1
|
Sadanandan J, Thomas S, Mathew IE, Huang Z, Blackburn SL, Tandon N, Lokhande H, McCrea PD, Bresnick EH, Dash PK, McBride DW, Harmanci A, Ahirwar LK, Jose D, Dienel AC, Zeineddine HA, Hong S, Kumar T P. Key epigenetic and signaling factors in the formation and maintenance of the blood-brain barrier. eLife 2024; 12:RP86978. [PMID: 39670988 PMCID: PMC11643625 DOI: 10.7554/elife.86978] [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] [Indexed: 12/14/2024] Open
Abstract
The blood-brain barrier (BBB) controls the movement of molecules into and out of the central nervous system (CNS). Since a functional BBB forms by mouse embryonic day E15.5, we reasoned that gene cohorts expressed in CNS endothelial cells (EC) at E13.5 contribute to BBB formation. In contrast, adult gene signatures reflect BBB maintenance mechanisms. Supporting this hypothesis, transcriptomic analysis revealed distinct cohorts of EC genes involved in BBB formation and maintenance. Here, we demonstrate that epigenetic regulator's histone deacetylase 2 (HDAC2) and polycomb repressive complex 2 (PRC2) control EC gene expression for BBB development and prevent Wnt/β-catenin (Wnt) target genes from being expressed in adult CNS ECs. Low Wnt activity during development modifies BBB genes epigenetically for the formation of functional BBB. As a Class-I HDAC inhibitor induces adult CNS ECs to regain Wnt activity and BBB genetic signatures that support BBB formation, our results inform strategies to promote BBB repair.
Collapse
Affiliation(s)
- Jayanarayanan Sadanandan
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Sithara Thomas
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Iny Elizabeth Mathew
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Zhen Huang
- Departments of Neurology & Neuroscience, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
| | - Spiros L Blackburn
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Nitin Tandon
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | | | - Pierre D McCrea
- Department of Genetics, TheUniversity of Texas MD Anderson Cancer CenterHoustonUnited States
| | - Emery H Bresnick
- Wisconsin Blood Cancer Research Institute, University of Wisconsin School of Medicine and Public HealthMadisonUnited States
| | - Pramod K Dash
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Devin W McBride
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Arif Harmanci
- UTHealth School of Biomedical InformaticsHoustonUnited States
| | - Lalit K Ahirwar
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Dania Jose
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Ari C Dienel
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Hussein A Zeineddine
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Sungha Hong
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| | - Peeyush Kumar T
- The Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center McGovern Medical SchoolHoustonUnited States
| |
Collapse
|
2
|
Spiteri Douglas R, Hartley MR, Yang JR, Franklin TB. Differential expression of Hdac2 in male and female mice of differing social status. Physiol Behav 2024; 273:114406. [PMID: 37949308 DOI: 10.1016/j.physbeh.2023.114406] [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: 05/23/2023] [Revised: 10/11/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Mice naturally form social hierarchies, and their experiences as subordinate or dominant mice inform future behavioural strategies. To better understand the neural bases of social dominance, we investigated hippocampal gene and protein expression of histone deacetylase 2 (HDAC2), an epigenetic regulator that decreases expression of synaptic plasticity genes and reduces excitatory synaptic function. Hdac2 in hippocampus was associated with social status. The gene for a closely related histone deacetylase (Hdac1), and HDAC2 protein expression, were not associated with social rank in hippocampus. These findings suggest that Hdac2 expression in hippocampus is distinctly linked with social status.
