1
|
Li Y, Izhar T, Kanekiyo T. HDAC3 as an Emerging Therapeutic Target for Alzheimer's Disease and other Neurological Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04866-w. [PMID: 40126601 DOI: 10.1007/s12035-025-04866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the aged population. Histone acetylation is a major epigenetic mechanism linked to memory formation and cognitive function. Histone deacetylases (HDACs) are responsible for the deacetylation of lysine residues in histone proteins. Although pan-HDAC inhibitors are effective in ameliorating AD phenotypes in preclinical models, they are associated with potential unfavorable adverse effects and barely translated into clinical trials. Therefore, the development of novel HDAC inhibitors with a well isoform-selectivity has been desired in AD drug discovery. Among various HDAC isoforms, HDAC3 is highly expressed in neurons and exhibits detrimental effects on synaptic plasticity and cognitive function. Moreover, HDAC3 provokes neuroinflammation and neurotoxicity and contributes to AD pathogenesis. In this review, we highlight HDAC3 as an attractive therapeutic target for disease-modifying therapy in AD. In addition, we discuss the therapeutic potential of HDAC3 inhibitors in other neurological disorders.
Collapse
Affiliation(s)
- Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Taha Izhar
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
2
|
Ding Y. Histone deacetylases: the critical enzymes for microglial activation involved in neuropathic pain. Front Pharmacol 2025; 16:1515787. [PMID: 40115267 PMCID: PMC11922887 DOI: 10.3389/fphar.2025.1515787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/23/2025] Open
Abstract
Neuropathic pain is a common health problem in clinical practice that can be caused by many different factors, including infection, ischemia, trauma, diabetes mellitus, nerve compression, autoimmune disorders, cancer, trigeminal neuralgia, and abuse of certain drugs. This type of pain can persistently affect patients for a long time, even after the rehabilitation of their damaged tissues. Researchers have identified the crucial role of microglial activation in the pathogenesis of neuropathic pain. Furthermore, emerging evidence has shown that the expression and/or activities of different histone deacetylases (HDACs) can modulate microglial function and neuropathic pain. In this review, we will summarize and discuss the functions and mechanisms of HDACs in microglial activation and neuropathic pain development. Additionally, we will also list the emerging HDAC inhibitors or activators that may contribute to therapeutic advancement in alleviating neuropathic pain.
Collapse
Affiliation(s)
- Yi Ding
- Department of Anesthesiology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Rosete C, Ciernia AV. The Two Faces of HDAC3: Neuroinflammation in Disease and Neuroprotection in Recovery. Epigenomics 2024; 16:1373-1388. [PMID: 39513228 PMCID: PMC11728336 DOI: 10.1080/17501911.2024.2419357] [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: 08/21/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024] Open
Abstract
Histone deacetylase 3 (HDAC3) is a critical regulator of gene expression, influencing a variety of cellular processes in the central nervous system. As such, dysfunction of this enzyme may serve as a key driver in the pathophysiology of various neuropsychiatric disorders and neurodegenerative diseases. HDAC3 plays a crucial role in regulating neuroinflammation, and is now widely recognized as a major contributor to neurological conditions, as well as in promoting neuroprotective recovery following brain injury, hemorrhage and stroke. Emerging evidence suggests that pharmacological inhibition of HDAC3 can mitigate behavioral and neuroimmune deficits in various brain diseases and disorders, offering a promising therapeutic strategy. Understanding HDAC3 in the healthy brain lays the necessary foundation to define and resolve its dysfunction in a disease state. This review explores the mechanisms of HDAC3 in various cell types and its involvement in disease pathology, emphasizing the potential of HDAC3 inhibition to address neuroimmune, gene expression and behavioral deficits in a range of neurodegenerative and neuropsychiatric conditions.
Collapse
Affiliation(s)
- Cal Rosete
- Djavad Mowafaghian Centre for Brain Health, Vancouver, V6T 1Z3, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 2A1, Canada
| | - Annie Vogel Ciernia
- Djavad Mowafaghian Centre for Brain Health, Vancouver, V6T 1Z3, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, V6T 2A1, Canada
| |
Collapse
|
4
|
Haage V, Tuddenham JF, Bautista A, White CC, Garcia F, Patel R, Comandante-Lou N, Marshe V, Soni RK, Sims PA, Menon V, Sproul AA, De Jager PL. HDAC Inhibitors recapitulate Human Disease-Associated Microglia Signatures in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617544. [PMID: 39416157 PMCID: PMC11482930 DOI: 10.1101/2024.10.11.617544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Disease-associated microglia (DAM), initially described in mouse models of neurodegenerative diseases, have been classified into two related states; starting from a TREM2-independent DAM1 state to a TREM2 dependent state termed DAM2, with each state being characterized by the expression of specific marker genes1. Recently, single-cell (sc)RNA-Seq studies have reported the existence of DAMs in humans2-6; however, whether DAMs play beneficial or detrimental roles in the context of neurodegeneration is still under debate7,8. Here, we present a pharmacological approach to mimic human DAM in vitro by exposing different human microglia models to selected histone deacetylase (HDAC) inhibitors. We also provide an initial functional characterization of our model system, showing a specific increase of amyloid beta phagocytosis along with a reduction of MCP-1 secretion. Additionally, we report an increase in MITF expression, a transcription factor previously described to drive expression towards the DAM phenotype. We further identify CADM1, LIPA and SCIN as DAM-marker genes shared across various proposed DAM signatures and in our model systems. Overall, our strategy for targeted microglial polarization bears great potential to further explore human DAM function and biology.
Collapse
Affiliation(s)
- Verena Haage
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - John F. Tuddenham
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Bautista
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Charles C. White
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Frankie Garcia
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Ronak Patel
- Department of Pathology and Cell Biology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Natacha Comandante-Lou
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Victoria Marshe
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York, NY
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Andrew A. Sproul
- Department of Pathology and Cell Biology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center
| |
Collapse
|
5
|
Xie Q, Li Q, Fang H, Zhang R, Tang H, Chen L. Gut-Derived Short-Chain Fatty Acids and Macrophage Modulation: Exploring Therapeutic Potentials in Pulmonary Fungal Infections. Clin Rev Allergy Immunol 2024; 66:316-327. [PMID: 38965168 DOI: 10.1007/s12016-024-08999-z] [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] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Short-chain fatty acids (SCFAs), such as acetate, propionate, and butyrate, modulate immune cell functions, particularly macrophages. This review explores the potential therapeutic applications of SCFAs in pulmonary fungal infections, a critical concern due to their high mortality rates and antifungal resistance. SCFAs enhance macrophage functions by promoting phagosome-lysosome fusion, increasing reactive oxygen species production, and balancing cytokine responses. Pulmonary fungal infections, caused by pathogens like Aspergillus fumigatus, are prevalent in immunocompromised patients, including those with diabetes, chronic obstructive pulmonary disease, and those on high-dose corticosteroids. SCFAs have shown promise in improving macrophage function in these contexts. However, the application of SCFAs must be balanced against potential side effects, including gut microbiota disruption and metabolic disorders. Further research is needed to optimize SCFA therapy for managing pulmonary fungal infections.
Collapse
Affiliation(s)
- Qian Xie
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Section, 1st Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Qinhui Li
- Medical Services Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Section, 1st Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Hong Fang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Section, 1st Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Rong Zhang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Section, 1st Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Huan Tang
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Section, 1st Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Lin Chen
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West 2nd Section, 1st Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
6
|
Davis N, Taylor B, Abelleira-Hervas L, Karimian-Marnani N, Aleksynas R, Syed N, Di Giovanni S, Palmisano I, Sastre M. Histone deacetylase-3 regulates the expression of the amyloid precursor protein and its inhibition promotes neuroregenerative pathways in Alzheimer's disease models. FASEB J 2024; 38:e23659. [PMID: 38733301 DOI: 10.1096/fj.202301762rr] [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: 08/30/2023] [Revised: 04/04/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024]
Abstract
HDAC3 inhibition has been shown to improve memory and reduce amyloid-β (Aβ) in Alzheimer's disease (AD) models, but the underlying mechanisms are unclear. We investigated the molecular effects of HDAC3 inhibition on AD pathology, using in vitro and ex vivo models of AD, based on our finding that HDAC3 expression is increased in AD brains. For this purpose, N2a mouse neuroblastoma cells as well as organotypic brain cultures (OBCSs) of 5XFAD and wild-type mice were incubated with various concentrations of the HDAC3 selective inhibitor RGFP966 (0.1-10 μM) for 24 h. Treatment with RGFP966 or HDAC3 knockdown in N2a cells was associated with an increase on amyloid precursor protein (APP) and mRNA expressions, without alterations in Aβ42 secretion. In vitro chromatin immunoprecipitation analysis revealed enriched HDAC3 binding at APP promoter regions. The increase in APP expression was also detected in OBCSs from 5XFAD mice incubated with 1 μM RGFP966, without changes in Aβ. In addition, HDAC3 inhibition resulted in a reduction of activated Iba-1-positive microglia and astrocytes in 5XFAD slices, which was not observed in OBCSs from wild-type mice. mRNA sequencing analysis revealed that HDAC3 inhibition modulated neuronal regenerative pathways related to neurogenesis, differentiation, axonogenesis, and dendritic spine density in OBCSs. Our findings highlight the complexity and diversity of the effects of HDAC3 inhibition on AD models and suggest that HDAC3 may have multiple roles in the regulation of APP expression and processing, as well as in the modulation of neuroinflammatory and neuroprotective genes.
Collapse
Affiliation(s)
- Nicola Davis
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Ben Taylor
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | | | | | - Robertas Aleksynas
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Nelofer Syed
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Simone Di Giovanni
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Ilaria Palmisano
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
- Department of Neuroscience, The Ohio State University, Columbus, Ohio, USA
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus, Ohio, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
7
|
Clayton BLL, Kristell JD, Allan KC, Cohn EF, Karl M, Jerome AD, Garrison E, Maeno-Hikichi Y, Sturno AM, Kerr A, Shick HE, Sepeda JA, Freundt EC, Sas AR, Segal BM, Miller RH, Tesar PJ. A phenotypic screening platform for identifying chemical modulators of astrocyte reactivity. Nat Neurosci 2024; 27:656-665. [PMID: 38378993 PMCID: PMC11034956 DOI: 10.1038/s41593-024-01580-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/15/2024] [Indexed: 02/22/2024]
Abstract
Disease, injury and aging induce pathological reactive astrocyte states that contribute to neurodegeneration. Modulating reactive astrocytes therefore represent an attractive therapeutic strategy. Here we describe the development of an astrocyte phenotypic screening platform for identifying chemical modulators of astrocyte reactivity. Leveraging this platform for chemical screening, we identify histone deacetylase 3 (HDAC3) inhibitors as effective suppressors of pathological astrocyte reactivity. We demonstrate that HDAC3 inhibition reduces molecular and functional characteristics of reactive astrocytes in vitro. Transcriptional and chromatin mapping studies show that HDAC3 inhibition disarms pathological astrocyte gene expression and function while promoting the expression of genes associated with beneficial astrocytes. Administration of RGFP966, a small molecule HDAC3 inhibitor, blocks reactive astrocyte formation and promotes neuroprotection in vivo in mice. Collectively, these results establish a platform for discovering modulators of reactive astrocyte states, inform the mechanisms that control astrocyte reactivity and demonstrate the therapeutic benefits of modulating astrocyte reactivity for neurodegenerative diseases.
