at the nadir in 24 hours, increased gradually thereafter, and was substantially lower than that in healthy controls during the 7-day period. A multivariate analysis showed plasma gelsolin level was an independent predictor for 1-month mortality (odds ratio, 0.941; 95% confidence interval, 0.895– 0.989; p = 0.017) and positively associated with Glasgow Coma Scale (GCS) score (t = 6.538, p 0.001). A receiver operating characteristic curve identified that a baseline plasma gelsolin level 52.7 mg/L predicted 1-month mortality with 88.5% sensitivity and 79.4% specificity (area under the curve, 0.869; 95%confidence interval, 0.783– 0.930). The predictive value of the gelsolin concentration was thus similar to that of GCS scores (p =0.185). However, gelsolin did not statistically significantly improve the area under the curve of GCS scores (p = 0.517). CONCLUSIONS Decreased plasma gelsolin level is associated with GCS scores and an independent prognostic marker of mortality after TBI. Reversing plasma gelsolin deficiency may be an effective treatment for TBI.
Collapse
Affiliation(s)
- Jin-Fang Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, China
| | | | | | | | | |
Collapse
|