1
|
Azman KF, Zakaria R. Brain-Derived Neurotrophic Factor (BDNF) in Huntington's Disease: Neurobiology and Therapeutic Potential. Curr Neuropharmacol 2025; 23:384-403. [PMID: 40123457 DOI: 10.2174/1570159x22666240530105516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2025] Open
Abstract
Huntington's disease is a hereditary neurodegenerative disorder marked by severe neurodegeneration in the striatum and cortex. Brain-derived neurotrophic factor (BDNF) is a member of the neurotrophin family of growth factors. It plays a crucial role in maintaining the survival and proper function of striatal neurons. Depletion of BDNF has been linked to impairment and death of striatal neurons, leading to the manifestation of motor, cognitive, and behavioral dysfunctions characteristic of Huntington's disease. This review highlights the current update on the neurobiology of BDNF in the pathogenesis of Huntington's disease. The molecular evidence and the affected signaling pathways are also discussed. In addition, the impact of experimental manipulation of BDNF levels and its pharmaceutical potential for Huntington's disease treatment are explicitly reviewed.
Collapse
Affiliation(s)
- Khairunnuur Fairuz Azman
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| | - Rahimah Zakaria
- Department of Physiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
2
|
Carvalho LB, Baroudi K, França C, Gonçalves AAS, Bernadi MM, da Silva RAF. LPS-induced neuroinflammation induces changes in the transcriptional profile of members of the CoRest repressive complex in the hippocampus. Mol Biol Rep 2024; 51:1156. [PMID: 39546128 DOI: 10.1007/s11033-024-09984-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/05/2024] [Indexed: 11/17/2024]
Abstract
The action of Corepressor for Element Silencing Transcription Factor 1 (CoREST) is primarily related to neural fate decisions. However, the molecular mechanisms linking neuroinflammation to the histone modifying complex remain unclear. CoREST is a hub for several cofactors that play important roles in epigenetic remodeling and transcriptional regulation. It allows us to question their functions during the inflammatory response in the Central Nervous System. The impact of LPS-induced neuroinflammation on the transcriptional epigenetic control of members with CoRest repressive complex in the hippocampus was investigated. Characterizing the basal transcriptional profile in the hippocampus of members with CoREST repressive complex showed that the Rcor3 is the most expressed gene, and the Rcor2 is the least expressed one. It was also demonstrated that the levels of Lsd1, CoREST1 (Rcor1), and CoREST2 (Rcor2) transcripts increased in the hippocampus after LPS i.p. administration, while for CoREST3 (Rcor3), no significant difference was observed. A significant increase was noticed in the percentages of the 5-meC mark (Hypermethylation) for the Rcor1 and Lsd1 genes with a positive Pearson correlation between methylation and expression. However, the correlation was directly proportional, ruling out DNA methylation as the main mechanism in transcriptional control.
Collapse
Affiliation(s)
| | - Kusai Baroudi
- Department of Clinical Sciences, College of Dentistry, Ajman University, Ajman, United Arab Emirates
| | - Cleiton França
- CEEpiRG - Center for Epigenetic Study and Genic Regulation, Program in Environmental and Experimental Pathology, Paulista University - UNIP, São Paulo, SP, Brazil
| | - Arila Adorno Scorzafava Gonçalves
- CEEpiRG - Center for Epigenetic Study and Genic Regulation, Program in Environmental and Experimental Pathology, Paulista University - UNIP, São Paulo, SP, Brazil
| | - Maria Martha Bernadi
- CEEpiRG - Center for Epigenetic Study and Genic Regulation, Program in Environmental and Experimental Pathology, Paulista University - UNIP, São Paulo, SP, Brazil
| | - Rodrigo Augusto Foganholi da Silva
- Postgraduate Program in Health Sciences, University of Taubaté, Taubaté, São Paulo, SP, Brazil.
- CEEpiRG - Center for Epigenetic Study and Genic Regulation, Program in Environmental and Experimental Pathology, Paulista University - UNIP, São Paulo, SP, Brazil.
- Postgraduate Program in Health Sciences, University of Taubaté - UNITAU, Taubaté, Brazil.
| |
Collapse
|
3
|
Tandon S, Aggarwal P, Sarkar S. Polyglutamine disorders: Pathogenesis and potential drug interventions. Life Sci 2024; 344:122562. [PMID: 38492921 DOI: 10.1016/j.lfs.2024.122562] [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: 11/02/2023] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Polyglutamine/poly(Q) diseases are a group nine hereditary neurodegenerative disorders caused due to abnormally expanded stretches of CAG trinucleotide in functionally distinct genes. All human poly(Q) diseases are characterized by the formation of microscopically discernable poly(Q) positive aggregates, the inclusion bodies. These toxic inclusion bodies are responsible for the impairment of several cellular pathways such as autophagy, transcription, cell death, etc., that culminate in disease manifestation. Although, these diseases remain largely without treatment, extensive research has generated mounting evidences that various events of poly(Q) pathogenesis can be developed as potential drug targets. The present review article briefly discusses the key events of disease pathogenesis, model system-based investigations that support the development of effective therapeutic interventions against pathogenesis of human poly(Q) disorders, and a comprehensive list of pharmacological and bioactive compounds that have been experimentally shown to alleviate poly(Q)-mediated neurotoxicity. Interestingly, due to the common cause of pathogenesis, all poly(Q) diseases share etiology, thus, findings from one disease can be potentially extrapolated to other poly(Q) diseases as well.
Collapse
Affiliation(s)
- Shweta Tandon
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Prerna Aggarwal
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India.
| |
Collapse
|
4
|
Jin L, Liu Y, Wu Y, Huang Y, Zhang D. REST Is Not Resting: REST/NRSF in Health and Disease. Biomolecules 2023; 13:1477. [PMID: 37892159 PMCID: PMC10605157 DOI: 10.3390/biom13101477] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Chromatin modifications play a crucial role in the regulation of gene expression. The repressor element-1 (RE1) silencing transcription factor (REST), also known as neuron-restrictive silencer factor (NRSF) and X2 box repressor (XBR), was found to regulate gene transcription by binding to chromatin and recruiting chromatin-modifying enzymes. Earlier studies revealed that REST plays an important role in the development and disease of the nervous system, mainly by repressing the transcription of neuron-specific genes. Subsequently, REST was found to be critical in other tissues, such as the heart, pancreas, skin, eye, and vascular. Dysregulation of REST was also found in nervous and non-nervous system cancers. In parallel, multiple strategies to target REST have been developed. In this paper, we provide a comprehensive summary of the research progress made over the past 28 years since the discovery of REST, encompassing both physiological and pathological aspects. These insights into the effects and mechanisms of REST contribute to an in-depth understanding of the transcriptional regulatory mechanisms of genes and their roles in the development and progression of disease, with a view to discovering potential therapeutic targets and intervention strategies for various related diseases.
Collapse
Affiliation(s)
- Lili Jin
- School of Life Sciences, Liaoning University, Shenyang 110036, China
| | - Ying Liu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Yifan Wu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Yi Huang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Dianbao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| |
Collapse
|
5
|
Lam XJ, Maniam S, Cheah PS, Ling KH. REST in the Road Map of Brain Development. Cell Mol Neurobiol 2023; 43:3417-3433. [PMID: 37517069 PMCID: PMC11410019 DOI: 10.1007/s10571-023-01394-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
Repressor element-1 silencing transcription factor (REST) or also known as neuron-restrictive silencing factor (NRSF), is the key initiator of epigenetic neuronal gene-expression modification. Identification of a massive number of REST-targeted genes in the brain signifies its broad involvement in maintaining the functionality of the nervous system. Additionally, REST plays a crucial role in conferring neuroprotection to the neurons against various stressors or insults during injuries. At the cellular level, nuclear localisation of REST is a key determinant for the functional transcriptional regulation of REST towards its target genes. Emerging studies reveal the implication of REST nuclear mislocalisation or dysregulation in several neurological diseases. The expression of REST varies depending on different types of neurological disorders, which has created challenges in the discovery of REST-targeted interventions. Hence, this review presents a comprehensive summary on the physiological roles of REST throughout brain development and its implications in neurodegenerative and neurodevelopmental disorders, brain tumours and cerebrovascular diseases. This review offers valuable insights to the development of potential therapeutic approaches targeting REST to improve pathologies in the brain. The important roles of REST as a key player in the nervous system development, and its implications in several neurological diseases.