Collapse
Affiliation(s)
- Benjamin L L Clayton
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - James D Kristell
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kevin C Allan
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Erin F Cohn
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Molly Karl
- Department of Anatomy and Cell Biology, George Washington University School of Medicine, Washington, DC, USA
| | - Andrew D Jerome
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Eric Garrison
- Department of Anatomy and Cell Biology, George Washington University School of Medicine, Washington, DC, USA
| | - Yuka Maeno-Hikichi
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Annalise M Sturno
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alexis Kerr
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - H Elizabeth Shick
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jesse A Sepeda
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Eric C Freundt
- Department of Biology, The University of Tampa, Tampa, FL, USA
| | - Andrew R Sas
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Benjamin M Segal
- Department of Neurology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| | - Robert H Miller
- Department of Anatomy and Cell Biology, George Washington University School of Medicine, Washington, DC, USA
| | - Paul J Tesar
- Institute for Glial Sciences, Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
8
|
Meleady L, Towriss M, Kim J, Bacarac V, Dang V, Rowland ME, Ciernia AV. Histone deacetylase 3 regulates microglial function through histone deacetylation. Epigenetics 2023; 18:2241008. [PMID: 37506371 PMCID: PMC10392760 DOI: 10.1080/15592294.2023.2241008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
As the primary innate immune cells of the brain, microglia respond to damage and disease through pro-inflammatory release of cytokines and neuroinflammatory molecules. Histone acetylation is an activating transcriptional mark that regulates inflammatory gene expression. Inhibition of histone deacetylase 3 (Hdac3) has been utilized in pre-clinical models of depression, stroke, and spinal cord injury to improve recovery following injury, but the molecular mechanisms underlying Hdac3's regulation of inflammatory gene expression in microglia is not well understood. To address this lack of knowledge, we examined how pharmacological inhibition of Hdac3 in an immortalized microglial cell line (BV2) impacted histone acetylation and gene expression of pro- and anti-inflammatory genes in response to immune challenge with lipopolysaccharide (LPS). Flow cytometry and cleavage under tags & release using nuclease (CUT & RUN) revealed that Hdac3 inhibition increases global and promoter-specific histone acetylation, resulting in the release of gene repression at baseline and enhanced responses to LPS. Hdac3 inhibition enhanced neuroprotective functions of microglia in response to LPS through reduced nitric oxide release and increased phagocytosis. The findings suggest Hdac3 serves as a regulator of microglial inflammation, and that inhibition of Hdac3 facilitates the microglial response to inflammation and its subsequent clearing of debris or damaged cells. Together, this work provides new mechanistic insights into therapeutic applications of Hdac3 inhibition which mediate reduced neuroinflammatory insults through microglial response.
Collapse
Affiliation(s)
- Laura Meleady
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Morgan Towriss
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jennifer Kim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Vince Bacarac
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Vivien Dang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Megan E. Rowland
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
9
|
Lanka G, Begum D, Banerjee S, Adhikari N, P Y, Ghosh B. Pharmacophore-based virtual screening, 3D QSAR, Docking, ADMET, and MD simulation studies: An in silico perspective for the identification of new potential HDAC3 inhibitors. Comput Biol Med 2023; 166:107481. [PMID: 37741229 DOI: 10.1016/j.compbiomed.2023.107481] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/19/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023]
Abstract
Histone deacetylase 3 (HDAC3) is an epigenetic regulator that involves gene expression, apoptosis, and cell cycle progression, and the overexpression of HDAC3 is accountable for several cancers, neurodegeneracy, and many other diseases. Therefore, HDAC3 emerged as a promising drug target for the novel drug design. Here, we carried out the pharmacophore modeling using 50 benzamide-based HDAC3 selective inhibitors and utilized it for PHASE ligand screening to retrieve the hits with similar pharmacophore features. The dataset inhibitors of best hypotheses used to build the 3D QSAR model and the generated 3D QSAR model resulted in good PLS statistics with a regression coefficient (R2) of 0.89, predictive coefficient (Q2) of 0.88, and Pearson-R factor of 0.94 indicating its excellent predictive ability. The hits retrieved from pharmacophore-based virtual screening were subjected to docking against HDAC3 for the identification of potential inhibitors. A total of 10 hitsM1 to M10 were ranked using their scoring functions and further subject to lead optimization. The Prime MM/GBSA, AutoDock binding free energies, and ADMET studies were implemented for the selection of lead candidates. The four ligand molecules M1, M2, M3, and M4 were identified as potential leads against HDAC3 after lead optimization. The top two leads M1 and M2 were subjected to MD simulations for their stability evaluation with HDAC3. The newly designed leads M11 and M12 were identified as HDAC3 potential inhibitors from MD simulations studies. Therefore, the outcomes of the present study could provide insights into the discovery of new potential HDAC3 inhibitors with improved selectivity and activity against a variety of cancers and neurodegenerative diseases.
Collapse
Affiliation(s)
- Goverdhan Lanka
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Shamirpet, Hyderabad, 500078, India
| | - Darakhshan Begum
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Shamirpet, Hyderabad, 500078, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, P. O. Box 17020, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, P. O. Box 17020, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Yogeeswari P
- Computer Aided Drug Design Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani Hyderabad Campus, Shamirpet, Hyderabad, 500078, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Shamirpet, Hyderabad, 500078, India.
| |
Collapse
|
10
|
Kuboyama T, Hotta K, Asanuma M, Ge YW, Toume K, Yamazaki T, Komatsu K. Quality assessment of Rheum species cultivated in Japan by focusing on M2 polarization of microglia. J Nat Med 2023; 77:699-711. [PMID: 37347410 DOI: 10.1007/s11418-023-01710-0] [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: 09/30/2022] [Accepted: 05/22/2023] [Indexed: 06/23/2023]
Abstract
In traditional Japanese medicine, Rhei Rhizoma is used as a purgative, blood stasis-resolving and antipsychotic drug. The latter two properties are possibly related to anti-inflammatory effects. Microglia regulate inflammation in the central nervous system. M1 microglia induce inflammation, while M2 microglia inhibit inflammation and show neurotrophic effects. This study investigated the effects from water extracts of roots of cultivated Rheum species in Nagano Prefecture, Japan (strain C, a related strain to a Japanese cultivar, 'Shinshu-Daio'; and strain 29, a Chinese strain) and 3 kinds of Rhei Rhizoma available in the Japanese market, and also examined their constituents on the polarization of cultured microglia. All extracts significantly decreased M1 microglia, and strains C and 29 significantly increased M2 microglia. Furthermore, the extracts of both strains significantly increased the M2/M1 ratio. Among the constituents of Rhei Rhizoma, ( +)-catechin (2), resveratrol 4'-O-β-D-(6″-O-galloyl) glucopyranoside (5), isolindleyin (8), and physcion (15) significantly increased the M2/M1 ratio. The contents of the constituents in water extract of each strain were quantified using HPLC. The extracts of strains C and 29 contained relatively large amounts of 2 and 5; and 2, 8, and 15, respectively. This study showed the water extracts of roots of cultivated Rheum strains in Japan had the effects of M2 polarization of microglia, suggesting that these strains become the candidate to develop anti-inflammatory Rhei Rhizoma. Moreover, the suitable chemical composition to possess anti-inflammatory activity in the brain was clarified for the future development of new type of Rhei Rhizoma.
Collapse
Affiliation(s)
- Tomoharu Kuboyama
- Laboratory of Pharmacognosy, Daiichi University of Pharmacy, 22-1 Tamagawa-Cho, Minami-Ku, Fukuoka, 815-8511, Japan.
| | - Kenichiro Hotta
- Section of Pharmacognosy, Division of Medicinal Resources, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Mai Asanuma
- Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, Kodo, Kyotanabe, Kyoto, 610-0395, Japan
| | - Yue-Wei Ge
- School of Traditional Chinese Medicine, Guangzhou Higher Education Mega Center, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou, China
| | - Kazufumi Toume
- Section of Pharmacognosy, Division of Medicinal Resources, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takuma Yamazaki
- Pharmaceutical Affairs Division, Health and Welfare, Department of Nagano Prefecture, 692-2 Habashita, Minami-Nagano, Nagano, 380-8570, Japan
| | - Katsuko Komatsu
- Section of Pharmacognosy, Division of Medicinal Resources, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| |
Collapse
|
11
|
Chen S, Ye J, Wu G, Shi J, Li X, Chen X, Wu W. Histone Deacetylase 3 Inhibition Ameliorates Microglia-Mediated Neuro-Inflammation Via the SIRT1/Nrf2 Pathway After Traumatic Spinal Cord Injury. Neurorehabil Neural Repair 2023; 37:503-518. [PMID: 37503724 DOI: 10.1177/15459683231183716] [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] [Indexed: 07/29/2023]
Abstract
BACKGROUND Microglial-induced inflammation plays a crucial role in the pathophysiological process of nervous system injury, however, still lacks effective therapeutic agents. Previously, we discovered that the inhibition of histone deacetylase 3 (HDAC3) exerts anti-inflammatory effects after traumatic spinal cord injury (SCI), whereas little is known about its underlying mechanism. Therefore, the present study aimed to explore the effects and potential mechanisms of HDAC3 on neuroinflammation and microglial function. METHODS Rats were randomized into 4 groups: sham group, SCI group, SCI + vehicle group, and SCI + RGF966 group. To examine the effect of HDAC3 on neurological deficit after SCI, we gathered data using the Basso Beattie Bresnahan locomotion scale, the inclined plane test, the blood-spinal cord barrier, junction protein expression, and Nissl staining. We also evaluated microglial activation and inflammatory factor levels. Immunofluorescence analysis, immunohistochemical analysis, western blotting, and quantitative real-time polymerase chain reaction were performed to examine the regulation of the Sirtuin 1 (SIRT1)/nuclear factor-erythroid 2-related factor 2 (Nrf2) pathway. RESULTS The results showed that HDAC3 inhibition significantly ameliorated Basso-Beattie-Bresnahan (BBB) permeability, brain edema, and improved neurological functions and junction protein levels. Additionally, HDAC3 inhibition significantly inhibited microglial activation, thereby reducing the levels of SCI-induced pro-inflammatory factors. Moreover, HDAC3 inhibition dramatically enhanced the expression of SIRT1 and increased both Nrf2 nuclear accumulation and transcriptional activity, thereby increasing downstream heme oxygenase-1 and NAD(P)H quinone oxidoreductase 1 expression. CONCLUSIONS The results of this study suggest that HDAC3 inhibition protects the spinal cord from injury following SCI by inhibiting SCI-induced microglial activation and the subsequent inflammatory response via SIRT1/Nrf2 signaling pathway, highlighting HDAC3 as a potential therapeutic target for the treatment of SCI.
Collapse
Affiliation(s)
- Shoubo Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jingfang Ye
- Department of Nursing Faculty, Quanzhou Medical College, Quanzhou, Fujian Province, China
| | - Guozhong Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jinnan Shi
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province, China
| | - Xiang Li
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province, China
| | - Wenhua Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
12
|
Shafqat A, Albalkhi I, Magableh HM, Saleh T, Alkattan K, Yaqinuddin A. Tackling the glial scar in spinal cord regeneration: new discoveries and future directions. Front Cell Neurosci 2023; 17:1180825. [PMID: 37293626 PMCID: PMC10244598 DOI: 10.3389/fncel.2023.1180825] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Axonal regeneration and functional recovery are poor after spinal cord injury (SCI), typified by the formation of an injury scar. While this scar was traditionally believed to be primarily responsible for axonal regeneration failure, current knowledge takes a more holistic approach that considers the intrinsic growth capacity of axons. Targeting the SCI scar has also not reproducibly yielded nearly the same efficacy in animal models compared to these neuron-directed approaches. These results suggest that the major reason behind central nervous system (CNS) regeneration failure is not the injury scar but a failure to stimulate axon growth adequately. These findings raise questions about whether targeting neuroinflammation and glial scarring still constitute viable translational avenues. We provide a comprehensive review of the dual role of neuroinflammation and scarring after SCI and how future research can produce therapeutic strategies targeting the hurdles to axonal regeneration posed by these processes without compromising neuroprotection.
