1
|
Morales-Herrejón G, García-Vázquez JB, Fernández-Pomares C, Bakalara N, Correa-Basurto J, Mendoza-Figueroa HL. Computationally Guided Design, Synthesis, and Evaluation of Novel Non-Hydroxamic Histone Deacetylase Inhibitors, Based on N-Trifluoroacetamide as a Zinc-Binding Group, Against Breast Cancer. Pharmaceuticals (Basel) 2025; 18:351. [PMID: 40143128 PMCID: PMC11944851 DOI: 10.3390/ph18030351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Histone deacetylases (HDACs) are enzymes that deacetylate histone proteins, impacting the transcriptional repression and activation of cancer-associated genes such as P53 and Ras. The overexpression of HDACs in breast cancer (BC) underscores their significance as therapeutic targets for modulating gene expression through epigenetic regulation. Methods: In this study, a novel series of SAHA (suberoylanilide hydroxamic acid) analogs were designed using an in silico ligand-based strategy. These analogs were then synthesized and evaluated for their HDAC-inhibitory capacity as well as their antiproliferative capacity on breast cancer cells. These compounds retained an aliphatic LINKER, mimicking the natural substrate acetyl-lysine, while differing from the hydroxamic fragment present in SAHA. Results: The synthesized compounds exhibited HDAC inhibitory activity, suggesting potential for binding to these pharmacological targets. Compounds 5b, 6a, and 6b were identified as promising candidates in the evaluation on breast cancer cell lines MCF-7 and MDA-MB-231 at 72 h. Specifically, compound 6b, which contains an N-trifluoroacetyl group as a zinc-binding group (ZBG), demonstrated an IC50 of 76.7 µM in the MDA-MB-231 cell line and 45.7 µM in the MCF-7 cell line. In the non-tumorigenic cell line, the compound exhibited an IC50 of 154.6 µM. Conversely, SAHA exhibited an almost negligible safety margin with regard to its cytotoxic activity when compared to breast cancer cells and healthy cells (MCF-10A). This observation underscores the elevated toxicity exhibited by hydroxamic acid-derived molecules. Conclusions: The bioisosteric modification of ZBG by N-trifluoroacetyl in 6a and 6b demonstrated favorable cytotoxic activity, exhibiting a higher safety margin. This study underscores the challenge of identifying novel ZBGs to replace hydroxamic acid in the development of HDAC inhibitors, with the objective of enhancing their physicochemical and toxicological profile for utilization in BC treatment.
Collapse
Affiliation(s)
- Gerardo Morales-Herrejón
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Ciudad de México 11340, Mexico; (G.M.-H.); (J.B.G.-V.); (C.F.-P.)
| | - Juan Benjamín García-Vázquez
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Ciudad de México 11340, Mexico; (G.M.-H.); (J.B.G.-V.); (C.F.-P.)
- Investigadoras e Investigadores por México CONAHCyT-Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Ciudad de México 11340, Mexico
| | - Cynthia Fernández-Pomares
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Ciudad de México 11340, Mexico; (G.M.-H.); (J.B.G.-V.); (C.F.-P.)
| | - Norbert Bakalara
- University Bordeaux, CNRS, Bordeaux INP-ENSTBB, CBMN, UMR 5248, F-33600 Pessac, France;
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Ciudad de México 11340, Mexico; (G.M.-H.); (J.B.G.-V.); (C.F.-P.)
| | - Humberto L. Mendoza-Figueroa
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina del Instituto Politécnico Nacional (ESM-IPN), Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Ciudad de México 11340, Mexico; (G.M.-H.); (J.B.G.-V.); (C.F.-P.)
| |
Collapse
|
2
|
Wood DJ, Tsvetkov E, Comte-Walters S, Welsh CL, Bloyd M, Wood TG, Akiki RM, Anderson EM, Penrod RD, Madan LK, Ball LE, Taniguchi M, Cowan CW. Epigenetic Control of an Auxiliary Subunit of Voltage-Gated Sodium Channels Regulates the Strength of Drug-Cue Associations and Relapse-Like Cocaine Seeking. Biol Psychiatry 2025:S0006-3223(25)00075-7. [PMID: 39923817 DOI: 10.1016/j.biopsych.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Repeated use of addictive drugs produces long-lasting and prepotent drug-cue associations that increase vulnerability for relapse in individuals with a substance use disorder. Epigenetic factors, such as HDAC5 (histone deacetylase 5), play a key role in regulating the formation of drug-cue associations, but the underlying mechanisms remain unclear. METHODS We used a combination of molecular biology, cultured cells, tandem mass spectrometry, deacetylase activity measurements, co-immunoprecipitation, and molecular dynamics simulations to assess HDAC5 structure-activity relationships. In male and female Long Evans rats, we used viral-mediated expression of HDAC5 mutants in the nucleus accumbens (NAc) to test effects on cocaine intravenous self-administration and cue-reinstated cocaine seeking. We also used in silico analysis of single-nucleus RNA sequencing data, quantitative reverse transcriptase-polymerase chain reaction, viral-mediated expression of Scn4b short hairpin RNA, patch-clamp electrophysiology, and rat cocaine or sucrose SA to assess Scn4b's effects on NAc intrinsic excitability and cued reward seeking. RESULTS We discovered that 2 conserved cysteines located near HDAC5's catalytic domain were required for its intrinsic deacetylase activity and that HDAC5's deacetylase activity was required in NAc medium spiny neurons (MSNs) to limit relapse-like cue-reinstated cocaine seeking. Moreover, we found that HDAC5 limited cocaine-seeking, but not sucrose-seeking, behavior by reducing NAc MSN intrinsic excitability through the deacetylase-dependent repression of Scn4b, which codes for an auxiliary subunit of voltage-gated sodium channels. CONCLUSIONS Our findings suggest that HDAC5's control of NAc Scn4b expression governs the formation of cocaine-cue, but not sucrose-cue, associations through modulation of NAc MSN intrinsic excitability and drug-induced NAc plasticity mechanisms.