Collapse
Affiliation(s)
- Xin-Jieh Lam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Sandra Maniam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Pike-See Cheah
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Malaysian Research Institute on Ageing (MyAgeing), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
- Malaysian Research Institute on Ageing (MyAgeing), Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
6
|
Speidell A, Bin Abid N, Yano H. Brain-Derived Neurotrophic Factor Dysregulation as an Essential Pathological Feature in Huntington's Disease: Mechanisms and Potential Therapeutics. Biomedicines 2023; 11:2275. [PMID: 37626771 PMCID: PMC10452871 DOI: 10.3390/biomedicines11082275] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a major neurotrophin whose loss or interruption is well established to have numerous intersections with the pathogenesis of progressive neurological disorders. There is perhaps no greater example of disease pathogenesis resulting from the dysregulation of BDNF signaling than Huntington's disease (HD)-an inherited neurodegenerative disorder characterized by motor, psychiatric, and cognitive impairments associated with basal ganglia dysfunction and the ultimate death of striatal projection neurons. Investigation of the collection of mechanisms leading to BDNF loss in HD highlights this neurotrophin's importance to neuronal viability and calls attention to opportunities for therapeutic interventions. Using electronic database searches of existing and forthcoming research, we constructed a literature review with the overarching goal of exploring the diverse set of molecular events that trigger BDNF dysregulation within HD. We highlighted research that investigated these major mechanisms in preclinical models of HD and connected these studies to those evaluating similar endpoints in human HD subjects. We also included a special focus on the growing body of literature detailing key transcriptomic and epigenetic alterations that affect BDNF abundance in HD. Finally, we offer critical evaluation of proposed neurotrophin-directed therapies and assessed clinical trials seeking to correct BDNF expression in HD individuals.
Collapse
Affiliation(s)
- Andrew Speidell
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (A.S.); (N.B.A.)
| | - Noman Bin Abid
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (A.S.); (N.B.A.)
| | - Hiroko Yano
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (A.S.); (N.B.A.)
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
7
|
Ferlazzo GM, Gambetta AM, Amato S, Cannizzaro N, Angiolillo S, Arboit M, Diamante L, Carbognin E, Romani P, La Torre F, Galimberti E, Pflug F, Luoni M, Giannelli S, Pepe G, Capocci L, Di Pardo A, Vanzani P, Zennaro L, Broccoli V, Leeb M, Moro E, Maglione V, Martello G. Genome-wide screening in pluripotent cells identifies Mtf1 as a suppressor of mutant huntingtin toxicity. Nat Commun 2023; 14:3962. [PMID: 37407555 DOI: 10.1038/s41467-023-39552-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by CAG-repeat expansions in the huntingtin (HTT) gene. The resulting mutant HTT (mHTT) protein induces toxicity and cell death via multiple mechanisms and no effective therapy is available. Here, we employ a genome-wide screening in pluripotent mouse embryonic stem cells (ESCs) to identify suppressors of mHTT toxicity. Among the identified suppressors, linked to HD-associated processes, we focus on Metal response element binding transcription factor 1 (Mtf1). Forced expression of Mtf1 counteracts cell death and oxidative stress caused by mHTT in mouse ESCs and in human neuronal precursor cells. In zebrafish, Mtf1 reduces malformations and apoptosis induced by mHTT. In R6/2 mice, Mtf1 ablates motor defects and reduces mHTT aggregates and oxidative stress. Our screening strategy enables a quick in vitro identification of promising suppressor genes and their validation in vivo, and it can be applied to other monogenic diseases.
Collapse
Affiliation(s)
- Giorgia Maria Ferlazzo
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
- Aptuit (Verona) S.r.l., an Evotec Company, Campus Levi-Montalcini, 37135, Verona, Italy
| | - Anna Maria Gambetta
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Sonia Amato
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
- Department of Neuroscience, University of Padova, Via Belzoni, 160, 35131, Padua, Italy
| | - Noemi Cannizzaro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Silvia Angiolillo
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Mattia Arboit
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Linda Diamante
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Elena Carbognin
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Federico La Torre
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy
| | - Elena Galimberti
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Florian Pflug
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Mirko Luoni
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Serena Giannelli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
| | | | | | | | - Paola Vanzani
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Lucio Zennaro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | - Vania Broccoli
- Division of Neuroscience, San Raffaele Scientific Institute, 20132, Milan, Italy
- CNR Institute of Neuroscience, 20854, Vedrano al Lambro, Italy
| | - Martin Leeb
- Max Perutz Laboratories Vienna, University of Vienna, Vienna Biocenter, Dr Bohr Gasse 9, 1030, Vienna, Austria
| | - Enrico Moro
- Department of Molecular Medicine, Medical School, University of Padua, 35131, Padua, Italy
| | | | - Graziano Martello
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131, Padua, Italy.
| |
Collapse
|
8
|
Hernández-Silva D, Alcaraz-Pérez F, Pérez-Sánchez H, Cayuela ML. Virtual screening and zebrafish models in tandem, for drug discovery and development. Expert Opin Drug Discov 2022:1-13. [DOI: 10.1080/17460441.2022.2147503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- David Hernández-Silva
- Telomerase, Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Structural Bioinformatics and High-Performance Computing Research Group (BIOHPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Francisca Alcaraz-Pérez
- Telomerase, Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| | - Horacio Pérez-Sánchez
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| | - Maria Luisa Cayuela
- Telomerase, Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| |
Collapse
|
9
|
Chia K, Klingseisen A, Sieger D, Priller J. Zebrafish as a model organism for neurodegenerative disease. Front Mol Neurosci 2022; 15:940484. [PMID: 36311026 PMCID: PMC9606821 DOI: 10.3389/fnmol.2022.940484] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
The zebrafish is increasingly recognized as a model organism for translational research into human neuropathology. The zebrafish brain exhibits fundamental resemblance with human neuroanatomical and neurochemical pathways, and hallmarks of human brain pathology such as protein aggregation, neuronal degeneration and activation of glial cells, for example, can be modeled and recapitulated in the fish central nervous system. Genetic manipulation, imaging, and drug screening are areas where zebrafish excel with the ease of introducing mutations and transgenes, the expression of fluorescent markers that can be detected in vivo in the transparent larval stages overtime, and simple treatment of large numbers of fish larvae at once followed by automated screening and imaging. In this review, we summarize how zebrafish have successfully been employed to model human neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, and Huntington’s disease. We discuss advantages and disadvantages of choosing zebrafish as a model for these neurodegenerative conditions.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Klingseisen
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
| | - Dirk Sieger
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Dirk Sieger,
| | - Josef Priller
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- United Kingdom Dementia Research Institute at University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, DZNE, Berlin, Germany
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Josef Priller,
| |
Collapse
|
10
|
Butler-Ryan R, Wood IC. The functions of repressor element 1-silencing transcription factor in models of epileptogenesis and post-ischemia. Metab Brain Dis 2021; 36:1135-1150. [PMID: 33813634 PMCID: PMC8272694 DOI: 10.1007/s11011-021-00719-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/17/2021] [Indexed: 12/14/2022]
Abstract
Epilepsy is a debilitating neurological disorder characterised by recurrent seizures for which 30% of patients are refractory to current treatments. The genetic and molecular aetiologies behind epilepsy are under investigation with the goal of developing new epilepsy medications. The transcriptional repressor REST (Repressor Element 1-Silencing Transcription factor) is a focus of interest as it is consistently upregulated in epilepsy patients and following brain insult in animal models of epilepsy and ischemia. This review analyses data from different epilepsy models and discusses the contribution of REST to epileptogenesis. We propose that in healthy brains REST acts in a protective manner to homeostatically downregulate increases in excitability, to protect against seizure through downregulation of BDNF (Brain-Derived Neurotrophic Factor) and its receptor, TrkB (Tropomyosin receptor kinase B). However, in epilepsy patients and post-seizure, REST may increase to a larger degree, which allows downregulation of the glutamate receptor subunit GluR2. This leads to AMPA glutamate receptors lacking GluR2 subunits, which have increased permeability to Ca2+, causing excitotoxicity, cell death and seizure. This concept highlights therapeutic potential of REST modulation through gene therapy in epilepsy patients.