Collapse
|
13
|
Ghosh D, Singh G, Mishra P, Singh A, Kumar A, Sinha N. Alteration in mitochondrial dynamics promotes the proinflammatory response of microglia and is involved in cerebellar dysfunction of young and aged mice following LPS exposure. Neurosci Lett 2023; 807:137262. [PMID: 37116576 DOI: 10.1016/j.neulet.2023.137262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023]
Abstract
Cerebellar dysfunction is implicated in impaired motor coordination and balance, thus disturbing the dynamics of sensorimotor integration. Neuroinflammation and aging could be prominent contributors to cerebellar aberration. Additionally, changes in mitochondrial dynamics may precede microglia activation in several chronic neurodegenerative diseases; however, the underlying mechanism remains largely unknown.Here using LPS (1 mg/kg i.p. for four consecutive days) stimulation in both young (3 months old) and aged (12 months old) mice, followed by molecular analysis on the 21st day, we have explored the correlation between aging and mitochondrial dynamic alteration in the backdrop of chronic neuroinflammation. Following LPS stimulation, we observed microglia activation and subsequent elevation in proinflammatory cytokines (M1; TNF-α, IFN-γ) with NLRP3 activationand a concomitant reduction in the expression of anti-inflammatory markers (M2; YM1, TGF-β1) in the cerebellar tissue of aged mice compared with the young LPS and aged controls. Remarkably, senescence (p21, p27, p53) and epigenetic (HDAC2) markers were found upregulated in the cerebellum tissue of the aged LPS group, suggesting their crucial role in LPS-induced cerebellar deficit. Further, we demonstrated alteration in the antagonistic forces of mitochondrial fusion and fission with increased expression of the mitochondrial fission-related gene [FIS1] and decreased fusion-related genes [MFN1 and MFN2]. We noted increased mtDNA copy number, microglia activation, and inflammatory response of IL1β and IFN-γ post-chronic neuroinflammation in aged LPS group. Our results suggest that the crosstalk between mitochondrial dynamics and altered microglial activation paradigm in chronic neuroinflammatory conditions may be the key to understanding the cerebellar molecular mechanism.
Collapse
Affiliation(s)
- Devlina Ghosh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomti Nagar Extension, Lucknow 226028, India; Centre of Biomedical Research, SGPGIMS-Campus, Raibareli Road, Lucknow 226014, India.
| | - Gajendra Singh
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Prabhaker Mishra
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Raebareli Road, Lucknow 226 014, Uttar Pradesh, India
| | - Aditi Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Gomti Nagar Extension, Lucknow 226028, India
| | - Alok Kumar
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Neeraj Sinha
- Centre of Biomedical Research, SGPGIMS-Campus, Raibareli Road, Lucknow 226014, India.
| |
Collapse
|
14
|
He R, Liu B, Geng B, Li N, Geng Q. The role of HDAC3 and its inhibitors in regulation of oxidative stress and chronic diseases. Cell Death Discov 2023; 9:131. [PMID: 37072432 PMCID: PMC10113195 DOI: 10.1038/s41420-023-01399-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 03/01/2023] [Accepted: 03/06/2023] [Indexed: 04/20/2023] Open
Abstract
HDAC3 is a specific and crucial member of the HDAC family. It is required for embryonic growth, development, and physiological function. The regulation of oxidative stress is an important factor in intracellular homeostasis and signal transduction. Currently, HDAC3 has been found to regulate several oxidative stress-related processes and molecules dependent on its deacetylase and non-enzymatic activities. In this review, we comprehensively summarize the knowledge of the relationship of HDAC3 with mitochondria function and metabolism, ROS-produced enzymes, antioxidant enzymes, and oxidative stress-associated transcription factors. We also discuss the role of HDAC3 and its inhibitors in some chronic cardiovascular, kidney, and neurodegenerative diseases. Due to the simultaneous existence of enzyme activity and non-enzyme activity, HDAC3 and the development of its selective inhibitors still need further exploration in the future.
Collapse
Affiliation(s)
- Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Boxin Geng
- School of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
Cheng Y, Song H, Ming GL, Weng YL. Epigenetic and epitranscriptomic regulation of axon regeneration. Mol Psychiatry 2023; 28:1440-1450. [PMID: 36922674 PMCID: PMC10650481 DOI: 10.1038/s41380-023-02028-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023]
Abstract
Effective axonal regeneration in the adult mammalian nervous system requires coordination of elevated intrinsic growth capacity and decreased responses to the inhibitory environment. Intrinsic regenerative capacity largely depends on the gene regulatory network and protein translation machinery. A failure to activate these pathways upon injury is underlying a lack of robust axon regeneration in the mature mammalian central nervous system. Epigenetics and epitranscriptomics are key regulatory mechanisms that shape gene expression and protein translation. Here, we provide an overview of different types of modifications on DNA, histones, and RNA, underpinning the regenerative competence of axons in the mature mammalian peripheral and central nervous systems. We highlight other non-neuronal cells and their epigenetic changes in determining the microenvironment for tissue repair and axon regeneration. We also address advancements of single-cell technology in charting transcriptomic and epigenetic landscapes that may further facilitate the mechanistic understanding of differential regenerative capacity in neuronal subtypes. Finally, as epigenetic and epitranscriptomic processes are commonly affected by brain injuries and psychiatric disorders, understanding their alterations upon brain injury would provide unprecedented mechanistic insights into etiology of injury-associated-psychiatric disorders and facilitate the development of therapeutic interventions to restore brain function.
Collapse
Affiliation(s)
- Yating Cheng
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, 77030, USA
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Yi-Lan Weng
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, 77030, USA.
- Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Kim HN, McCrea MR, Li S. Advances in molecular therapies for targeting pathophysiology in spinal cord injury. Expert Opin Ther Targets 2023; 27:171-187. [PMID: 37017093 PMCID: PMC10148912 DOI: 10.1080/14728222.2023.2194532] [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: 01/23/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Spinal cord injury (SCI) affects 25,000-50,000 people around the world each year and there is no cure for SCI patients currently. The primary injury damages spinal cord tissues and secondary injury mechanisms, including ischemia, apoptosis, inflammation, and astrogliosis, further exacerbate the lesions to the spinal cord. Recently, researchers have designed various therapeutic approaches for SCI by targeting its major cellular or molecular pathophysiology. AREAS COVERED Some strategies have shown promise in repairing injured spinal cord for functional recoveries, such as administering neuroprotective reagents, targeting specific genes to promote robust axon regeneration of disconnected spinal fiber tracts, targeting epigenetic factors to enhance cell survival and neural repair, and facilitating neuronal relay pathways and neuroplasticity for restoration of function after SCI. This review focuses on the major advances in preclinical molecular therapies for SCI reported in recent years. EXPERT OPINION Recent progress in developing novel and effective repairing strategies for SCI is encouraging, but many challenges remain for future design of effective treatments, including developing highly effective neuroprotectants for early interventions, stimulating robust neuronal regeneration with functional synaptic reconnections among disconnected neurons, maximizing the recovery of lost neural functions with combination strategies, and translating the most promising therapies into human use.
Collapse
Affiliation(s)
- Ha Neui Kim
- Shriners Hospitals Pediatric Research Center, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Madeline R. McCrea
- Shriners Hospitals Pediatric Research Center, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
17
|
Tang H, Gu Y, Jiang L, Zheng G, Pan Z, Jiang X. The role of immune cells and associated immunological factors in the immune response to spinal cord injury. Front Immunol 2023; 13:1070540. [PMID: 36685599 PMCID: PMC9849245 DOI: 10.3389/fimmu.2022.1070540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological condition prevalent worldwide. Where the pathological mechanisms underlying SCI are concerned, we can distinguish between primary injury caused by initial mechanical damage and secondary injury characterized by a series of biological responses, such as vascular dysfunction, oxidative stress, neurotransmitter toxicity, lipid peroxidation, and immune-inflammatory response. Secondary injury causes further tissue loss and dysfunction, and the immune response appears to be the key molecular mechanism affecting injured tissue regeneration and functional recovery from SCI. Immune response after SCI involves the activation of different immune cells and the production of immunity-associated chemicals. With the development of new biological technologies, such as transcriptomics, the heterogeneity of immune cells and chemicals can be classified with greater precision. In this review, we focus on the current understanding of the heterogeneity of these immune components and the roles they play in SCI, including reactive astrogliosis and glial scar formation, neutrophil migration, macrophage transformation, resident microglia activation and proliferation, and the humoral immunity mediated by T and B cells. We also summarize findings from clinical trials of immunomodulatory therapies for SCI and briefly review promising therapeutic drugs currently being researched.
Collapse
Affiliation(s)
- Huaguo Tang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yuanjie Gu
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Lei Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Gang Zheng
- Department of Neurosurgery, The Central Hospital Affiliated to Shaoxing University, Jiaxing, China
| | - Zhuoer Pan
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xiugui Jiang
- Department of Hand and Foot Surgery, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
18
|
Sun W, Wen M, Liu M, Wang Q, Liu Q, Li L, Siebert HC, Loers G, Zhang R, Zhang N. Effect of β-hydroxybutyrate on behavioral alterations, molecular and morphological changes in CNS of multiple sclerosis mouse model. Front Aging Neurosci 2022; 14:1075161. [PMID: 36533180 PMCID: PMC9752847 DOI: 10.3389/fnagi.2022.1075161] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/14/2022] [Indexed: 09/29/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of central nervous system (CNS). Aging is the most significant risk factor for the progression of MS. Dietary modulation (such as ketogenic diet) and caloric restriction, can increase ketone bodies, especially β-hydroxybutyrate (BHB). Increased BHB has been reported to prevent or improve age-related disease. The present studies were performed to understand the therapeutic effect and potential mechanisms of exogenous BHB in cuprizone (CPZ)-induced demyelinating model. In this study, a continuous 35 days CPZ mouse model with or without BHB was established. The changes of behavior function, pathological hallmarks of CPZ, and intracellular signal pathways in mice were detected by Open feld test, Morris water maze, RT-PCR, immuno-histochemistry, and western blot. The results showed that BHB treatment improved behavioral performance, prevented myelin loss, decreased the activation of astrocyte as well as microglia, and up-regulated the neurotrophin brain-derived neurotrophic factor in both the corpus callosum and hippocampus. Meanwhile, BHB treatment increased the number of MCT1+ cells and APC+ oligodendrocytes. Furthermore, the treatment decreased the expression of HDAC3, PARP1, AIF and TRPA1 which is related to oligodendrocyte (OL) apoptosis in the corpus callosum, accompanied by increased expression of TrkB. This leads to an increased density of doublecortin (DCX)+ neuronal precursor cells and mature NeuN+ neuronal cells in the hippocampus. As a result, BHB treatment effectively promotes the generation of PDGF-Ra+ (oligodendrocyte precursor cells, OPCs), Sox2+ cells and GFAP+ (astrocytes), and decreased the production of GFAP+ TRAP1+ cells, and Oligo2+ TRAP1+ cells in the corpus callosum of mouse brain. Thus, our results demonstrate that BHB treatment efficiently supports OPC differentiation and decreases the OLs apoptosis in CPZ-intoxicated mice, partly by down-regulating the expression of TRPA1 and PARP, which is associated with the inhibition of the p38-MAPK/JNK/JUN pathway and the activation of ERK1/2, PI3K/AKT/mTOR signaling, supporting BHB treatment adjunctive nutritional therapy for the treatment of chronic demyelinating diseases, such as multiple sclerosis (MS).