Collapse
Affiliation(s)
- Renée Spiteri Douglas
- The Social Lab, Dalhousie University, Department of Psychology and Neuroscience, Halifax, NS, Canada
| | - Mackenzie R Hartley
- The Social Lab, Dalhousie University, Department of Psychology and Neuroscience, Halifax, NS, Canada
| | - J Renee Yang
- The Social Lab, Dalhousie University, Department of Psychology and Neuroscience, Halifax, NS, Canada
| | - Tamara B Franklin
- The Social Lab, Dalhousie University, Department of Psychology and Neuroscience, Halifax, NS, Canada.
| |
Collapse
|
3
|
Lex RK, Vokes SA. Timing is everything: Transcriptional repression is not the default mode for regulating Hedgehog signaling. Bioessays 2022; 44:e2200139. [PMID: 36251875 PMCID: PMC9691524 DOI: 10.1002/bies.202200139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022]
Abstract
Hedgehog (HH) signaling is a conserved pathway that drives developmental growth and is essential for the formation of most organs. The expression of HH target genes is regulated by a dual switch mechanism where GLI proteins function as bifunctional transcriptional activators (in the presence of HH signaling) and transcriptional repressors (in the absence of HH signaling). This results in a tight control of GLI target gene expression during rapidly changing levels of pathway activity. It has long been presumed that GLI proteins also repress target genes prior to the initial expression of HH in a given tissue. This idea forms the basis for the limb bud pre-patterning model for regulating digit number. Recent findings indicate that GLI repressor proteins are indeed present prior to HH signaling but contrary to this model, GLI proteins are inert as they do not regulate transcriptional responses or enhancer chromatin modifications at this time. These findings suggest that GLI transcriptional repressor activity is not a default state as assumed, but is itself regulated in an unknown fashion. We discuss these findings and their implications for understanding pre-patterning, digit regulation, and HH-driven disease.
Collapse
Affiliation(s)
- Rachel K. Lex
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
| | - Steven A. Vokes
- Department of Molecular Bioscienc es, University of Texas at Austin, 100 E 24th Street Stop A5000, Austin, TX 78712 USA
| |
Collapse
|
4
|
Chotitumnavee J, Yamashita Y, Takahashi Y, Takada Y, Iida T, Oba M, Itoh Y, Suzuki T. Selective degradation of histone deacetylase 8 mediated by a proteolysis targeting chimera (PROTAC). Chem Commun (Camb) 2022; 58:4635-4638. [PMID: 35311871 DOI: 10.1039/d2cc00272h] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We developed a first-in-class proteolysis targeting chimera (PROTAC) for selective degradation of histone deacetylase 8 (HDAC8). The PROTAC induced degradation of HDAC8 without affecting the levels of other HDACs in cellular assays, and inhibited the growth of T-cell leukemia Jurkat cells more potently than a conventional HDAC8 inhibitor.
Collapse
Affiliation(s)
- Jiranan Chotitumnavee
- SANKEN, Osaka University, Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan. .,Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Yasunobu Yamashita
- SANKEN, Osaka University, Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan.
| | - Yukari Takahashi
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Yuri Takada
- SANKEN, Osaka University, Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan.
| | - Tetsuya Iida
- SANKEN, Osaka University, Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan. .,Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Makoto Oba
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Yukihiro Itoh
- SANKEN, Osaka University, Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan. .,Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| | - Takayoshi Suzuki
- SANKEN, Osaka University, Mihogaoka, Ibaraki-shi, Osaka 567-0047, Japan. .,Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto 606-0823, Japan
| |
Collapse
|
5
|
GLI transcriptional repression is inert prior to Hedgehog pathway activation. Nat Commun 2022; 13:808. [PMID: 35145123 PMCID: PMC8831537 DOI: 10.1038/s41467-022-28485-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/28/2022] [Indexed: 12/28/2022] Open
Abstract
The Hedgehog (HH) pathway regulates a spectrum of developmental processes through the transcriptional mediation of GLI proteins. GLI repressors control tissue patterning by preventing sub-threshold activation of HH target genes, presumably even before HH induction, while lack of GLI repression activates most targets. Despite GLI repression being central to HH regulation, it is unknown when it first becomes established in HH-responsive tissues. Here, we investigate whether GLI3 prevents precocious gene expression during limb development. Contrary to current dogma, we find that GLI3 is inert prior to HH signaling. While GLI3 binds to most targets, loss of Gli3 does not increase target gene expression, enhancer acetylation or accessibility, as it does post-HH signaling. Furthermore, GLI repression is established independently of HH signaling, but after its onset. Collectively, these surprising results challenge current GLI pre-patterning models and demonstrate that GLI repression is not a default state for the HH pathway. GLI repression has been presumed to be the default transcriptional state and important for pre-patterning tissues. Challenging current models, the authors show that GLI3 repression is inert in the limb bud before the onset of Hedgehog signaling.