Collapse
Affiliation(s)
- Ruth Butler-Ryan
- School of Biomedical Sciences, Faculty of Biological Sciences, The University of Leeds, Leeds, LS2 9JT UK
| | - Ian C. Wood
- School of Biomedical Sciences, Faculty of Biological Sciences, The University of Leeds, Leeds, LS2 9JT UK
| |
Collapse
|
11
|
Pogoda A, Chmielewska N, Maciejak P, Szyndler J. Transcriptional Dysregulation in Huntington's Disease: The Role in Pathogenesis and Potency for Pharmacological Targeting. Curr Med Chem 2021; 28:2783-2806. [PMID: 32628586 DOI: 10.2174/0929867327666200705225821] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/15/2020] [Accepted: 06/19/2020] [Indexed: 11/22/2022]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a mutation in the gene that encodes a critical cell regulatory protein, huntingtin (Htt). The expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats causes improper folding of functional proteins and is an initial trigger of pathological changes in the brain. Recent research has indicated that the functional dysregulation of many transcription factors underlies the neurodegenerative processes that accompany HD. These disturbances are caused not only by the loss of wild-type Htt (WT Htt) function but also by the occurrence of abnormalities that result from the action of mutant Htt (mHtt). In this review, we aim to describe the role of transcription factors that are currently thought to be strongly associated with HD pathogenesis, namely, RE1-silencing transcription factor, also known as neuron-restrictive silencer factor (REST/NRSF), forkhead box proteins (FOXPs), peroxisome proliferator-activated receptor gamma coactivator-1a (PGC1α), heat shock transcription factor 1 (HSF1), and nuclear factor κ light-chain-enhancer of activated B cells (NF- κB). We also take into account the role of these factors in the phenotype of HD as well as potential pharmacological interventions targeting the analyzed proteins. Furthermore, we considered whether molecular manipulation resulting in changes in transcription factor function may have clinical potency for treating HD.
Collapse
Affiliation(s)
- Aleksandra Pogoda
- Faculty of Medicine, Medical University of Warsaw, Zwirki i Wigury Street 61, 02-097 Warsaw, Poland
| | - Natalia Chmielewska
- Department of Neurochemistry, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland
| | - Piotr Maciejak
- Department of Neurochemistry, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland
| | - Janusz Szyndler
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, Banacha Street 1B, 02-097 Warsaw, Poland
| |
Collapse
|
12
|
Difference of binding modes among three ligands to a receptor mSin3B corresponding to their inhibitory activities. Sci Rep 2021; 11:6178. [PMID: 33731831 PMCID: PMC7971087 DOI: 10.1038/s41598-021-85612-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 02/19/2021] [Indexed: 11/08/2022] Open
Abstract
A preceding experiment suggested that a compound, which inhibits binding of the REST/NRSF segment to the cleft of a receptor protein mSin3B, can be a potential drug candidate to ameliorate many neuropathies. We have recently developed an enhanced conformational sampling method, genetic-algorithm-guided multi-dimensional virtual-system-coupled canonical molecular dynamics, and in the present study, applied it to three systems consisting of mSin3B and one of three compounds, sertraline, YN3, and acitretin. Other preceding experiments showed that only sertraline inhibits the binding of REST/NRSF to mSin3B. The current simulation study produced the spatial distribution of the compounds around mSin3B, and showed that sertraline and YN3 bound to the cleft of mSin3B with a high propensity, although acitretin did not. Further analyses of the simulation data indicated that only the sertraline-mSin3B complex produced a hydrophobic core similar to that observed in the molecular interface of the REST/NRSF-mSin3B complex: An aromatic ring of sertraline sunk deeply in the mSin3B's cleft forming a hydrophobic core contacting to hydrophobic amino-acid residues located at the bottom of the cleft. The present study proposes a step to design a compound that inhibits competitively the binding of a ligand to its receptor.
Collapse
|
13
|
More than a Corepressor: The Role of CoREST Proteins in Neurodevelopment. eNeuro 2020; 7:ENEURO.0337-19.2020. [PMID: 32075869 PMCID: PMC7070449 DOI: 10.1523/eneuro.0337-19.2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/19/2020] [Accepted: 02/05/2020] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms governing normal neurodevelopment are tightly regulated by the action of transcription factors. Repressor element 1 (RE1) silencing transcription factor (REST) is widely documented as a regulator of neurogenesis that acts by recruiting corepressor proteins and repressing neuronal gene expression in non-neuronal cells. The REST corepressor 1 (CoREST1), CoREST2, and CoREST3 are best described for their role as part of the REST complex. However, recent evidence has shown the proteins have the ability to repress expression of distinct target genes in a REST-independent manner. These findings indicate that each CoREST paralogue may have distinct and critical roles in regulating neurodevelopment and are more than simply “REST corepressors,” whereby they act as independent repressors orchestrating biological processes during neurodevelopment.
Collapse
|
14
|
Theoretical and Experimental Approaches Aimed at Drug Design Targeting Neurodegenerative Diseases. Processes (Basel) 2019. [DOI: 10.3390/pr7120940] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In recent years, green chemistry has been strengthening, showing how basic and applied sciences advance globally, protecting the environment and human health. A clear example of this evolution is the synergy that now exists between theoretical and computational methods to design new drugs in the most efficient possible way, using the minimum of reagents and obtaining the maximum yield. The development of compounds with potential therapeutic activity against multiple targets associated with neurodegenerative diseases/disorders (NDD) such as Alzheimer’s disease is a hot topic in medical chemistry, where different scientists from various disciplines collaborate to find safe, active, and effective drugs. NDD are a public health problem, affecting mainly the population over 60 years old. To generate significant progress in the pharmacological treatment of NDD, it is necessary to employ different experimental strategies of green chemistry, medical chemistry, and molecular biology, coupled with computational and theoretical approaches such as molecular simulations and chemoinformatics, all framed in the rational drug design targeting NDD. Here, we review how green chemistry and computational approaches have been used to develop new compounds with the potential application against NDD, as well as the challenges and new directions of the drug development multidisciplinary process.
Collapse
|
15
|
Marisetty AL, Lu L, Veo BL, Liu B, Coarfa C, Kamal MM, Kassem DH, Irshad K, Lu Y, Gumin J, Henry V, Paulucci-Holthauzen A, Rao G, Baladandayuthapani V, Lang FF, Fuller GN, Majumder S. REST-DRD2 mechanism impacts glioblastoma stem cell-mediated tumorigenesis. Neuro Oncol 2019; 21:775-785. [PMID: 30953587 PMCID: PMC6556851 DOI: 10.1093/neuonc/noz030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a lethal, heterogeneous human brain tumor, with regulatory mechanisms that have yet to be fully characterized. Previous studies have indicated that the transcriptional repressor REST (repressor element-1 silencing transcription factor) regulates the oncogenic potential of GBM stem cells (GSCs) based on level of expression. However, how REST performs its regulatory role is not well understood. METHODS We examined 2 independent high REST (HR) GSC lines using genome-wide assays, biochemical validations, gene knockdown analysis, and mouse tumor models. We analyzed in-house patient tumors and patient data present in The Cancer Genome Atlas (TCGA). RESULTS Genome-wide transcriptome and DNA-binding analyses suggested the dopamine receptor D2 (DRD2) gene, a dominant regulator of neurotransmitter signaling, as a direct target of REST. Biochemical analyses and mouse intracranial tumor models using knockdown of REST and double knockdown of REST and DRD2 validated this target and suggested that DRD2 is a downstream target of REST regulating tumorigenesis, at least in part, through controlling invasion and apoptosis. Further, TCGA GBM data support the presence of the REST-DRD2 axis and reveal that high REST/low DRD2 (HRLD) and low REST/high DRD2 (LRHD) tumors are specific subtypes, are molecularly different from the known GBM subtypes, and represent functional groups with distinctive patterns of enrichment of gene sets and biological pathways. The inverse HRLD/LRHD expression pattern is also seen in in-house GBM tumors. CONCLUSIONS These findings suggest that REST regulates neurotransmitter signaling pathways through DRD2 in HR-GSCs to impact tumorigenesis. They further suggest that the REST-DRD2 mechanism forms distinct subtypes of GBM.