Collapse
Affiliation(s)
- Wei Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Quiqin Liu
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Lanjie Li
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Hans-Christian Siebert
- Schauenburgerstr, RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Kiel University, Kiel, Germany
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
- Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
19
|
Ashok A, Pooranawattanakul S, Tai WL, Cho KS, Utheim TP, Cestari DM, Chen DF. Epigenetic Regulation of Optic Nerve Development, Protection, and Repair. Int J Mol Sci 2022; 23:8927. [PMID: 36012190 PMCID: PMC9408916 DOI: 10.3390/ijms23168927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Epigenetic factors are known to influence tissue development, functionality, and their response to pathophysiology. This review will focus on different types of epigenetic regulators and their associated molecular apparatus that affect the optic nerve. A comprehensive understanding of epigenetic regulation in optic nerve development and homeostasis will help us unravel novel molecular pathways and pave the way to design blueprints for effective therapeutics to address optic nerve protection, repair, and regeneration.
Collapse
Affiliation(s)
- Ajay Ashok
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sarita Pooranawattanakul
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Wai Lydia Tai
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tor P. Utheim
- Department of Medical Biochemistry, Oslo University Hospital, 0372 Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, 0372 Oslo, Norway
| | - Dean M. Cestari
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Dong Feng Chen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
20
|
Zhao Y, Mu H, Huang Y, Li S, Wang Y, Stetler RA, Bennett MVL, Dixon CE, Chen J, Shi Y. Microglia-specific deletion of histone deacetylase 3 promotes inflammation resolution, white matter integrity, and functional recovery in a mouse model of traumatic brain injury. J Neuroinflammation 2022; 19:201. [PMID: 35933343 PMCID: PMC9357327 DOI: 10.1186/s12974-022-02563-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Histone deacetylases (HDACs) are believed to exacerbate traumatic brain injury (TBI) based on studies using pan-HDAC inhibitors. However, the HDAC isoform responsible for the detrimental effects and the cell types involved remain unknown, which may hinder the development of specific targeting strategies that boost therapeutic efficacy while minimizing side effects. Microglia are important mediators of post-TBI neuroinflammation and critically impact TBI outcome. HDAC3 was reported to be essential to the inflammatory program of in vitro cultured macrophages, but its role in microglia and in the post-TBI brain has not been investigated in vivo. METHODS We generated HDAC3LoxP mice and crossed them with CX3CR1CreER mice, enabling in vivo conditional deletion of HDAC3. Microglia-specific HDAC3 knockout (HDAC3 miKO) was induced in CX3CR1CreER:HDAC3LoxP mice with 5 days of tamoxifen treatment followed by a 30-day development interval. The effects of HDAC3 miKO on microglial phenotype and neuroinflammation were examined 3-5 days after TBI induced by controlled cortical impact. Neurological deficits and the integrity of white matter were assessed for 6 weeks after TBI by neurobehavioral tests, immunohistochemistry, electron microscopy, and electrophysiology. RESULTS HDAC3 miKO mice harbored specific deletion of HDAC3 in microglia but not in peripheral monocytes. HDAC3 miKO reduced the number of microglia by 26%, but did not alter the inflammation level in the homeostatic brain. After TBI, proinflammatory microglial responses and brain inflammation were markedly alleviated by HDAC3 miKO, whereas the infiltration of blood immune cells was unchanged, suggesting a primary effect of HDAC3 miKO on modulating microglial phenotype. Importantly, HDAC3 miKO was sufficient to facilitate functional recovery for 6 weeks after TBI. TBI-induced injury to axons and myelin was ameliorated, and signal conduction by white matter fiber tracts was significantly enhanced in HDAC3 miKO mice. CONCLUSION Using a novel microglia-specific conditional knockout mouse model, we delineated for the first time the role of microglial HDAC3 after TBI in vivo. HDAC3 miKO not only reduced proinflammatory microglial responses, but also elicited long-lasting improvement of white matter integrity and functional recovery after TBI. Microglial HDAC3 is therefore a promising therapeutic target to improve long-term outcomes after TBI.
Collapse
Affiliation(s)
- Yongfang Zhao
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Hongfeng Mu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yichen Huang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sicheng Li
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Yangfan Wang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - C Edward Dixon
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
21
|
Zhou X, Qiao B. Inhibition of HDAC3 and ATXN3 by miR-25 prevents neuronal loss and ameliorates neurological recovery in cerebral stroke experimental rats. J Physiol Biochem 2022; 78:139-149. [PMID: 35025075 DOI: 10.1007/s13105-021-00848-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/14/2021] [Indexed: 11/26/2022]
Abstract
HDAC3 plays important role in regulating memory and plasticity of neurons. We studied the role of miR-25 against HDAC3-induced neuronal injury in acute ischemic stroke. Subjects reported for acute stroke were included in the study. The rat model of middle cerebral artery occlusion was developed and received miR-25 agomir and antagomir treatments via intra-cerebroventricular injection. The brain tissues were processed and neuronal cells were isolated and submitted to oxygen glucose derivation-mediated injury. mRNA levels were studied by RT-PCR and protein levels by ELISA method. TUNEL and nuclear protein staining was done to find the ischemic area. Behavioral studies were carried out by Morris water maze test and beam balance test. Results suggested a significant increase in plasma miR-25 levels observed in acute ischemic stroke subjects. The levels of miR-25 were increased in the tissues of infarcted area of brain tissues of rats. However, the expression of miR-25 decreased in neuronal cells but increased in supernatant post-oxygen glucose deprivation. The treatment of miR-25 agomir decreased the infarct volume and apoptosis of neurons in MCAO rats, and it also improved the plasticity of neurons and axons, down-regulated ATXN3 and HDAC3 levels, and improved acetyl-H3K9 expression. In vitro outcomes suggested that miR-25 inhibited HDAC3 neurons and modulated the oxygen glucose deprivation injury in neurons. The treatment of RGFP966 increased the acetyl-H3K9 levels and prevented the miR-25 antagomir-induced injury. The study suggested miR-25 as an important predicting biomarker in acute ischemia injury. KEY POINTS: • HDAC3 is key regulator for neuronal health. • miR-25 was overexpressed in acute ischemic stroke condition. • miR-25 inhibits loss of neurons and provides neuronal recovery in animal model of stroke via inhibiting HDAC3 and ATXN3.
Collapse
Affiliation(s)
- Xiaomei Zhou
- Department of Neurology, The First People's Hospital of Lianyungang, Lianyungang, 222002, Jiangsu, China
| | - Benyu Qiao
- Department of Neurology, Lianyungang Hospital of Traditional Chinese Medicine, No.160 Chaoyang Road, Haizhou District, Lianyungang, 222001, Jiangsu, China.
| |
Collapse
|
22
|
Salvador AFM, Kipnis J. Immune response after central nervous system injury. Semin Immunol 2022; 59:101629. [PMID: 35753867 DOI: 10.1016/j.smim.2022.101629] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Traumatic injuries of the central nervous system (CNS) affect millions of people worldwide, and they can lead to severely damaging consequences such as permanent disability and paralysis. Multiple factors can obstruct recovery after CNS injury. One of the most significant is the progressive neuronal death that follows the initial mechanical impact, leading to the loss of undamaged cells via a process termed secondary neurodegeneration. Efforts to define treatments that limit the spread of damage, while important, have been largely ineffectual owing to gaps in the mechanistic understanding that underlies the persisting neuronal cell death. Inflammation, with its influx of immune cells that occurs shortly after injury, has been associated with secondary neurodegeneration. However, the role of the immune system after CNS injury is far more complex. Studies have indicated that the immune response after CNS injury is detrimental, owing to immune cell-produced factors (e.g., pro-inflammatory cytokines, free radicals, neurotoxic glutamate) that worsen tissue damage. Our lab and others have also demonstrated the beneficial immune response that occurs after CNS injury, with the release of growth factors such as brain-derived growth factor (BDNF) and interleukin (IL-10) and the clearance of apoptotic and myelin debris by immune cells1-4. In this review, we first discuss the multifaceted roles of the immune system after CNS injury. We then speculate on how advancements in single-cell RNA technologies can dramatically change our understanding of the immune response, how the spinal cord meninges serve as an important site for hosting immunological processes critical for recovery, and how the origin of peripherally recruited immune cells impacts their function in the injured CNS.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
Depleted histone deacetylase 3 or restored microRNA-19b-1-5p facilitates recovery of spinal cord injury via inactivating JAK2/STAT3 signaling pathway. Genomics 2021; 114:110262. [PMID: 34971719 DOI: 10.1016/j.ygeno.2021.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 11/23/2022]
Abstract
We intended to discuss the influence of histone deacetylase 3 (HDAC3) on spinal cord injury (SCI) by regulating microRNA-19b-1-5p (miR-19b-1-5p) and janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway. In a rat model, the role of HDAC3 and miR-19b-1-5p in SCI was identified through detecting motor function, serum inflammation, pathological damage, cell apoptosis and GFAP expression. Also, by measuring GFAP expression and migration of spinal cord astrocytes, the effects of HDAC3 and miR-19b-1-5p in SCI were identified in vitro. Restoration of miR-19b-1-5p or depletion of HDAC3 attenuated motor function, inflammation, pathological damage and apoptosis, and reduced GFAP expression in the spinal cord tissue of SCI rats. Up-regulating miR-19b-1-5p or down-regulating HDAC3 decreased migration and GFAP expression of injured astrocytes. Our study presents that down-regulated HDAC3 can facilitate the recovery of SCI via inhibiting the activation of JAK2/STAT3 pathway by up-regulating miR-19b-1-5p.
Collapse
|
24
|
Adhikari N, Jha T, Ghosh B. Dissecting Histone Deacetylase 3 in Multiple Disease Conditions: Selective Inhibition as a Promising Therapeutic Strategy. J Med Chem 2021; 64:8827-8869. [PMID: 34161101 DOI: 10.1021/acs.jmedchem.0c01676] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The acetylation of histone and non-histone proteins has been implicated in several disease states. Modulation of such epigenetic modifications has therefore made histone deacetylases (HDACs) important drug targets. HDAC3, among various class I HDACs, has been signified as a potentially validated target in multiple diseases, namely, cancer, neurodegenerative diseases, diabetes, obesity, cardiovascular disorders, autoimmune diseases, inflammatory diseases, parasitic infections, and HIV. However, only a handful of HDAC3-selective inhibitors have been reported in spite of continuous efforts in design and development of HDAC3-selective inhibitors. In this Perspective, the roles of HDAC3 in various diseases as well as numerous potent and HDAC3-selective inhibitors have been discussed in detail. It will surely open up a new vista in the discovery of newer, more effective, and more selective HDAC3 inhibitors.
Collapse
Affiliation(s)
- Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, P.O. Box 17020, Kolkata, 700032 West Bengal, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, P.O. Box 17020, Kolkata, 700032 West Bengal, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Shamirpet, Hyderabad 500078, India
| |
Collapse
|
25
|
Wu CH, Tsai YC, Tsai TH, Kuo KL, Su YF, Chang CH, Lin CL. Valproic Acid Reduces Vasospasm through Modulation of Akt Phosphorylation and Attenuates Neuronal Apoptosis in Subarachnoid Hemorrhage Rats. Int J Mol Sci 2021; 22:ijms22115975. [PMID: 34205883 PMCID: PMC8198375 DOI: 10.3390/ijms22115975] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/27/2021] [Accepted: 05/31/2021] [Indexed: 11/16/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is a devastating emergent event associated with high mortality and morbidity. Survivors usually experience functional neurological sequelae caused by vasospasm-related delayed ischemia. In this study, male Sprague-Dawley rats were randomly assigned to five groups: sham (non-SAH) group, SAH group, and three groups with SAH treated with different doses of valproic acid (VPA) (10, 20, 40 mg/kg, once-daily, for 7 days). The severity of vasospasm was determined by the ratio of cross-sectional areas to intima-media thickness of the basilar arteries (BA) on the seventh day after SAH. The BA showed decreased expression of phospho-Akt proteins. The dentate gyrus showed increased expression of cleaved caspase-3 and Bax proteins and decreased expression of Bcl-2, phospho-ERK 1/2, phospho-Akt and acetyl-histone H3 proteins. The incidence of SAH-induced vasospasm was significantly lower in the SAH group treated with VPA 40 mg/kg (p < 0.001). Moreover, all groups treated with VPA showed reversal of the above-mentioned protein expression in BA and the dentate gyrus. Treatment with VPA upregulated histone H3 acetylation and conferred anti-vasospastic and neuro-protective effects by enhancing Akt and/or ERK phosphorylation. This study demonstrated that VPA could alleviate delayed cerebral vasospasm induced neuro-apoptosis after SAH.