Collapse
|
6
|
Donovan LL, Magnussen JH, Dyssegaard A, Lehel S, Hooker JM, Knudsen GM, Hansen HD. Imaging HDACs In Vivo: Cross-Validation of the [ 11C]Martinostat Radioligand in the Pig Brain. Mol Imaging Biol 2021; 22:569-577. [PMID: 31290052 DOI: 10.1007/s11307-019-01403-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE With the emerging knowledge about the impact of epigenetic alterations on behavior and brain disorders, the ability to measure epigenetic alterations in brain tissue in vivo has become critically important. We present the first in vivo/in vitro cross-validation of the novel positron emission tomography (PET) radioligand [11C]Martinostat in the pig brain with regard to its ability to measure histone deacetylase 1-3 (HDAC1-3) levels in vivo. PROCEDURES Nine female Danish landrace pigs underwent 121-min dynamic PET scans with [11C]Martinostat. We quantified [11C]Martinostat uptake using both a simple ratio method and kinetic models with arterial input function. By the end of the scan, the animals were euthanized and the brains were extracted. We measured HDAC1-3 protein levels in frontal cortex, cerebellum vermis, and hippocampus and compared the protein levels and regional outcome values to the [11C]Martinostat PET quantification. RESULTS [11C]Martinostat distributed widely across brain regions, with the highest uptake in the cerebellum vermis and the lowest in the olfactory bulbs. Based on the Akaike information criterion, the quantification was most reliably performed by Ichise MA1 kinetic modeling, but since the radioligand displayed very slow kinetics, we also calculated standard uptake value (SUV) ratios which correlated well with VT. The western blots revealed higher brain tissue protein levels of HDAC1/2 compared to HDAC3, and HDAC1 and HDAC2 levels were highly correlated in all three investigated brain regions. The in vivo SUV ratio measure correlated well with the in vitro HDAC1-3 levels, whereas no correlation was found between VT values and HDAC levels. CONCLUSIONS We found good correlation between in vivo measured SUV ratios and in vitro measures of HDAC 1-3 proteins, supporting that [11C]Martinostat provides a good in vivo measure of the cerebral HDAC1-3 protein levels.
Collapse
Affiliation(s)
- L L Donovan
- Neurobiology Research Unit and Center for NeuroPharm, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen O, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - J H Magnussen
- Neurobiology Research Unit and Center for NeuroPharm, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen O, Denmark
| | - A Dyssegaard
- Neurobiology Research Unit and Center for NeuroPharm, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen O, Denmark
| | - S Lehel
- PET and Cyclotron Unit, Copenhagen University Hospital Rigshospitalet, 2100, Copenhagen O, Denmark
| | - J M Hooker
- MGH/HST A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - G M Knudsen
- Neurobiology Research Unit and Center for NeuroPharm, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen O, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen N, Denmark
| | - H D Hansen
- Neurobiology Research Unit and Center for NeuroPharm, Copenhagen University Hospital Rigshospitalet, 9 Blegdamsvej, 2100, Copenhagen O, Denmark.