Collapse
Affiliation(s)
- Anantha L Marisetty
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Li Lu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bethany L Veo
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bin Liu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Mohamed Mostafa Kamal
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dina Hamada Kassem
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Khushboo Irshad
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yungang Lu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Verlene Henry
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sadhan Majumder
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
16
|
Hu X, Contini A. Rescoring Virtual Screening Results with the MM-PBSA Methods: Beware of Internal Dielectric Constants. J Chem Inf Model 2019; 59:2714-2728. [PMID: 31063686 DOI: 10.1021/acs.jcim.9b00095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
With the potential of improving virtual screening outcome, MM-PB/GBSA has become a disputed method that requires extensive testing and tuning to provide the optimal results. One of the tuning factors is the internal or solute dielectric constant. We have applied three test sets with receptors of different categories and libraries from different sources to investigate the underlying issue related to this constant. We discovered that increasing internal dielectric value does not improve the virtual screening enrichment qualitatively. More interestingly, nonpolar and polar calculated energies act differently in libraries with different molecular weight distributions. From this work, the performance of MM-PBSA rescoring in virtual screening is more library- than receptor-dependent.
Collapse
Affiliation(s)
- Xiao Hu
- Dipartimento di Scienze Farmaceutiche - Sezione di Chimica Generale e Organica "Alessandro Marchesini" , Università degli Studi di Milano , Via Venezian, 21 , 20133 Milano , Italy
| | - Alessandro Contini
- Dipartimento di Scienze Farmaceutiche - Sezione di Chimica Generale e Organica "Alessandro Marchesini" , Università degli Studi di Milano , Via Venezian, 21 , 20133 Milano , Italy
| |
Collapse
|
17
|
NRSF and Its Epigenetic Effectors: New Treatments for Neurological Disease. Brain Sci 2018; 8:brainsci8120226. [PMID: 30572571 PMCID: PMC6316267 DOI: 10.3390/brainsci8120226] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/02/2022] Open
Abstract
The Neuron Restrictive Silencer Factor (NRSF) is the well-known master transcriptional repressor of the neuronal phenotype. Research to date has shown that it is an important player in the growth and development of the nervous system. Its role in the maturation of neural precursor cells to adult neurons has been well characterized in stem cell models. While much has been characterized from a developmental perspective, research is revealing that NRSF plays a role in various neurological diseases, ranging from neurodegenerative, neuropsychiatric, to cancer. Dysregulation of NRSF activity disrupts downstream gene expression that is responsible for neuronal cell homeostasis in several models that contribute to pathologic states. Interestingly, it is now becoming apparent that the dysregulation of NRSF contributes to neurological disease through epigenetic mechanisms. Although NRSF itself is a transcription factor, its major effectors are chromatin modifiers. At the level of epigenetics, changes in NRSF activity have been well characterized in models of neuropathic pain and epilepsy. Better understanding of the epigenetic basis of brain diseases has led to design and use of small molecules that can prevent NRSF from repressing gene expression by neutralizing its interactions with its chromatin remodelers. This review will address the basic function of NRSF and its cofactors, investigate their mechanisms, then explore how their dysfunction can cause disease states. This review will also address research on NRSF as a therapeutic target and delve into new therapeutic strategies that focus on disrupting NRSF’s ability to recruit chromatin remodelers.
Collapse
|
18
|
Sassone J, Papadimitriou E, Thomaidou D. Regenerative Approaches in Huntington's Disease: From Mechanistic Insights to Therapeutic Protocols. Front Neurosci 2018; 12:800. [PMID: 30450032 PMCID: PMC6224350 DOI: 10.3389/fnins.2018.00800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/15/2018] [Indexed: 01/10/2023] Open
Abstract
Huntington’s Disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the exon-1 of the IT15 gene encoding the protein Huntingtin. Expression of mutated Huntingtin in humans leads to dysfunction and ultimately degeneration of selected neuronal populations of the striatum and cerebral cortex. Current available HD therapy relies on drugs to treat chorea and control psychiatric symptoms, however, no therapy has been proven to slow down disease progression or prevent disease onset. Thus, although 24 years have passed since HD gene identification, HD remains a relentless progressive disease characterized by cognitive dysfunction and motor disability that leads to death of the majority of patients, on average 10–20 years after its onset. Up to now several molecular pathways have been implicated in the process of neurodegeneration involved in HD and have provided potential therapeutic targets. Based on these data, approaches currently under investigation for HD therapy aim on the one hand at getting insight into the mechanisms of disease progression in a human-based context and on the other hand at silencing mHTT expression by using antisense oligonucleotides. An innovative and still poorly investigated approach is to identify new factors that increase neurogenesis and/or induce reprogramming of endogenous neuroblasts and parenchymal astrocytes to generate new healthy neurons to replace lost ones and/or enforce neuroprotection of pre-existent striatal and cortical neurons. Here, we review studies that use human disease-in-a-dish models to recapitulate HD pathogenesis or are focused on promoting in vivo neurogenesis of endogenous striatal neuroblasts and direct neuronal reprogramming of parenchymal astrocytes, which combined with neuroprotective protocols bear the potential to re-establish brain homeostasis lost in HD.
Collapse
Affiliation(s)
- Jenny Sassone
- Vita-Salute University and San Raffaele Scientific Institute, Milan, Italy
| | | | - Dimitra Thomaidou
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
19
|
Makhouri FR, Ghasemi JB. In Silico Studies in Drug Research Against Neurodegenerative Diseases. Curr Neuropharmacol 2018; 16:664-725. [PMID: 28831921 PMCID: PMC6080098 DOI: 10.2174/1570159x15666170823095628] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 07/24/2017] [Accepted: 08/16/2017] [Indexed: 01/14/2023] Open
Abstract
Background Neurodegenerative diseases such as Alzheimer's disease (AD), amyotrophic lateral sclerosis, Parkinson's disease (PD), spinal cerebellar ataxias, and spinal and bulbar muscular atrophy are described by slow and selective degeneration of neurons and axons in the central nervous system (CNS) and constitute one of the major challenges of modern medicine. Computer-aided or in silico drug design methods have matured into powerful tools for reducing the number of ligands that should be screened in experimental assays. Methods In the present review, the authors provide a basic background about neurodegenerative diseases and in silico techniques in the drug research. Furthermore, they review the various in silico studies reported against various targets in neurodegenerative diseases, including homology modeling, molecular docking, virtual high-throughput screening, quantitative structure activity relationship (QSAR), hologram quantitative structure activity relationship (HQSAR), 3D pharmacophore mapping, proteochemometrics modeling (PCM), fingerprints, fragment-based drug discovery, Monte Carlo simulation, molecular dynamic (MD) simulation, quantum-mechanical methods for drug design, support vector machines, and machine learning approaches. Results Detailed analysis of the recently reported case studies revealed that the majority of them use a sequential combination of ligand and structure-based virtual screening techniques, with particular focus on pharmacophore models and the docking approach. Conclusion Neurodegenerative diseases have a multifactorial pathoetiological origin, so scientists have become persuaded that a multi-target therapeutic strategy aimed at the simultaneous targeting of multiple proteins (and therefore etiologies) involved in the development of a disease is recommended in future.