Collapse
Affiliation(s)
- Chieh-Hsin Wu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (C.-H.W.); (T.-H.T.); (K.-L.K.); (Y.-F.S.); (C.-H.C.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Yi-Cheng Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Tai-Hsin Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (C.-H.W.); (T.-H.T.); (K.-L.K.); (Y.-F.S.); (C.-H.C.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Keng-Liang Kuo
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (C.-H.W.); (T.-H.T.); (K.-L.K.); (Y.-F.S.); (C.-H.C.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Yu-Feng Su
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (C.-H.W.); (T.-H.T.); (K.-L.K.); (Y.-F.S.); (C.-H.C.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
| | - Chih-Hui Chang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (C.-H.W.); (T.-H.T.); (K.-L.K.); (Y.-F.S.); (C.-H.C.)
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
| | - Chih-Lung Lin
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (C.-H.W.); (T.-H.T.); (K.-L.K.); (Y.-F.S.); (C.-H.C.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80756, Taiwan;
- Correspondence: ; Tel.: +886-7-3121101
| |
Collapse
|
26
|
INTACT vs. FANS for Cell-Type-Specific Nuclei Sorting: A Comprehensive Qualitative and Quantitative Comparison. Int J Mol Sci 2021; 22:ijms22105335. [PMID: 34069481 PMCID: PMC8159132 DOI: 10.3390/ijms22105335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/11/2021] [Accepted: 05/14/2021] [Indexed: 12/15/2022] Open
Abstract
Increasing numbers of studies seek to characterize the different cellular sub-populations present in mammalian tissues. The techniques “Isolation of Nuclei Tagged in Specific Cell Types” (INTACT) or “Fluorescence-Activated Nuclei Sorting” (FANS) are frequently used for isolating nuclei of specific cellular subtypes. These nuclei are then used for molecular characterization of the cellular sub-populations. Despite the increasing popularity of both techniques, little is known about their isolation efficiency, advantages, and disadvantages or downstream molecular effects. In our study, we compared the physical and molecular attributes of sfGFP+ nuclei isolated by the two methods—INTACT and FANS—from the neocortices of Arc-CreERT2 × CAG-Sun1/sfGFP animals. We identified differences in efficiency of sfGFP+ nuclei isolation, nuclear size as well as transcriptional (RNA-seq) and chromatin accessibility (ATAC-seq) states. Therefore, our study presents a comprehensive comparison between the two widely used nuclei sorting techniques, identifying the advantages and disadvantages for both INTACT and FANS. Our conclusions are summarized in a table to guide researchers in selecting the most suitable methodology for their individual experimental design.
Collapse
|
27
|
Wahane S, Zhou X, Zhou X, Guo L, Friedl MS, Kluge M, Ramakrishnan A, Shen L, Friedel CC, Zhang B, Friedel RH, Zou H. Diversified transcriptional responses of myeloid and glial cells in spinal cord injury shaped by HDAC3 activity. SCIENCE ADVANCES 2021; 7:eabd8811. [PMID: 33637528 PMCID: PMC7909890 DOI: 10.1126/sciadv.abd8811] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/12/2021] [Indexed: 05/02/2023]
Abstract
The innate immune response influences neural repair after spinal cord injury (SCI). Here, we combined myeloid-specific transcriptomics and single-cell RNA sequencing to uncover not only a common core but also temporally distinct gene programs in injury-activated microglia and macrophages (IAM). Intriguingly, we detected a wide range of microglial cell states even in healthy spinal cord. Upon injury, IAM progressively acquired overall reparative, yet diversified transcriptional profiles, each comprising four transcriptional subtypes with specialized tasks. Notably, IAM have both distinct and common gene signatures as compared to neurodegeneration-associated microglia, both engaging phagocytosis, autophagy, and TyroBP pathways. We also identified an immediate response microglia subtype serving as a source population for microglial transformation and a proliferative subtype controlled by the epigenetic regulator histone deacetylase 3 (HDAC3). Together, our data unveil diversification of myeloid and glial subtypes in SCI and an extensive influence of HDAC3, which may be exploited to enhance functional recovery.
Collapse
Affiliation(s)
- Shalaka Wahane
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiang Zhou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lei Guo
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marie-Sophie Friedl
- Institut für Informatik, Ludwig-Maximilians-Universität München, 80333 Munich, Germany
| | - Michael Kluge
- Institut für Informatik, Ludwig-Maximilians-Universität München, 80333 Munich, Germany
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Caroline C Friedel
- Institut für Informatik, Ludwig-Maximilians-Universität München, 80333 Munich, Germany
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
28
|
Kuboyama T, Kominato S, Nagumo M, Tohda C. Recovery from spinal cord injury via M2 microglial polarization induced by Polygalae Radix. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 82:153452. [PMID: 33418139 DOI: 10.1016/j.phymed.2020.153452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Spinal cord injury (SCI) is a refractory neurodegenerative disease caused by inflammation. M1 microglia induce inflammation, whereas M2 suppress inflammation and exhibit neuroprotective effects. Following SCI, M1 cells are more predominant than M2 cells, and hence, increasing the predominance of M2 microglia may improve SCI. PURPOSE We aimed to evaluate the active constituents of herbal medicine that induce M2 predominance and to investigate their effects using SCI model mice. METHODS Herbal medicine inducing M2 were screened using cultured microglia. After orally administering the active herbal medicine, Polygalae Radix (PR), to SCI model mice, motor function was evaluated. Compounds in the spinal cord following treatment were assessed using liquid chromatography-mass spectrometry. The effects of compounds detected in the spinal cord were investigated in cultured microglia. RESULTS PR induced M2 predominance in cultured microglia, improved motor function in SCI model mice, and showed a tendency to increase M2 microglia and protect against axonal degeneration in the inured spinal cord. Sibiricose A5 and 3,6'-disinapoyl sucrose were identified as active constituents in PR. CONCLUSION PR may be a promising candidate for the treatment of SCI by inducing M2 predominance.
Collapse
Affiliation(s)
- Tomoharu Kuboyama
- Laboratory of Pharmacognosy, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka, 815-8511, Japan; Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama, 930-0194, Japan.
| | - Seiya Kominato
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama, 930-0194, Japan
| | - Misaki Nagumo
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama, 930-0194, Japan
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama, 930-0194, Japan
| |
Collapse
|
29
|
Al Mamun A, Monalisa I, Tul Kubra K, Akter A, Akter J, Sarker T, Munir F, Wu Y, Jia C, Afrin Taniya M, Xiao J. Advances in immunotherapy for the treatment of spinal cord injury. Immunobiology 2020; 226:152033. [PMID: 33321368 DOI: 10.1016/j.imbio.2020.152033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/19/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a leading cause of morbidity and disability in the world. Over the past few decades, the exact molecular mechanisms describing secondary, persistent injuries, as well as primary and transient injuries, have attracted massive attention to the clinicians and researchers. Recent investigations have distinctly shown the critical roles of innate and adaptive immune responses in regulating sterile neuroinflammation and functional outcomes after SCI. In past years, some promising advances in immunotherapeutic options have efficaciously been identified for the treatment of SCI. In our narrative review, we have mainly focused on the new therapeutic strategies such as the maturation and apoptosis of immune cells by several agents, mesenchymal stem cells (MSCs) as well as multi-factor combination therapy, which have recently provided novel ideas and prospects for the future treatment of SCI. This article also illustrates the latest progress in clarifying the potential roles of innate and adaptive immune responses in SCI, the progression and specification of prospective immunotherapy and outstanding issues in the area.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China
| | - Ilma Monalisa
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Khadija Tul Kubra
- Department of Pharmacy, University of Development Alternative, Dhaka 1209, Bangladesh
| | - Afroza Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Jaheda Akter
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chattogram-4318, Chittagong, Bangladesh
| | - Tamanna Sarker
- Department of Pharmacy, University of Asia Pacific, Dhaka 1205, Bangladesh
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035 Zhejiang Province, China
| | - Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027 Zhejiang Province, China
| | - Masuma Afrin Taniya
- Department of Life Sciences, School of Environment and Life Sciences, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China.
| |
Collapse
|
30
|
A New Therapeutic Strategy Targeting Protein Deacetylation for Spinal Cord Injury. Neuroscience 2020; 451:197-206. [PMID: 33039524 DOI: 10.1016/j.neuroscience.2020.09.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
Lysine acetylation is a post-translational modification that regulates a diversity of biological processes. However, its implication in spinal cord injury (SCI) remains unclear. Here we investigated the acetylation events in injured spinal cords on a proteomic scale for the first time. Additionally, whether promoting acetylation could mitigate SCI was evaluated. A total of 268 differentially acetylated peptides were identified. Among them, 2 peptides were up-acetylated and 141 peptides were down-acetylated in the injured spinal cord tissues (Fold change >2 and P < 0.05). There were also 116 unique acetylated peptides in the sham group and 9 unique acetylated peptides in the SCI group. Functional enrichment analysis revealed that differently acetylated proteins were involved in multiple cellular processes and metabolic processes. Kyoto Encyclopaedia of Genes and Genomes analysis showed that several pathways, including cGMP-PKG signaling pathway and hypoxia-inducible factor-1 (HIF-1) signaling pathway, were predominantly presented. Moreover, promoting acetylation using glycerol triacetate (GTA) showed a therapeutic effect on SCI, with improved Basso-Beattie-Bresnahan scores and histologic morphology, and decreased neuronal apoptosis and inflammation. In conclusion, our data indicated that protein deacetylation might play crucial roles in the development of secondary injury of SCI, and promoting acetylation by GTA effectively mitigated SCI. Our data not only enhance our understanding on acetylproteome dataset in the spinal cord tissues, but also provide novel insights for the treatment of SCI.
Collapse
|
31
|
Wang H, Zhang C, Yang LE, Yang Z. Hederagenin Modulates M1 Microglial Inflammatory Responses and Neurite Outgrowth. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20946252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder. Neurite atrophy and synaptic loss initiate the onset of neuronal death, while the activated M1 microglia-induced neuroinflammatory microenvironment inhibits neurite regeneration and exacerbates neuronal loss. Thus, optimizing the brain microenvironment using small compounds through suppressing activated M1 microglia and promoting neurite regrowth might be an effective therapeutic strategy for AD. We found that hederagenin (HED), a naturally occurring triterpene compound, inhibited lipopolysaccharide-induced nitric oxide generation and downregulated expression of proinflammatory cytokines, such as tumor necrosis factor-α, interleukin-1β (IL-1β), and IL-6. Further investigation of primary microglia confirmed that HED inhibited Iba-1 positive M1 microglia. However, no changes were seen in CD206 positive M2 microglia polarization. HED remarkably suppressed phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells subunit p65 signaling. In addition, HED ameliorated Aβ25-35-induced neuritic atrophy and neuronal death. Therefore, HED might be a therapeutic candidate for AD.