| |
Collapse
|
7
|
Lazarchuk P, Hernandez-Villanueva J, Pavlova MN, Federation A, MacCoss M, Sidorova JM. Mutual Balance of Histone Deacetylases 1 and 2 and the Acetyl Reader ATAD2 Regulates the Level of Acetylation of Histone H4 on Nascent Chromatin of Human Cells. Mol Cell Biol 2020; 40:e00421-19. [PMID: 32015101 PMCID: PMC7156220 DOI: 10.1128/mcb.00421-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/25/2019] [Accepted: 01/21/2020] [Indexed: 01/04/2023] Open
Abstract
Newly synthesized histone H4 that is incorporated into chromatin during DNA replication is acetylated on lysines 5 and 12. Histone deacetylase 1 (HDAC1) and HDAC2 are responsible for reducing H4 acetylation as chromatin matures. Using CRISPR-Cas9-generated hdac1- or hdac2-null fibroblasts, we determined that HDAC1 and HDAC2 do not fully compensate for each other in removing de novo acetyls on H4 in vivo Proteomics of nascent chromatin and proximity ligation assays with newly replicated DNA revealed the binding of ATAD2, a bromodomain-containing posttranslational modification (PTM) reader that recognizes acetylated H4. ATAD2 is a transcription facilitator overexpressed in several cancers and in the simian virus 40 (SV40)-transformed human fibroblast model cell line used in this study. The recruitment of ATAD2 to nascent chromatin was increased in hdac2 cells over the wild type, and ATAD2 depletion reduced the levels of nascent chromatin-associated, acetylated H4 in wild-type and hdac2 cells. We propose that overexpressed ATAD2 shifts the balance of H4 acetylation by protecting this mark from removal and that HDAC2 but not HDAC1 can effectively compete with ATAD2 for the target acetyls. ATAD2 depletion also reduced global RNA synthesis and nascent DNA-associated RNA. A moderate dependence on ATAD2 for replication fork progression was noted only for hdac2 cells overexpressing the protein.
Collapse
Affiliation(s)
- Pavlo Lazarchuk
- University of Washington, Department of Pathology, Seattle, Washington, USA
| | | | - Maria N Pavlova
- University of Washington, Department of Pathology, Seattle, Washington, USA
| | | | - Michael MacCoss
- University of Washington, Department of Genome Sciences, Seattle, Washington, USA
| | - Julia M Sidorova
- University of Washington, Department of Pathology, Seattle, Washington, USA
| |
Collapse
|
8
|
Aboukhatwa SM, Hanigan TW, Taha TY, Neerasa J, Ranjan R, El-Bastawissy EE, Elkersh MA, El-Moselhy TF, Frasor J, Mahmud N, McLachlan A, Petukhov PA. Structurally Diverse Histone Deacetylase Photoreactive Probes: Design, Synthesis, and Photolabeling Studies in Live Cells and Tissue. ChemMedChem 2019; 14:1096-1107. [PMID: 30921497 PMCID: PMC6548601 DOI: 10.1002/cmdc.201900114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/28/2019] [Indexed: 01/27/2023]
Abstract
Histone deacetylase (HDAC) activity is modulated in vivo by post-translational modifications and formation of multiprotein complexes. Novel chemical tools to study how these factors affect engagement of HDAC isoforms by HDAC inhibitors (HDACi) in cells and tissues are needed. In this study, a synthetic strategy to access chemically diverse photoreactive probes (PRPs) was developed and used to prepare seven novel HDAC PRPs 9-15. The class I HDAC isoform engagement by PRPs was determined in biochemical assays and photolabeling experiments in live SET-2, HepG2, HuH7, and HEK293T cell lines and in mouse liver tissue. Unlike the HDAC protein abundance and biochemical activity against recombinant HDACs, the chemotype of the PRPs and the type of cells were key in defining the engagement of HDAC isoforms in live cells. Our findings suggest that engagement of HDAC isoforms by HDACi in vivo may be substantially modulated in a cell- and tissue-type-dependent manner.