Collapse
Affiliation(s)
| | - Jahan B Ghasemi
- Chemistry Department, Faculty of Sciences, University of Tehran, Tehran, Iran
| |
Collapse
|
20
|
Lee JH, Kim JH, Kim S, Cho KS, Lee SB. Chromatin Changes Associated with Neuronal Maintenance and Their Pharmacological Application. Curr Neuropharmacol 2018; 16:118-125. [PMID: 28571546 PMCID: PMC5883374 DOI: 10.2174/1570159x15666170601124220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 05/03/2017] [Accepted: 05/31/2017] [Indexed: 01/07/2023] Open
Abstract
Background: The transcriptional control of neuronal specification and early development has been intensively stud-ied over the past few decades. However, relatively little is known about transcriptional programs associated with the mainte-nance of terminally differentiated neuronal cells with respect to their functions, structures, and cell type-specific identity features. Methods: Notably, largely because of the recent advances in related techniques such as next generation sequencing and chromatin immunoprecipitation sequencing, the physiological implications of system-wide regulation of gene expression through changes in chromatin states have begun to be extensively studied in various contexts and systems, including the nervous system. Results: Here, we attempt to review our current understanding of the link between chromatin changes and neuronal mainte-nance in the period of life after the completion of neuronal development. Perturbations involving chromatin changes in the system-wide transcriptional control are believed to be closely associated with diverse aspects of neuronal aging and neuro-degenerative conditions. Conclusion: In this review, we focused on heterochromatin and epigenetic dysregulation in neurodegenerative conditions as well as neuronal aging, the most important risk factor leading to neuronal degeneration, in order to highlight the close association between chromatin changes and neuronal maintenance. Lastly, we reviewed the cur-rently available and potential future applications of pharmacological control of the chromatin states associated with neuronal maintenance.
Collapse
Affiliation(s)
- Jang Ho Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029. Korea
| | - Jeong-Hoon Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Korea
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.,Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
| | - Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul 05029. Korea
| | - Sung Bae Lee
- Department of Brain & Cognitive Sciences, DGIST, Daegu 42988. Korea
| |
Collapse
|
21
|
Sertraline, chlorprothixene, and chlorpromazine characteristically interact with the REST-binding site of the corepressor mSin3, showing medulloblastoma cell growth inhibitory activities. Sci Rep 2018; 8:13763. [PMID: 30213984 PMCID: PMC6137095 DOI: 10.1038/s41598-018-31852-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022] Open
Abstract
Dysregulation of repressor-element 1 silencing transcription factor REST/NRSF is related to several neuropathies, including medulloblastoma, glioblastoma, Huntington’s disease, and neuropathic pain. Inhibitors of the interaction between the N-terminal repressor domain of REST/NRSF and the PAH1 domain of its corepressor mSin3 may ameliorate such neuropathies. In-silico screening based on the complex structure of REST/NRSF and mSin3 PAH1 yielded 52 active compounds, including approved neuropathic drugs. We investigated their binding affinity to PAH1 by NMR, and their inhibitory activity toward medulloblastoma cell growth. Interestingly, three antidepressant and antipsychotic medicines, sertraline, chlorprothixene, and chlorpromazine, were found to strongly bind to PAH1. Multivariate analysis based on NMR chemical shift changes in PAH1 residues induced by ligand binding was used to identify compound characteristics associated with cell growth inhibition. Active compounds showed a new chemo-type for inhibitors of the REST/NRSF-mSin3 interaction, raising the possibility of new therapies for neuropathies caused by dysregulation of REST/NRSF.
Collapse
|
22
|
Lu L, Marisetty A, Liu B, Kamal MM, Gumin J, Veo B, Cai Y, Kassem DH, Weng C, Maynard ME, Hood KN, Fuller GN, Pan ZZ, Cykowski MD, Dash PK, Majumder S. REST overexpression in mice causes deficits in spontaneous locomotion. Sci Rep 2018; 8:12083. [PMID: 30108242 PMCID: PMC6092433 DOI: 10.1038/s41598-018-29441-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/04/2018] [Indexed: 12/22/2022] Open
Abstract
Overexpression of REST has been implicated in brain tumors, ischemic insults, epilepsy, and movement disorders such as Huntington's disease. However, owing to the lack of a conditional REST overexpression animal model, the mechanism of action of REST overexpression in these disorders has not been established in vivo. We created a REST overexpression mouse model using the human REST (hREST) gene. Our results using these mice confirm that hREST expression parallels endogenous REST expression in embryonic mouse brains. Further analyses indicate that REST represses the dopamine receptor 2 (Drd2) gene, which encodes a critical nigrostriatal receptor involved in regulating movement, in vivo. Overexpression of REST using Drd2-Cre in adult mice results in increased REST and decreased DRD2 expression in the striatum, a major site of DRD2 expression, and phenocopies the spontaneous locomotion deficits seen upon global DRD2 deletion or specific DRD2 deletion from indirect-pathway medium spiny neurons. Thus, our studies using this mouse model not only reveal a new function of REST in regulating spontaneous locomotion but also suggest that REST overexpression in DRD2-expressing cells results in spontaneous locomotion deficits.
Collapse
Affiliation(s)
- Li Lu
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anantha Marisetty
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Neurosurgery, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bin Liu
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Epigenetics and Molecular Carcinogenesis, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mohamed Mostafa Kamal
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bethany Veo
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Pediatrics/Hematology and Oncology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - YouQing Cai
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dina Hamada Kassem
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Connie Weng
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mark E Maynard
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Kimberly N Hood
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Gregory N Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhizhong Z Pan
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Matthew D Cykowski
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Sadhan Majumder
- Departments of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
23
|
Xiang C, Zhang S, Dong X, Ma S, Cong S. Transcriptional Dysregulation and Post-translational Modifications in Polyglutamine Diseases: From Pathogenesis to Potential Therapeutic Strategies. Front Mol Neurosci 2018; 11:153. [PMID: 29867345 PMCID: PMC5962650 DOI: 10.3389/fnmol.2018.00153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
Polyglutamine (polyQ) diseases are hereditary neurodegenerative disorders caused by an abnormal expansion of a trinucleotide CAG repeat in the coding region of their respective associated genes. PolyQ diseases mainly display progressive degeneration of the brain and spinal cord. Nine polyQ diseases are known, including Huntington's disease (HD), spinal and bulbar muscular atrophy (SBMA), dentatorubral-pallidoluysian atrophy (DRPLA), and six forms of spinocerebellar ataxia (SCA). HD is the best characterized polyQ disease. Many studies have reported that transcriptional dysregulation and post-translational disruptions, which may interact with each other, are central features of polyQ diseases. Post-translational modifications, such as the acetylation of histones, are closely associated with the regulation of the transcriptional activity. A number of groups have studied the interactions between the polyQ proteins and transcription factors. Pharmacological drugs or genetic manipulations aimed at correcting the dysregulation have been confirmed to be effective in the treatment of polyQ diseases in many animal and cellular models. For example, histone deaceylase inhibitors have been demonstrated to have beneficial effects in cases of HD, SBMA, DRPLA, and SCA3. In this review, we describe the transcriptional and post-translational dysregulation in polyQ diseases with special focus on HD, and we summarize and comment on potential treatment approaches targeting disruption of transcription and post-translation processes in these diseases.
Collapse
Affiliation(s)
| | | | | | | | - Shuyan Cong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Sehgal SA, Hammad MA, Tahir RA, Akram HN, Ahmad F. Current Therapeutic Molecules and Targets in Neurodegenerative Diseases Based on in silico Drug Design. Curr Neuropharmacol 2018; 16:649-663. [PMID: 29542412 PMCID: PMC6080102 DOI: 10.2174/1570159x16666180315142137] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 01/01/2018] [Accepted: 03/02/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND As the number of elderly persons increases, neurodegenerative diseases are becoming ubiquitous. There is currently a great need for knowledge concerning management of oldage neurodegenerative diseases; the most important of which are: Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis, and Huntington's disease. OBJECTIVE To summarize the potential of computationally predicted molecules and targets against neurodegenerative diseases. METHOD Review of literature published since 1997 against neurodegenerative diseases, utilizing as keywords: in silico, Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis ALS, and Huntington's disease was conducted. RESULTS AND CONCLUSION Due to the costs associated with experimentation and current ethical law, performing experiments directly on living organisms has become much more difficult. In this scenario, in silico techniques have been successful and have become powerful tools in the search to cure disease. Researchers use the Computer Aided Drug Design pipeline which: 1) generates 3- dimensional structures of target proteins through homology modeling 2) achieves stabilization through molecular dynamics simulation, and 3) exploits molecular docking through large compound libraries. Next generation sequencing is continually producing enormous amounts of raw sequence data while neuroimaging is producing a multitude of raw image data. To solve such pressing problems, these new tools and algorithms are required. This review elaborates precise in silico tools and techniques for drug targets, active molecules, and molecular docking studies, together with future prospects and challenges concerning possible breakthroughs in Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis, and Huntington's disease.