Collapse
Affiliation(s)
- Hua Wang
- Department of Pharmacy, The Second Hospital of Shandong University, Jinan, P. R. China
| | - Cai Zhang
- Department of Nutrition and Marine Drugs, College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang, P. R. China
| | - Long-en Yang
- Department of Nutrition and Marine Drugs, College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang, P. R. China
| | - Zhiyou Yang
- Department of Nutrition and Marine Drugs, College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Ocean University, Zhanjiang, P. R. China
- Department of Neuropharmacology, Shenzhen Institute of Guangdong Ocean University, Shenzhen, P. R. China
| |
Collapse
|
32
|
Abstract
IMPACT STATEMENT Brain development and degeneration are highly complex processes that are regulated by a large number of molecules and signaling pathways the identities of which are being unraveled. Accumulating evidence points to histone deacetylases and epigenetic mechanisms as being important regulators of these processes. In this review, we describe that histone deacetylase-3 (HDAC3) is a particularly crucial regulator of both neurodevelopment and neurodegeneration. In addition, HDAC3 regulates memory formation, synaptic plasticity, and the cognitive impairment associated with normal aging. Understanding how HDAC3 functions contributes to the normal development and functioning of the brain while also promoting neurodegeneration could lead to the development of therapeutic approaches for neurodevelopmental, neuropsychiatric, and neurodegenerative disorders.
Collapse
|
33
|
Kuboyama T, Yang X, Tohda C. Natural Medicines and Their Underlying Mechanisms of Prevention and Recovery from Amyloid Β-Induced Axonal Degeneration in Alzheimer's Disease. Int J Mol Sci 2020; 21:E4665. [PMID: 32630004 PMCID: PMC7369795 DOI: 10.3390/ijms21134665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 01/26/2023] Open
Abstract
In Alzheimer's disease (AD), amyloid β (Aβ) induces axonal degeneration, neuronal network disruption, and memory impairment. Although many candidate drugs to reduce Aβ have been clinically investigated, they failed to recover the memory function in AD patients. Reportedly, Aβ deposition occurred before the onset of AD. Once neuronal networks were disrupted by Aβ, they could hardly be recovered. Therefore, we speculated that only removal of Aβ was not enough for AD therapy, and prevention and recovery from neuronal network disruption were also needed. This review describes the challenges related to the condition of axons for AD therapy. We established novel in vitro models of Aβ-induced axonal degeneration. Using these models, we found that several traditional medicines and their constituents prevented or helped recover from Aβ-induced axonal degeneration. These drugs also prevented or helped recover from memory impairment in in vivo models of AD. One of these drugs ameliorated memory decline in AD patients in a clinical study. These results indicate that prevention and recovery from axonal degeneration are possible strategies for AD therapy.
Collapse
Affiliation(s)
- Tomoharu Kuboyama
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan; (T.K.); (X.Y.)
- Laboratory of Pharmacognosy, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan
| | - Ximeng Yang
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan; (T.K.); (X.Y.)
| | - Chihiro Tohda
- Section of Neuromedical Science, Institute of Natural Medicine, University of Toyama, Sugitani 2630, Toyama 930-0194, Japan; (T.K.); (X.Y.)
| |
Collapse
|
34
|
Zhou X, Wahane S, Friedl MS, Kluge M, Friedel CC, Avrampou K, Zachariou V, Guo L, Zhang B, He X, Friedel RH, Zou H. Microglia and macrophages promote corralling, wound compaction and recovery after spinal cord injury via Plexin-B2. Nat Neurosci 2020; 23:337-350. [PMID: 32112058 PMCID: PMC7412870 DOI: 10.1038/s41593-020-0597-7] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tissue repair after spinal cord injury requires the mobilization of immune and glial cells to form a protective barrier that seals the wound and facilitates debris clearing, inflammatory containment and matrix compaction. This process involves corralling, wherein phagocytic immune cells become confined to the necrotic core, which is surrounded by an astrocytic border. Here we elucidate a temporally distinct gene signature in injury-activated microglia and macrophages (IAMs) that engages axon guidance pathways. Plexin-B2 is upregulated in IAMs and is required for motor sensory recovery after spinal cord injury. Plexin-B2 deletion in myeloid cells impairs corralling, leading to diffuse tissue damage, inflammatory spillover and hampered axon regeneration. Corralling begins early and requires Plexin-B2 in both microglia and macrophages. Mechanistically, Plexin-B2 promotes microglia motility, steers IAMs away from colliding cells and facilitates matrix compaction. Our data therefore establish Plexin-B2 as an important link that integrates biochemical cues and physical interactions of IAMs with the injury microenvironment during wound healing.
Collapse
Affiliation(s)
- Xiang Zhou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shalaka Wahane
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marie-Sophie Friedl
- Institut für Informatik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Kluge
- Institut für Informatik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Caroline C Friedel
- Institut für Informatik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kleopatra Avrampou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lei Guo
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xijing He
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an International Medical Center, Xi'an, China
| | - Roland H Friedel
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Hongyan Zou
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
35
|
Matheson R, Chida K, Lu H, Clendaniel V, Fisher M, Thomas A, Lo EH, Selim M, Shehadah A. Neuroprotective Effects of Selective Inhibition of Histone Deacetylase 3 in Experimental Stroke. Transl Stroke Res 2020; 11:1052-1063. [PMID: 32016769 DOI: 10.1007/s12975-020-00783-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/15/2022]
Abstract
Histone deacetylase 3 (HDAC3) has been implicated as neurotoxic in several neurodegenerative conditions. However, the role of HDAC3 in ischemic stroke has not been thoroughly explored. We tested the hypothesis that selective inhibition of HDAC3 after stroke affords neuroprotection. Adult male Wistar rats (n = 8/group) were subjected to 2 h of middle cerebral artery occlusion (MCAO), and randomly selected animals were treated intraperitoneally twice with either vehicle (1% Tween 80) or a selective HDAC3 inhibitor (RGFP966, 10 mg/kg) at 2 and 24 h after MCAO. Long-term behavioral tests were performed up to 28 days after MCAO. Another set of rats (n = 7/group) were sacrificed at 3 days for histological analysis. Immunostaining for HDAC3, acetyl-Histone 3 (AcH3), NeuN, TNF-alpha, toll-like receptor 4 (TLR4), cleaved caspase-3, cleaved poly (ADP-ribose) polymerase (PARP), Akt, and TUNEL were performed. Selective HDAC3 inhibition improved long-term functional outcome (p < 0.05) and reduced infarct volume (p < 0.0001). HDAC3 inhibition increased levels of AcH3 in the ischemic brain (p = 0.016). Higher levels of AcH3 were significantly correlated with better neurological scores and smaller infarct volumes (r = 0.74, p = 0.002; r = 0.6, p = 0.02, respectively). The RGFP966 treatment reduced apoptosis-TUNEL+, cleaved caspase-3+, and cleaved PARP+ cells-and neuroinflammation-TNF-alpha+ and TLR4+ cells-in the ischemic border compared to vehicle control (p < 0.05). The RGFP966 treatment also increased Akt expression in the ipsilateral cortex (p < 0.001). Selective HDAC3 inhibition after stroke improves long-term neurological outcome and decreases infarct volume. The neuroprotective effects of HDAC3 inhibition are associated with a reduction in apoptosis and inflammation and upregulation of the Akt pathway.
Collapse
Affiliation(s)
- Rudy Matheson
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Kohei Chida
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Hui Lu
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.,Xuan Wu Hospital/Capital Medical University, Xicheng district, Beijing, 100053, People's Republic of China
| | - Victoria Clendaniel
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Marc Fisher
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Ajith Thomas
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Magdy Selim
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Amjad Shehadah
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
36
|
Zhao H, Li G, Zhang S, Li F, Wang R, Tao Z, Ma Q, Han Z, Yan F, Fan J, Li L, Ji X, Luo Y. Inhibition of histone deacetylase 3 by MiR-494 alleviates neuronal loss and improves neurological recovery in experimental stroke. J Cereb Blood Flow Metab 2019; 39:2392-2405. [PMID: 31510852 PMCID: PMC6893973 DOI: 10.1177/0271678x19875201] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
HDAC3 is an essential negative regulator of neuronal plasticity and memory formation. Although a chemical inhibitor has been invented, little is known about its endogenous modulators. We explored whether miR-494 affects HDAC3-mediated neuronal injury following acute ischemic stroke. A substantial increase in plasma miR-494 was detected in AIS patients and was positively associated with the mRS at one year after symptom onset. The miR-494 levels were transiently increased in the infarcted brain tissue of mice. In contrast, miR-494 levels were reduced in neurons but increased in the medium after OGD. Intracerebroventricular injection of miR-494 agomir reduced neuronal apoptosis and infarct volume at the acute stage of MCAO, promoted axonal plasticity and long-term outcomes at the recovery stage, suppressed neuronal ataxin-3 and HDAC3 expression and increased acetyl-H3K9 levels in the ipsilateral hemisphere. In vitro studies confirmed that miR-494 posttranslationally inhibited HDAC3 in neurons and prevented OGD-induced neuronal axonal injury. The HDAC3 inhibitor increased acetyl-H3K9 levels and reversed miR-494 antagomir-aggravated acute cerebral ischemic injury, as well as brain atrophy and long-term functional recovery. These results suggest that miR-494 may serve as a predictive biomarker of functional outcomes in AIS patients and a potential therapeutic target for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Haiping Zhao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Guangwen Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Sijia Zhang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Fangfang Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Zhen Tao
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Qingfeng Ma
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ziping Han
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Feng Yan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lingzhi Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| | - Xunming Ji
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
The inflammatory effect of epigenetic factors and modifications in type 2 diabetes. Inflammopharmacology 2019; 28:345-362. [PMID: 31707555 DOI: 10.1007/s10787-019-00663-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023]
Abstract
Inflammation has a central role in the etiology of type 2 diabetes (T2D) and its complications. Both genetic and epigenetic factors have been implicated in the development of T2D-associated inflammation. Epigenetic mechanisms regulate the function of several components of the immune system. Diabetic conditions trigger aberrant epigenetic alterations that contribute to the progression of insulin resistance and β-cell dysfunction by induction of inflammatory responses. Thus, targeting epigenetic factors and modifications, as one of the underlying causes of inflammation, could lead to the development of novel immune-based strategies for the treatment of T2D. The aim of this review is to provide an overview of the epigenetic mechanisms involved in the propagation and perpetuation of chronic inflammation in T2D. We also discuss the possible anti-inflammatory approaches that target epigenetic factors for the treatment of T2D.
Collapse
|
38
|
Kuboyama T. [Development of New Therapies for Neurodegenerative Diseases via Axonal Growth]. YAKUGAKU ZASSHI 2019; 139:1385-1390. [PMID: 31685734 DOI: 10.1248/yakushi.19-00147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In neurodegenerative diseases, such as Alzheimer's disease (AD) and spinal cord injury (SCI), inhibited axonal regeneration lead to irreversible functional impairment. Although many agents that eliminate axonal growth impediments have been clinically investigated, none induced functional recovery. I hypothesized that the removal of impediments alone was not enough and that promoting axonal growth and neuronal network reconstruction were needed for recovery from neurodegenerative diseases. To promote axonal growth, I have focused on neurons and microglia. In vitro models of AD and SCI were developed by culturing neurons in the presence of amyloid β (Aβ) and chondroitin sulfate proteoglycan, respectively. These were then used to identify several extracts of herbal medicines and their constituents that promoted axonal growth. Oral administration of these extracts and their constituents improved memory and motor function in in vivo mouse models of AD and SCI, respectively. The bioactive compounds in these extracts were identified by analyzing brain and spinal cord samples from the mice. Their protein targets were identified using the drug affinity responsive target stability method. Analysis of early events in the axons after culture with Aβ revealed that the inhibition of endocytosis was sufficient to prevent the axonal atrophy and memory deficits caused by Aβ. The compounds that increased M2 microglia were observed to promote axonal normalization and growth; they were also found to recover memory and motor function in mice models of AD and SCI, respectively. The above results indicate that axonal growth plays important roles in the recovery from AD and SCI.