Collapse
Affiliation(s)
- Shaimaa M Aboukhatwa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Thomas W Hanigan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Taha Y Taha
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Jayaprakash Neerasa
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Rajeev Ranjan
- Section of Hematology/Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Eman E El-Bastawissy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Mohamed A Elkersh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University, Alexandria, 21311, Egypt
| | - Tarek F El-Moselhy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt
| | - Jonna Frasor
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Nadim Mahmud
- Section of Hematology/Oncology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Alan McLachlan
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Pavel A Petukhov
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| |
Collapse
|
9
|
Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid Suppresses Human Adenovirus Gene Expression and Replication. J Virol 2019; 93:JVI.00088-19. [PMID: 30944181 DOI: 10.1128/jvi.00088-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022] Open
Abstract
Human adenovirus (HAdV) causes minor illnesses in most patients but can lead to severe disease and death in pediatric, geriatric, and immunocompromised individuals. No approved antiviral therapy currently exists for the treatment of these severe HAdV-induced diseases. In this study, we show that the pan-histone deacetylase (HDAC) inhibitor SAHA reduces HAdV-5 gene expression and DNA replication in tissue culture, ultimately decreasing virus yield from infected cells. Importantly, SAHA also reduced gene expression from more virulent and clinically relevant serotypes, including HAdV-4 and HAdV-7. In addition to SAHA, several other HDAC inhibitors (e.g., trichostatin A, apicidin, and panobinostat) also affected HAdV gene expression. We determined that loss of class I HDAC activity, mainly HDAC2, impairs efficient expression of viral genes, and that E1A physically interacts with HDAC2. Our results suggest that HDAC activity is necessary for HAdV replication, which may represent a novel pharmacological target in HAdV-induced disease.IMPORTANCE Although human adenovirus (HAdV) can cause severe diseases that can be fatal in some populations, there are no effective treatments to combat HAdV infection. In this study, we determined that the pan-histone deacetylase (HDAC) inhibitor SAHA has inhibitory activity against several clinically relevant serotypes of HAdV. This U.S. Food and Drug Administration-approved compound affects various stages of the virus lifecycle and reduces virus yield even at low concentrations. We further report that class I HDAC activity, particularly HDAC2, is required for efficient expression of viral genes during lytic infection. Investigation of the mechanism underlying SAHA-mediated suppression of HAdV gene expression and replication will enhance current knowledge of virus-cell interaction and may aid in the development of more effective antivirals with lower toxicity for the treatment of HAdV infections.
Collapse
|
10
|
von Knethen A, Brüne B. Histone Deacetylation Inhibitors as Therapy Concept in Sepsis. Int J Mol Sci 2019; 20:ijms20020346. [PMID: 30654448 PMCID: PMC6359123 DOI: 10.3390/ijms20020346] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/11/2019] [Accepted: 01/12/2019] [Indexed: 12/15/2022] Open
Abstract
Sepsis is characterized by dysregulated gene expression, provoking a hyper-inflammatory response occurring in parallel to a hypo-inflammatory reaction. This is often associated with multi-organ failure, leading to the patient’s death. Therefore, reprogramming of these pro- and anti-inflammatory, as well as immune-response genes which are involved in acute systemic inflammation, is a therapy approach to prevent organ failure and to improve sepsis outcomes. Considering epigenetic, i.e., reversible, modifications of chromatin, not altering the DNA sequence as one tool to adapt the expression profile, inhibition of factors mediating these changes is important. Acetylation of histones by histone acetyltransferases (HATs) and initiating an open-chromatin structure leading to its active transcription is counteracted by histone deacetylases (HDACs). Histone deacetylation triggers a compact nucleosome structure preventing active transcription. Hence, inhibiting the activity of HDACs by specific inhibitors can be used to restore the expression profile of the cells. It can be assumed that HDAC inhibitors will reduce the expression of pro-, as well as anti-inflammatory mediators, which blocks sepsis progression. However, decreased cytokine expression might also be unfavorable, because it can be associated with decreased bacterial clearance.
Collapse
Affiliation(s)
- Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt/Main, 60590 Frankfurt, Germany.
- Fraunhofer⁻IME, Project Group Translational Medicine and Pharmacology (TMP), 60596 Frankfurt, Germany.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt/Main, 60590 Frankfurt, Germany.
- Fraunhofer⁻IME, Project Group Translational Medicine and Pharmacology (TMP), 60596 Frankfurt, Germany.
| |
Collapse
|