Collapse
Affiliation(s)
- Sheikh Arslan Sehgal
- Address correspondence to this author at the State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences; Beijing, China; E-mail:
| | | | | | | | | |
Collapse
|
25
|
Patterson KP, Barry JM, Curran MM, Singh-Taylor A, Brennan G, Rismanchi N, Page M, Noam Y, Holmes GL, Baram TZ. Enduring Memory Impairments Provoked by Developmental Febrile Seizures Are Mediated by Functional and Structural Effects of Neuronal Restrictive Silencing Factor. J Neurosci 2017; 37:3799-3812. [PMID: 28275159 PMCID: PMC5394897 DOI: 10.1523/jneurosci.3748-16.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/25/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022] Open
Abstract
In a subset of children experiencing prolonged febrile seizures (FSs), the most common type of childhood seizures, cognitive outcomes are compromised. However, the underlying mechanisms are unknown. Here we identified significant, enduring spatial memory problems in male rats following experimental prolonged FS (febrile status epilepticus; eFSE). Remarkably, these deficits were abolished by transient, post hoc interference with the chromatin binding of the transcriptional repressor neuron restrictive silencing factor (NRSF or REST). This transcriptional regulator is known to contribute to neuronal differentiation during development and to programmed gene expression in mature neurons. The mechanisms of the eFSE-provoked memory problems involved complex disruption of memory-related hippocampal oscillations recorded from CA1, likely resulting in part from impairments of dendritic filtering of cortical inputs as well as abnormal synaptic function. Accordingly, eFSE provoked region-specific dendritic loss in the hippocampus, and aberrant generation of excitatory synapses in dentate gyrus granule cells. Blocking NRSF transiently after eFSE prevented granule cell dysmaturation, restored a functional balance of γ-band network oscillations, and allowed treated eFSE rats to encode and retrieve spatial memories. Together, these studies provide novel insights into developing networks that underlie memory, the mechanisms by which early-life seizures influence them, and the means to abrogate the ensuing cognitive problems.SIGNIFICANCE STATEMENT Whereas seizures have been the central focus of epilepsy research, they are commonly accompanied by cognitive problems, including memory impairments that contribute to poor quality of life. These deficits often arise before the onset of spontaneous seizures, or independent from them, yet the mechanisms involved are unclear. Here, using a rodent model of common developmental seizures that provoke epilepsy in a subset of individuals, we identify serious consequent memory problems. We uncover molecular, cellular, and circuit-level mechanisms that underlie these deficits and successfully abolish them by targeted therapeutic interventions. These findings may be important for understanding and preventing cognitive problems in individuals suffering long febrile seizures.
Collapse
Affiliation(s)
| | - Jeremy M Barry
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | | | | | - Gary Brennan
- Departments of Anatomy/Neurobiology
- Pediatrics, and
| | | | - Matias Page
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | | | - Gregory L Holmes
- Department of Neurological Sciences, University of Vermont College of Medicine, Burlington, Vermont 05405
| | - Tallie Z Baram
- Departments of Anatomy/Neurobiology,
- Pediatrics, and
- Neurology, University of California-Irvine, Irvine, California 92697-4475, and
| |
Collapse
|
26
|
Barger SW. Gene regulation and genetics in neurochemistry, past to future. J Neurochem 2016; 139 Suppl 2:24-57. [PMID: 27747882 DOI: 10.1111/jnc.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/01/2016] [Accepted: 03/30/2016] [Indexed: 12/14/2022]
Abstract
Ask any neuroscientist to name the most profound discoveries in the field in the past 60 years, and at or near the top of the list will be a phenomenon or technique related to genes and their expression. Indeed, our understanding of genetics and gene regulation has ushered in whole new systems of knowledge and new empirical approaches, many of which could not have even been imagined prior to the molecular biology boon of recent decades. Neurochemistry, in the classic sense, intersects with these concepts in the manifestation of neuropeptides, obviously dependent upon the central dogma (the established rules by which DNA sequence is eventually converted into protein primary structure) not only for their conformation but also for their levels and locales of expression. But, expanding these considerations to non-peptide neurotransmitters illustrates how gene regulatory events impact neurochemistry in a much broader sense, extending beyond the neurochemicals that translate electrical signals into chemical ones in the synapse, to also include every aspect of neural development, structure, function, and pathology. From the beginning, the mutability - yet relative stability - of genes and their expression patterns were recognized as potential substrates for some of the most intriguing phenomena in neurobiology - those instances of plasticity required for learning and memory. Near-heretical speculation was offered in the idea that perhaps the very sequence of the genome was altered to encode memories. A fascinating component of the intervening progress includes evidence that the central dogma is not nearly as rigid and consistent as we once thought. And this mutability extends to the potential to manipulate that code for both experimental and clinical purposes. Astonishing progress has been made in the molecular biology of neurochemistry during the 60 years since this journal debuted. Many of the gains in conceptual understanding have been driven by methodological progress, from automated high-throughput sequencing instruments to recombinant-DNA vectors that can convey color-coded genetic modifications in the chromosomes of live adult animals. This review covers the highlights of these advances, both theoretical and technological, along with a brief window into the promising science ahead. This article is part of the 60th Anniversary special issue.
Collapse
Affiliation(s)
- Steven W Barger
- Department of Geriatrics, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA.
| |
Collapse
|
27
|
Regulation of neural gene transcription by optogenetic inhibition of the RE1-silencing transcription factor. Proc Natl Acad Sci U S A 2015; 113:E91-100. [PMID: 26699507 DOI: 10.1073/pnas.1507355112] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Optogenetics provides new ways to activate gene transcription; however, no attempts have been made as yet to modulate mammalian transcription factors. We report the light-mediated regulation of the repressor element 1 (RE1)-silencing transcription factor (REST), a master regulator of neural genes. To tune REST activity, we selected two protein domains that impair REST-DNA binding or recruitment of the cofactor mSin3a. Computational modeling guided the fusion of the inhibitory domains to the light-sensitive Avena sativa light-oxygen-voltage-sensing (LOV) 2-phototrophin 1 (AsLOV2). By expressing AsLOV2 chimeras in Neuro2a cells, we achieved light-dependent modulation of REST target genes that was associated with an improved neural differentiation. In primary neurons, light-mediated REST inhibition increased Na(+)-channel 1.2 and brain-derived neurotrophic factor transcription and boosted Na(+) currents and neuronal firing. This optogenetic approach allows the coordinated expression of a cluster of genes impinging on neuronal activity, providing a tool for studying neuronal physiology and correcting gene expression changes taking place in brain diseases.
Collapse
|
28
|
Schiffer D, Caldera V, Mellai M, Conforti P, Cattaneo E, Zuccato C. Repressor element-1 silencing transcription factor (REST) is present in human control and Huntington's disease neurones. Neuropathol Appl Neurobiol 2015; 40:899-910. [PMID: 24634989 DOI: 10.1111/nan.12137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 03/12/2014] [Indexed: 01/25/2023]
Abstract
AIMS The repressor element-1 silencing transcription factor/neurone-restrictive silencer factor (REST/NRSF) is a master regulator of neuronal gene expression. REST/NRSF functions by recruiting other cofactors to genomic loci that contain the repressor element 1/neurone restrictive silencer element (RE1/NRSE) binding motif. In brain, demonstration of REST protein presence in neurones has remained controversial. However, RE1/NRSE containing neuronal genes are actively modulated and REST dysregulation is implicated in Huntington's disease (HD). We aimed to investigate REST distribution in autopsy brain from control and HD patients. METHODS Brain tissues from six controls and six HD cases (Vonsattel grade 3 and 4) were investigated using immunohistochemical analysis. RESULTS REST was present in neurones and glial cells of the cortex, caudate nucleus, hippocampus and cerebellum. REST labelling was mainly cytoplasmic in neurones while preferential nuclear staining of REST was found in glial cells. We also found that REST and huntingtin (HTT) colocalize in human neurones. Low levels of cytoplasmic REST were detected in neurones of the HD cortex and caudate but no direct relationship between decreased neuronal REST expression and disease grade was observed. CONCLUSIONS These data support the notion of REST presence in human brain neurones and glial cells and indicate the importance of developing compounds able to restore REST-regulated transcription of neuronal genes in HD.