Collapse
Affiliation(s)
- Tomoharu Kuboyama
- Division of Neuromedical Science, Institute of Natural Medicine, University of Toyama
| |
Collapse
|
39
|
Platycodigenin as Potential Drug Candidate for Alzheimer's Disease via Modulating Microglial Polarization and Neurite Regeneration. Molecules 2019; 24:molecules24183207. [PMID: 31487775 PMCID: PMC6767002 DOI: 10.3390/molecules24183207] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022] Open
Abstract
Neuroinflammatory microenvironment, regulating neurite regrowth and neuronal survival, plays a critical role in Alzheimer’s disease (AD). During neuroinflammation, microglia are activated, inducing the release of inflammatory or anti-inflammatory factors depending on their polarization into classical M1 microglia or alternative M2 phenotype. Therefore, optimizing brain microenvironment by small molecule-targeted microglia polarization and promoting neurite regeneration might be a potential therapeutic strategy for AD. In this study, we found platycodigenin, a naturally occurring triterpenoid, promoted M2 polarization and inhibited M1 polarization in lipopolysaccharide (LPS)-stimulated BV2 and primary microglia. Platycodigenin downregulated pro-inflammatory molecules such as interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-6 and nitric oxide (NO), while upregulated anti-inflammatory cytokine IL-10. Further investigation confirmed that platycodigenin inhibited cyclooxygenase-2 (Cox2) positive M1 but increased Ym1/2 positive M2 microglial polarization in primary microglia. In addition, platycodigenin significantly decreased LPS-induced the hyperphosphorylation of mitogen-activated protein kinase (MAPK) p38 and nuclear factor-κB (NF-κB) p65 subunits. Furthermore, the inactivation of peroxisome proliferators-activated receptor γ (PPARγ) induced by LPS was completely ameliorated by platycodigenin. Platycodigenin also promoted neurite regeneration and neuronal survival after Aβ treatment in primary cortical neurons. Taken together, our study for the first time clarified that platycodigenin effectively ameliorated LPS-induced inflammation and Aβ-induced neurite atrophy and neuronal death.
Collapse
|
40
|
Wang S, Smith GM, Selzer ME, Li S. Emerging molecular therapeutic targets for spinal cord injury. Expert Opin Ther Targets 2019; 23:787-803. [PMID: 31460807 DOI: 10.1080/14728222.2019.1661381] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: Spinal cord injury (SCI) is a complicated and devastating neurological disorder. Patients with SCI usually have dramatically reduced quality of life. In recent years, numerous studies have reported advances in understanding the pathophysiology of SCI and developing preclinical therapeutic strategies for SCI, including various molecular therapies, and yet there is still no cure. Areas covered: After SCI, tissue damage, responses and repair involve interactions among many cellular components, including neurons, axons, glia, leukocytes, and other cells. Accordingly, numerous cellular genes and molecules have become therapeutic targets for neural tissue repair, circuit reconstruction, and behavioral restoration. Here, we review the major recent advances in biological and molecular strategies to enhance neuroprotection, axon regeneration, remyelination, neuroplasticity and functional recovery in preclinical studies of SCI. Expert opinion: Researchers have made tremendous progress in identifying individual and combined molecular therapies in animal studies. It is very important to identify additional highly effective treatments for early neuroprotective intervention and for functionally meaningful axon regeneration and neuronal reconnections. Because multiple mechanisms contribute to the functional loss after SCI, combining the most promising approaches that target different pathophysiological and molecular mechanisms should exhibit synergistic actions for maximal functional restoration. [Databases searched: PubMed; inclusive dates: 6/27/2019].
Collapse
Affiliation(s)
- Shuo Wang
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Anatomy and Cell Biology, Temple University School of Medicine , Philadelphia , PA , USA
| | - George M Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Neuroscience, Temple University School of Medicine , Philadelphia , PA , USA
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Neurology, Temple University School of Medicine , Philadelphia , PA , USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Anatomy and Cell Biology, Temple University School of Medicine , Philadelphia , PA , USA
| |
Collapse
|
41
|
Akhmetzyanova E, Kletenkov K, Mukhamedshina Y, Rizvanov A. Different Approaches to Modulation of Microglia Phenotypes After Spinal Cord Injury. Front Syst Neurosci 2019; 13:37. [PMID: 31507384 PMCID: PMC6718713 DOI: 10.3389/fnsys.2019.00037] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/29/2019] [Indexed: 01/04/2023] Open
Abstract
Microglial cells, which are highly plastic, immediately respond to any change in the microenvironment by becoming activated and shifting the phenotype toward neurotoxicity or neuroprotection. The polarization of microglia/macrophages after spinal cord injury (SCI) seems to be a dynamic process and can change depending on the microenvironment, stage, course, and severity of the posttraumatic process. Effective methods to modulate microglia toward a neuroprotective phenotype in order to stimulate neuroregeneration are actively sought for. In this context, available approaches that can selectively impact the polarization of microglia/macrophages regulate synthesis of trophic factors and cytokines/chemokines in them, and their phagocytic function and effects on the course and outcome of SCI are discussed in this review.
Collapse
Affiliation(s)
- Elvira Akhmetzyanova
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Konstantin Kletenkov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Yana Mukhamedshina
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Albert Rizvanov
- OpenLab Gene and Cell Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
42
|
Placek K, Schultze JL, Aschenbrenner AC. Epigenetic reprogramming of immune cells in injury, repair, and resolution. J Clin Invest 2019; 129:2994-3005. [PMID: 31329166 DOI: 10.1172/jci124619] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune cells are pivotal in the reaction to injury, whereupon, under ideal conditions, repair and resolution phases restore homeostasis following initial acute inflammation. Immune cell activation and reprogramming require transcriptional changes that can only be initiated if epigenetic alterations occur. Recently, accelerated deciphering of epigenetic mechanisms has extended knowledge of epigenetic regulation, including long-distance chromatin remodeling, DNA methylation, posttranslational histone modifications, and involvement of small and long noncoding RNAs. Epigenetic changes have been linked to aspects of immune cell development, activation, and differentiation. Furthermore, genome-wide epigenetic landscapes have been established for some immune cells, including tissue-resident macrophages, and blood-derived cells including T cells. The epigenetic mechanisms underlying developmental steps from hematopoietic stem cells to fully differentiated immune cells led to development of epigenetic technologies and insights into general rules of epigenetic regulation. Compared with more advanced research areas, epigenetic reprogramming of immune cells in injury remains in its infancy. While the early epigenetic mechanisms supporting activation of the immune response to injury have been studied, less is known about resolution and repair phases and cell type-specific changes. We review prominent recent findings concerning injury-mediated epigenetic reprogramming, particularly in stroke and myocardial infarction. Lastly, we illustrate how single-cell technologies will be crucial to understanding epigenetic reprogramming in the complex sequential processes following injury.
Collapse
Affiliation(s)
- Katarzyna Placek
- Immunology and Metabolism, LIMES Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Platform for Single Cell Genomics and Epigenomics at the German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany.,Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
43
|
Wahane S, Halawani D, Zhou X, Zou H. Epigenetic Regulation Of Axon Regeneration and Glial Activation in Injury Responses. Front Genet 2019; 10:640. [PMID: 31354788 PMCID: PMC6629966 DOI: 10.3389/fgene.2019.00640] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
Injury to the nervous system triggers a multicellular response in which epigenetic mechanisms play an important role in regulating cell type-specific transcriptional changes. Here, we summarize recent progress in characterizing neuronal intrinsic and extrinsic chromatin reconfigurations and epigenetic changes triggered by axonal injury that shape neuroplasticity and glial functions. We specifically discuss regeneration-associated transcriptional modules comprised of transcription factors and epigenetic regulators that control axon growth competence. We also review epigenetic regulation of neuroinflammation and astroglial responses that impact neural repair. These advances provide a framework for developing epigenetic strategies to maximize adaptive alterations while minimizing maladaptive stress responses in order to enhance axon regeneration and achieve functional recovery after injury.
Collapse
Affiliation(s)
- Shalaka Wahane
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Dalia Halawani
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiang Zhou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongyan Zou
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
44
|
Hervera A, Zhou L, Palmisano I, McLachlan E, Kong G, Hutson TH, Danzi MC, Lemmon VP, Bixby JL, Matamoros‐Angles A, Forsberg K, De Virgiliis F, Matheos DP, Kwapis J, Wood MA, Puttagunta R, del Río JA, Di Giovanni S. PP4-dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure. EMBO J 2019; 38:e101032. [PMID: 31268609 PMCID: PMC6600644 DOI: 10.15252/embj.2018101032] [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] [Received: 10/29/2018] [Revised: 04/15/2019] [Accepted: 04/17/2019] [Indexed: 01/01/2023] Open
Abstract
The molecular mechanisms discriminating between regenerative failure and success remain elusive. While a regeneration-competent peripheral nerve injury mounts a regenerative gene expression response in bipolar dorsal root ganglia (DRG) sensory neurons, a regeneration-incompetent central spinal cord injury does not. This dichotomic response offers a unique opportunity to investigate the fundamental biological mechanisms underpinning regenerative ability. Following a pharmacological screen with small-molecule inhibitors targeting key epigenetic enzymes in DRG neurons, we identified HDAC3 signalling as a novel candidate brake to axonal regenerative growth. In vivo, we determined that only a regenerative peripheral but not a central spinal injury induces an increase in calcium, which activates protein phosphatase 4 that in turn dephosphorylates HDAC3, thus impairing its activity and enhancing histone acetylation. Bioinformatics analysis of ex vivo H3K9ac ChIPseq and RNAseq from DRG followed by promoter acetylation and protein expression studies implicated HDAC3 in the regulation of multiple regenerative pathways. Finally, genetic or pharmacological HDAC3 inhibition overcame regenerative failure of sensory axons following spinal cord injury. Together, these data indicate that PP4-dependent HDAC3 dephosphorylation discriminates between axonal regeneration and regenerative failure.