Collapse
Affiliation(s)
- Davide Schiffer
- Neuro-Bio-Oncology Research Center, Policlinico di Monza Foundation, Vercelli; Consorzio per le Neuroscienze, University of Pavia, Pavia
| | | | | | | | | | | |
Collapse
|
29
|
Qureshi IA, Mehler MF. Epigenetics and therapeutic targets mediating neuroprotection. Brain Res 2015; 1628:265-272. [PMID: 26236020 DOI: 10.1016/j.brainres.2015.07.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/14/2015] [Accepted: 07/22/2015] [Indexed: 11/29/2022]
Abstract
The rapidly evolving science of epigenetics is transforming our understanding of the nervous system in health and disease and holds great promise for the development of novel diagnostic and therapeutic approaches targeting neurological diseases. Increasing evidence suggests that epigenetic factors and mechanisms serve as important mediators of the pathogenic processes that lead to irrevocable neural injury and of countervailing homeostatic and regenerative responses. Epigenetics is, therefore, of considerable translational significance to the field of neuroprotection. In this brief review, we provide an overview of epigenetic mechanisms and highlight the emerging roles played by epigenetic processes in neural cell dysfunction and death and in resultant neuroprotective responses. This article is part of a Special Issue entitled SI: Neuroprotection.
Collapse
Affiliation(s)
- Irfan A Qureshi
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Mark F Mehler
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Ruth L. and David S. Gottesman Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Center for Epigenomics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
30
|
The Transcription Repressor REST in Adult Neurons: Physiology, Pathology, and Diseases. eNeuro 2015; 2:eN-REV-0010-15. [PMID: 26465007 PMCID: PMC4596026 DOI: 10.1523/eneuro.0010-15.2015] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/11/2015] [Accepted: 06/18/2015] [Indexed: 12/21/2022] Open
Abstract
REST [RE1-silencing transcription factor (also called neuron-restrictive silencer factor)] is known to repress thousands of possible target genes, many of which are neuron specific. To date, REST repression has been investigated mostly in stem cells and differentiating neurons. Current evidence demonstrates its importance in adult neurons as well. Low levels of REST, which are acquired during differentiation, govern the expression of specific neuronal phenotypes. REST-dependent genes encode important targets, including transcription factors, transmitter release proteins, voltage-dependent and receptor channels, and signaling proteins. Additional neuronal properties depend on miRNAs expressed reciprocally to REST and on specific splicing factors. In adult neurons, REST levels are not always low. Increases occur during aging in healthy humans. Moreover, extensive evidence demonstrates that prolonged stimulation with various agents induces REST increases, which are associated with the repression of neuron-specific genes with appropriate, intermediate REST binding affinity. Whether neuronal increases in REST are protective or detrimental remains a subject of debate. Examples of CA1 hippocampal neuron protection upon depolarization, and of neurodegeneration upon glutamate treatment and hypoxia have been reported. REST participation in psychiatric and neurological diseases has been shown, especially in Alzheimer’s disease and Huntington’s disease, as well as epilepsy. Distinct, complex roles of the repressor in these different diseases have emerged. In conclusion, REST is certainly very important in a large number of conditions. We suggest that the conflicting results reported for the role of REST in physiology, pathology, and disease depend on its complex, direct, and indirect actions on many gene targets and on the diverse approaches used during the investigations.
Collapse
|
31
|
Ieraci A, Mallei A, Musazzi L, Popoli M. Physical exercise and acute restraint stress differentially modulate hippocampal brain-derived neurotrophic factor transcripts and epigenetic mechanisms in mice. Hippocampus 2015; 25:1380-92. [PMID: 25820928 DOI: 10.1002/hipo.22458] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2015] [Indexed: 12/15/2022]
Abstract
Physical exercise and stressful experiences have been shown to exert opposite effects on behavioral functions and brain plasticity, partly by involving the action of brain-derived neurotrophic factor (BDNF). Although epigenetic modifications are known to play a pivotal role in the regulation of the different BDNF transcripts, it is poorly understood whether epigenetic mechanisms are also implied in the BDNF modulation induced by physical exercise and stress. Here, we show that total BDNF mRNA levels and BDNF transcripts 1, 2, 3, 4, 6, and 7 were reduced immediately after acute restraint stress (RS) in the hippocampus of mice, and returned to control levels 24 h after the stress session. On the contrary, exercise increased BDNF mRNA expression and counteracted the stress-induced decrease of BDNF transcripts. Physical exercise-induced up-regulation of BDNF transcripts was accounted for by increase in histone H3 acetylated levels at specific BDNF promoters, whereas the histone H3 trimethylated lysine 27 and dimethylated lysine 9 levels were unaffected. Acute RS did not change the levels of acetylated and methylated histone H3 at the BDNF promoters. Furthermore, we found that physical exercise and RS were able to differentially modulate the histone deacetylases mRNA levels. Finally, we report that a single treatment with histone deacetylase inhibitors, prior to acute stress exposure, prevented the down-regulation of total BDNF and BDNF transcripts 1, 2, 3, and 6, partially reproducing the effect of physical exercise. Overall, these results suggest that physical exercise and stress are able to differentially modulate the expression of BDNF transcripts by possible different epigenetic mechanisms.
Collapse
Affiliation(s)
- Alessandro Ieraci
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università degli Studi di Milano, Milano, Italy
| | - Alessandra Mallei
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università degli Studi di Milano, Milano, Italy
| | - Laura Musazzi
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università degli Studi di Milano, Milano, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
32
|
Maes T, Mascaró C, Ortega A, Lunardi S, Ciceri F, Somervaille TCP, Buesa C. KDM1 histone lysine demethylases as targets for treatments of oncological and neurodegenerative disease. Epigenomics 2015; 7:609-26. [PMID: 26111032 DOI: 10.2217/epi.15.9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Histone methylation and demethylation are important processes associated with the regulation of gene transcription, and alterations in histone methylation status have been linked to a large number of human diseases. Initially thought to be an irreversible process, histone methylation is now known to be reversed by two families of proteins containing over 30 members that act to remove methyl groups from specific lysine residues present in the tails of histone H3 and histone H4. A rapidly growing number of reports have implicated the FAD-dependent lysine specific demethylase (KDM1) family in cancer, and several small-molecule inhibitors are in development for the treatment of cancer. An additional role has emerged for KDM1 in brain function, offering additional opportunities for the development of novel therapeutic strategies in neurodegenerative disease. A decade after the identification of KDM1A as a histone demethylase, the first selective inhibitors have now reached the clinic.
Collapse
Affiliation(s)
- Tamara Maes
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornella de Llobregat, Barcelona, España
| | - Cristina Mascaró
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornella de Llobregat, Barcelona, España
| | - Alberto Ortega
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornella de Llobregat, Barcelona, España
| | - Serena Lunardi
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornella de Llobregat, Barcelona, España
| | - Filippo Ciceri
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornella de Llobregat, Barcelona, España
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, Manchester, M20 4BX, UK
| | - Carlos Buesa
- Oryzon Genomics S.A., Carrer Sant Ferran 74, 08940 Cornella de Llobregat, Barcelona, España
| |
Collapse
|
33
|
NRSF: an Angel or a Devil in Neurogenesis and Neurological Diseases. J Mol Neurosci 2014; 56:131-44. [DOI: 10.1007/s12031-014-0474-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/18/2014] [Indexed: 12/12/2022]
|
34
|
Wild EJ, Tabrizi SJ. Targets for future clinical trials in Huntington's disease: what's in the pipeline? Mov Disord 2014; 29:1434-45. [PMID: 25155142 PMCID: PMC4265300 DOI: 10.1002/mds.26007] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 01/08/2023] Open
Abstract
The known genetic cause of Huntington's disease (HD) has fueled considerable progress in understanding its pathobiology and the development of therapeutic approaches aimed at correcting specific changes linked to the causative mutation. Among the most promising is reducing expression of mutant huntingtin protein (mHTT) with RNA interference or antisense oligonucleotides; human trials are now being planned. Zinc-finger transcriptional repression is another innovative method to reduce mHTT expression. Modulation of mHTT phosphorylation, chaperone upregulation, and autophagy enhancement represent attempts to alter cellular homeostasis to favor removal of mHTT. Inhibition of histone deacetylases (HDACs) remains of interest; recent work affirms HDAC4 as a target but questions the assumed centrality of its catalytic activity in HD. Phosphodiesterase inhibition, aimed at restoring synaptic function, has progressed rapidly to human trials. Deranged cellular signaling provides several tractable targets, but specificity and complexity are challenges. Restoring neurotrophic support in HD remains a key potential therapeutic approach. with several approaches being pursued, including brain-derived neurotrophic factor (BDNF) mimesis through tyrosine receptor kinase B (TrkB) agonism and monoclonal antibodies. An increasing understanding of the role of glial cells in HD has led to several new therapeutic avenues, including kynurenine monooxygenase inhibition, immunomodulation by laquinimod, CB2 agonism, and others. The complex metabolic derangements in HD remain under study, but no clear therapeutic strategy has yet emerged. We conclude that many exciting therapeutics are progressing through the development pipeline, and combining a better understanding of HD biology in human patients, with concerted medicinal chemistry efforts, will be crucial for bringing about an era of effective therapies.
Collapse
Affiliation(s)
- Edward J Wild
- Department of Neurodegenerative Disease, UCL Institute of Neurology, National Hospital for Neurology & NeurosurgeryQueen Square, London, WC1N 3BG, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, National Hospital for Neurology & NeurosurgeryQueen Square, London, WC1N 3BG, UK
| |
Collapse
|
35
|
Valor LM. Transcription, epigenetics and ameliorative strategies in Huntington's Disease: a genome-wide perspective. Mol Neurobiol 2014; 51:406-23. [PMID: 24788684 PMCID: PMC4309905 DOI: 10.1007/s12035-014-8715-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/11/2014] [Indexed: 12/18/2022]
Abstract
Transcriptional dysregulation in Huntington’s disease (HD) is an early event that shapes the brain transcriptome by both the depletion and ectopic activation of gene products that eventually affect survival and neuronal functions. Disruption in the activity of gene expression regulators, such as transcription factors, chromatin-remodeling proteins, and noncoding RNAs, accounts for the expression changes observed in multiple animal and cellular models of HD and in samples from patients. Here, I review the recent advances in the study of HD transcriptional dysregulation and its causes to finally discuss the possible implications in ameliorative strategies from a genome-wide perspective. To date, the use of genome-wide approaches, predominantly based on microarray platforms, has been successful in providing an extensive catalog of differentially regulated genes, including biomarkers aimed at monitoring the progress of the pathology. Although still incipient, the introduction of combined next-generation sequencing techniques is enhancing our comprehension of the mechanisms underlying altered transcriptional dysregulation in HD by providing the first genomic landscapes associated with epigenetics and the occupancy of transcription factors. In addition, the use of genome-wide approaches is becoming more and more necessary to evaluate the efficacy and safety of ameliorative strategies and to identify novel mechanisms of amelioration that may help in the improvement of current preclinical therapeutics. Finally, the major conclusions obtained from HD transcriptomics studies have the potential to be extrapolated to other neurodegenerative disorders.
Collapse
Affiliation(s)
- Luis M Valor
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas, Av. Santiago Ramón y Cajal s/n, Sant Joan d'Alacant, 03550, Alicante, Spain,
| |
Collapse
|
36
|
Kumar A, Vaish M, Ratan RR. Transcriptional dysregulation in Huntington's disease: a failure of adaptive transcriptional homeostasis. Drug Discov Today 2014; 19:956-62. [PMID: 24662036 DOI: 10.1016/j.drudis.2014.03.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/14/2014] [Indexed: 01/02/2023]
Abstract
Huntington's disease (HD) is a signature polyglutamine disorder. An enduring theory of HD pathogenesis has involved dysregulation of transcription. Indeed, transcriptional regulatory proteins can be modulated to overcome cardinal features of HD-modeled mice, and efforts to move these into human studies are ongoing. Here, we discuss a unifying hypothesis emerging from these studies, which is that HD represents the pathological disruption of evolutionarily conserved adaptive gene programs to counteract oxidative stress, mitochondrial dysfunction and accumulation of misfolded proteins. Transcriptional dyshomeostasis of adaptive genes is further exacerbated by repression of genes involved in normal synaptic activity or growth factor signaling.
Collapse
Affiliation(s)
- Amit Kumar
- Burke Medical Research Institute, White Plains, NY 10065, USA; Brain and Mind Research Institute, Weill Medical College, Cornell University, White Plains, NY 10605, USA.
| | - Manisha Vaish
- Burke Medical Research Institute, White Plains, NY 10065, USA
| | - Rajiv R Ratan
- Burke Medical Research Institute, White Plains, NY 10065, USA; Brain and Mind Research Institute, Weill Medical College, Cornell University, White Plains, NY 10605, USA; Department of Neurology, Weill Medical College, Cornell University, White Plains, NY 10605, USA.
| |
Collapse
|
37
|
Ruffoni A, Ferri N, Bernini SK, Ricci C, Corsini A, Maffucci I, Clerici F, Contini A. 2-Amino-3-(phenylsulfanyl)norbornane-2-carboxylate: An Appealing Scaffold for the Design of Rac1–Tiam1 Protein–Protein Interaction Inhibitors. J Med Chem 2014; 57:2953-62. [DOI: 10.1021/jm401924s] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Alessandro Ruffoni
- Dipartimento
di Scienze Farmaceutiche—Sezione di Chimica Generale e Organica
“Alessandro Marchesini”, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Nicola Ferri
- Dipartimento
di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
- Multimedica IRCCS, Milan, Italy
| | - Sergio K. Bernini
- Dipartimento
di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Chiara Ricci
- Dipartimento
di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Alberto Corsini
- Dipartimento
di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Irene Maffucci
- Dipartimento
di Scienze Farmaceutiche—Sezione di Chimica Generale e Organica
“Alessandro Marchesini”, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Francesca Clerici
- Dipartimento
di Scienze Farmaceutiche—Sezione di Chimica Generale e Organica
“Alessandro Marchesini”, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Alessandro Contini
- Dipartimento
di Scienze Farmaceutiche—Sezione di Chimica Generale e Organica
“Alessandro Marchesini”, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| |
Collapse
|
38
|
Abstract
Changes in the level and activity of brain-derived neurotrophic factor (BDNF) have been described in a number of neurodegenerative disorders since early 1990s. However, only in Huntington disease (HD) gain- and loss-of-function experiments have mechanistically linked these abnormalities with the genetic defect.In this chapter we will describe how huntingtin protein, whose mutation causes HD, is involved in the physiological control of BDNF synthesis and transport in neurons and how both processes are simultaneously disrupted in HD. We will describe the underlying molecular mechanisms and discuss pre-clinical data concerning the impact of the experimental manipulation of BDNF levels on HD progression. These studies have revealed that a major loss of BDNF protein in the brain of HD patients may contribute to the clinical manifestations of the disease. The experimental strategies under investigation to increase brain BDNF levels in animal models of HD will also be described, with a view to ultimately improving the clinical treatment of this condition.
Collapse
Affiliation(s)
- Chiara Zuccato
- Department of Biosciences and Centre for Stem cell Research, Università degli Studi di Milano, Via Viotti 3/5, 20133, Milan, Italy,
| | | |
Collapse
|
39
|
Mrzljak L, Munoz-Sanjuan I. Therapeutic Strategies for Huntington's Disease. Curr Top Behav Neurosci 2013; 22:161-201. [PMID: 24277342 DOI: 10.1007/7854_2013_250] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Huntington's disease (HD) is a devastating autosomal dominant neurodegenerative disease, caused by expansion of the CAG repeat in the huntingtin (HTT) gene and characterized pathologically by the loss of pyramidal neurons in several cortical areas, of striatal medium spiny neurons, and of hypothalamic neurons. Clinically, a distinguishing feature of the disease is uncontrolled involuntary movements (chorea, dyskensias) accompanied by progressive cognitive, motor, and psychiatric impairment. This review focuses on the current state of therapeutic development for the treatment of HD, including the preclinical and clinical development of small molecules and molecular therapies.
Collapse
|