Collapse
Affiliation(s)
- Arnau Hervera
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
- Molecular and Cellular NeurobiotechnologyInstitute for Bioengineering of Catalonia (IBEC)Parc Científic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat de BarcelonaBarcelonaSpain
- Institute of NeuroscienceUniversity of BarcelonaBarcelonaSpain
| | - Luming Zhou
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
- Laboratory for NeuroRegeneration and RepairCenter for NeurologyHertie Institute for Clinical Brain ResearchUniversity of TuebingenTuebingenGermany
- Graduate School for Cellular and Molecular NeuroscienceUniversity of TuebingenTuebingenGermany
| | - Ilaria Palmisano
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
| | - Eilidh McLachlan
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
| | - Guiping Kong
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
- Laboratory for NeuroRegeneration and RepairCenter for NeurologyHertie Institute for Clinical Brain ResearchUniversity of TuebingenTuebingenGermany
| | - Thomas H Hutson
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
| | - Matt C Danzi
- The Miami Project to Cure ParalysisDepartment of Neurological SurgeryMiller School of MedicineUniversity of MiamiMiamiFLUSA
| | - Vance P Lemmon
- The Miami Project to Cure ParalysisDepartment of Neurological SurgeryMiller School of MedicineUniversity of MiamiMiamiFLUSA
| | - John L Bixby
- The Miami Project to Cure ParalysisDepartment of Neurological SurgeryMiller School of MedicineUniversity of MiamiMiamiFLUSA
| | - Andreu Matamoros‐Angles
- Molecular and Cellular NeurobiotechnologyInstitute for Bioengineering of Catalonia (IBEC)Parc Científic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat de BarcelonaBarcelonaSpain
- Institute of NeuroscienceUniversity of BarcelonaBarcelonaSpain
| | - Kirsi Forsberg
- Laboratory for NeuroRegeneration and RepairCenter for NeurologyHertie Institute for Clinical Brain ResearchUniversity of TuebingenTuebingenGermany
| | - Francesco De Virgiliis
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
- Laboratory for NeuroRegeneration and RepairCenter for NeurologyHertie Institute for Clinical Brain ResearchUniversity of TuebingenTuebingenGermany
- Graduate School for Cellular and Molecular NeuroscienceUniversity of TuebingenTuebingenGermany
| | - Dina P Matheos
- Center for the Neurobiology of Learning & MemoryDepartment of Neurobiology & BehaviorUniversity of CaliforniaIrvineCAUSA
| | - Janine Kwapis
- Center for the Neurobiology of Learning & MemoryDepartment of Neurobiology & BehaviorUniversity of CaliforniaIrvineCAUSA
| | - Marcelo A Wood
- Center for the Neurobiology of Learning & MemoryDepartment of Neurobiology & BehaviorUniversity of CaliforniaIrvineCAUSA
| | - Radhika Puttagunta
- Laboratory for NeuroRegeneration and RepairCenter for NeurologyHertie Institute for Clinical Brain ResearchUniversity of TuebingenTuebingenGermany
- Spinal Cord Injury CenterUniversity Hospital HeidelbergHeidelbergGermany
| | - José Antonio del Río
- Molecular and Cellular NeurobiotechnologyInstitute for Bioengineering of Catalonia (IBEC)Parc Científic de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Department of Cell Biology, Physiology and ImmunologyUniversitat de BarcelonaBarcelonaSpain
- Institute of NeuroscienceUniversity of BarcelonaBarcelonaSpain
| | - Simone Di Giovanni
- Department of MedicineDivision of Brain SciencesMolecular NeuroregenerationImperial College LondonLondonUK
- Laboratory for NeuroRegeneration and RepairCenter for NeurologyHertie Institute for Clinical Brain ResearchUniversity of TuebingenTuebingenGermany
| |
Collapse
|
45
|
Yang Z, Kuboyama T, Tohda C. Naringenin promotes microglial
M2
polarization and
Aβ
degradation enzyme expression. Phytother Res 2019; 33:1114-1121. [DOI: 10.1002/ptr.6305] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/22/2018] [Accepted: 01/11/2019] [Indexed: 11/05/2022]
Affiliation(s)
- Zhiyou Yang
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural MedicineUniversity of Toyama Toyama Japan
- Research Institute and Key Laboratory for Marine Drugs and Nutrition, College of Food Science and TechnologyGuangdong Ocean University Zhanjiang China
| | - Tomoharu Kuboyama
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural MedicineUniversity of Toyama Toyama Japan
| | - Chihiro Tohda
- Division of Neuromedical Science, Department of Bioscience, Institute of Natural MedicineUniversity of Toyama Toyama Japan
| |
Collapse
|
46
|
Sanchez S, Lemmens S, Baeten P, Sommer D, Dooley D, Hendrix S, Gou Fabregas M. HDAC3 Inhibition Promotes Alternative Activation of Macrophages but Does Not Affect Functional Recovery after Spinal Cord Injury. Exp Neurobiol 2018; 27:437-452. [PMID: 30429652 PMCID: PMC6221838 DOI: 10.5607/en.2018.27.5.437] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/07/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022] Open
Abstract
After spinal cord injury (SCI), monocyte derived macrophages play a detrimental role. Histone deacetylases (HDACs) are central epigenetic regulators of macrophage-polarization. We hypothesized that HDAC3 inhibition suppresses the pro-inflammatory macrophage phenotype (M1), promotes the anti-inflammatory phenotype (M2) and improves functional recovery after SCI. Therefore, two inhibitors of HDAC3 were selected, namely scriptaid and RGFP966. The impact on macrophage polarization was studied by investigating the effect on gene and protein expression of selected M1 and M2 markers. We show that scriptaid differentially influences M1 and M2 markers. It increases CD86 and iNOS gene expression and decreases GPR18, CD38, FPR2 and Arg-1 gene expression as well as the production of IL-6 and NO. RGFP966 primarily increased the expression of the M2 markers Arg-1 and Ym1 and reduced the production of IL-6 (M1). RGFP966 and scriptaid reduced the formation of foamy macrophages. Finally, to investigate the impact of HDAC3 inhibition on functional recovery after SCI, we studied the effects of RGFP966 and scriptaid in an in vivo T-cut hemisection SCI model. Histological analyses were performed on spinal cord sections to determine lesion size and astrogliosis, demyelinated area and selected infiltrating immune cells. RGFP966 and scriptaid did not affect functional recovery or histopathological outcome after SCI. In conclusion, these results indicate that specific HDAC3 inhibition with RGFP966 promotes alternative activation of macrophages and reduces the formation of foamy macrophages, but does not lead to a better functional recovery after SCI.
Collapse
Affiliation(s)
- Selien Sanchez
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Stefanie Lemmens
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Paulien Baeten
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Daniela Sommer
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Dearbhaile Dooley
- Health Science Centre, School of Medicine, University College Dublin, Dublin D04 V1W8, Ireland
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| | - Myriam Gou Fabregas
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek BE3590, Belgium
| |
Collapse
|
47
|
Adhikari N, Amin SA, Trivedi P, Jha T, Ghosh B. HDAC3 is a potential validated target for cancer: An overview on the benzamide-based selective HDAC3 inhibitors through comparative SAR/QSAR/QAAR approaches. Eur J Med Chem 2018; 157:1127-1142. [DOI: 10.1016/j.ejmech.2018.08.081] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/08/2018] [Accepted: 08/27/2018] [Indexed: 02/06/2023]
|
48
|
Palmisano I, Di Giovanni S. Advances and Limitations of Current Epigenetic Studies Investigating Mammalian Axonal Regeneration. Neurotherapeutics 2018; 15:529-540. [PMID: 29948919 PMCID: PMC6095777 DOI: 10.1007/s13311-018-0636-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Axonal regeneration relies on the expression of regenerative associated genes within a coordinated transcriptional programme, which is finely tuned as a result of the activation of several regenerative signalling pathways. In mammals, this chain of events occurs in neurons following peripheral axonal injury, however it fails upon axonal injury in the central nervous system, such as in the spinal cord and the brain. Accumulating evidence has been suggesting that epigenetic control is a key factor to initiate and sustain the regenerative transcriptional response and that it might contribute to regenerative success versus failure. This review will discuss experimental evidence so far showing a role for epigenetic regulation in models of peripheral and central nervous system axonal injury. It will also propose future directions to fill key knowledge gaps and to test whether epigenetic control might indeed discriminate between regenerative success and failure.
Collapse
Affiliation(s)
- Ilaria Palmisano
- Laboratory for Neuroregeneration, Centre for Restorative Neuroscience, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.
| | - Simone Di Giovanni
- Laboratory for Neuroregeneration, Centre for Restorative Neuroscience, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
49
|
Putatunda R, Bethea JR, Hu WH. Potential immunotherapies for traumatic brain and spinal cord injury. Chin J Traumatol 2018; 21:125-136. [PMID: 29759918 PMCID: PMC6033730 DOI: 10.1016/j.cjtee.2018.02.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 02/08/2018] [Indexed: 02/04/2023] Open
Abstract
Traumatic injury of the central nervous system (CNS) including brain and spinal cord remains a leading cause of morbidity and disability in the world. Delineating the mechanisms underlying the secondary and persistent injury versus the primary and transient injury has been drawing extensive attention for study during the past few decades. The sterile neuroinflammation during the secondary phase of injury has been frequently identified substrate underlying CNS injury, but as of now, no conclusive studies have determined whether this is a beneficial or detrimental role in the context of repair. Recent pioneering studies have demonstrated the key roles for the innate and adaptive immune responses in regulating sterile neuroinflammation and CNS repair. Some promising immunotherapeutic strategies have been recently developed for the treatment of CNS injury. This review updates the recent progress on elucidating the roles of the innate and adaptive immune responses in the context of CNS injury, the development and characterization of potential immunotherapeutics, as well as outstanding questions in this field.
Collapse
Affiliation(s)
- Raj Putatunda
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, USA
| | - John R. Bethea
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Wen-Hui Hu
- Center for Metabolic Disease Research, Department of Pathology and Laboratory Medicine, Temple University Lewis Katz School of Medicine, 3500 N Broad Street, Philadelphia, PA, USA,Corresponding author.
| |
Collapse
|
50
|
Chen S, Ye J, Chen X, Shi J, Wu W, Lin W, Lin W, Li Y, Fu H, Li S. Valproic acid attenuates traumatic spinal cord injury-induced inflammation via STAT1 and NF-κB pathway dependent of HDAC3. J Neuroinflammation 2018; 15:150. [PMID: 29776446 PMCID: PMC5960086 DOI: 10.1186/s12974-018-1193-6] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Microglial polarization with M1/M2 phenotype shifts and the subsequent neuroinflammatory responses are vital contributing factors for spinal cord injury (SCI)-induced secondary injury. Nuclear factor-κB (NF-κB) is considered the central transcription factor of inflammatory mediators, which plays a crucial role in microglial activation. Lysine acetylation of STAT1 seems necessary for NF-kB pathway activity, as it is regulated by histone deacetylases (HDACs). There have been no studies that have explained if HDAC inhibition by valproic acid (VPA) affects the NF-κB pathway via acetylation of STAT1 dependent of HDAC activity in the microglia-mediated central inflammation following SCI. We investigated the potential molecular mechanisms that focus on the phenotypic transition of microglia and the STAT1-mediated NF-κB acetylation after a VPA treatment. METHODS The Basso-Beattie-Bresnahan locomotion scale, the inclined plane test, the blood-spinal cord barrier, and Nissl staining were employed to determine the neuroprotective effects of VPA treatment after SCI. Assessment of microglia polarization and pro-inflammatory markers, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and interferon (INF)-γ was used to evaluate the neuroinflammatory responses and the anti-inflammatory effects of VPA treatment. Immunofluorescent staining and Western blot analysis were used to detect HDAC3 nuclear translocation, activity, and NF-κB signaling pathway activation to evaluate the effects of VPA treatment. The impact of STAT1 acetylation on NF-kB pathway and the interaction between STAT1 and NF-kB were assessed to evaluate anti-inflammation effects of VPA treatment and also whether these effects were dependent on a STAT1/NF-κB pathway to gain further insight into the mechanisms underlying the development of the neuroinflammatory response after SCI. RESULTS The results showed that the VPA treatment promoted the phenotypic shift of microglia from M1 to M2 phenotype and inhibited microglial activation, thus reducing the SCI-induced inflammatory factors. The VPA treatment upregulation of the acetylation of STAT1/NF-κB pathway was likely caused by the HDAC3 translocation to the nucleus and activity. These results indicated that the treatment with the VPA suppressed the expression and the activity of HDAC3 and enhanced STAT1, as well as NF-κB p65 acetylation following a SCI. The acetylation status of NF-kB p65 and the complex with NF-κB p65 and STAT1 inhibited the NF-kB p65 transcriptional activity and attenuated the microglia-mediated central inflammatory response following SCI. CONCLUSIONS These results suggested that the VPA treatment attenuated the inflammatory response by modulating microglia polarization through STAT1-mediated acetylation of the NF-κB pathway, dependent of HDAC3 activity. These effects led to neuroprotective effects following SCI.
Collapse
Affiliation(s)
- Shoubo Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Jingfang Ye
- Department of nursing faculty, Quanzhou Medical College, Quanzhou, 362000, Fujian Province, China
| | - Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
| | - Jinnan Shi
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Wenhua Wu
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Wenping Lin
- Department of Orthopaedics, The Second Affiliated Hospital, Fujian Medical Universityz, Quanzhou, 362000, Fujian Province, China
| | - Weibin Lin
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Yasong Li
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Huangde Fu
- Department of Neurosurgery, Affiliated Hospital of YouJiang Medical University for Nationalities, Baise, 533000, Guangxi Province, China
| | - Shun Li
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan Province, China.
| |
Collapse
|