Collapse
Affiliation(s)
- Daniel J Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina
| | - Evgeny Tsvetkov
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Susana Comte-Walters
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Colin L Welsh
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Michelle Bloyd
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina
| | - Timothy G Wood
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Rose Marie Akiki
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Medical Scientist Training Program, Medical University of South Carolina, Charleston, South Carolina
| | - Ethan M Anderson
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Rachel D Penrod
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Lalima K Madan
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Lauren E Ball
- Department of Pharmacology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Makoto Taniguchi
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Christopher W Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
3
|
Jasim SA, Altalbawy FMA, Abohassan M, Oghenemaro EF, Bishoyi AK, Singh RP, Kaur P, Sivaprasad GV, Mohammed JS, Hulail HM. Histone Deacetylases (HDACs) Roles in Inflammation-mediated Diseases; Current Knowledge. Cell Biochem Biophys 2024:10.1007/s12013-024-01587-0. [PMID: 39419931 DOI: 10.1007/s12013-024-01587-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
The histone acetyl transferases (HATs) and histone deacetylases (HDACs), which are mostly recognized for their involvement in regulating chromatin remodeling via histone acetylation/deacetylation, have been shown to also change several non-histone proteins to regulate other cellular processes. Acetylation affects the activity or function of cytokine receptors, nuclear hormone receptors, intracellular signaling molecules, and transcription factors in connection to inflammation. Some small-molecule HDAC inhibitors are utilized as anticancer medications in clinical settings due to their capability to regulate cellular growth arrest, differentiation, and death. Here, we summarize our present knowledge of the innate and adaptive immunological pathways that classical HDAC enzymes control. The aim is to justify the targeted (or non-targeted) use of inhibitors against certain HDAC enzymes in inflammatory diseases such as arthritis, inflammatory bowel diseases (IBD), airways inflammation and neurological diseases.
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques department, College of Health and Medical Technology, University of Al-maarif, Anbar, Iraq
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Ravindra Pal Singh
- Department of Pharmaceutics, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | | | - Hanen Mahmod Hulail
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
4
|
Turkman N, Xu S, Huang CH, Eyermann C, Salino J, Khan P. High-Contrast PET Imaging with [ 18F]NT160, a Class-IIa Histone Deacetylase Probe for In Vivo Imaging of Epigenetic Machinery in the Central Nervous System. J Med Chem 2023; 66:5611-5621. [PMID: 37068265 PMCID: PMC10150721 DOI: 10.1021/acs.jmedchem.2c02064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Indexed: 04/19/2023]
Abstract
We utilized positron emission tomography (PET) imaging in vivo to map the spatiotemporal biodistribution/expression of class-IIa histone deacetylases (class-IIa HDACs) in the central nervous system (CNS). Herein we report an improved radiosynthesis of [18F]NT160 using 4-hydroxy-TEMPO which led to a significant improvement in radiochemical yield and molar activity. PET imaging with [18F]NT160, a highly potent class-IIa HDAC inhibitor, led to high-quality and high-contrast images of the brain. [18F]NT160 displayed excellent pharmacokinetic and imaging characteristics: brain uptake is high in gray matter regions, tissue kinetics are appropriate for a 18F-tracer, and specific binding for class-IIa HDACs is demonstrated by self-blockade. Higher uptake with [18F]NT160 was observed in the hippocampus, thalamus, and cortex while the uptake in the cerebellum was relatively low. Overall, our current studies with [18F]NT160 will likely facilitate the development and clinical translation of PET tracers for imaging of class-IIa HDACs biodistribution/expression in cancer and the CNS.
Collapse
Affiliation(s)
- Nashaat Turkman
- Stony
Brook Cancer Center, Stony Brook, Long Island, New York 11794, United States
- Department
of Radiology, School of Medicine, Stony
Brook University, Long Island, New York 11794, United States
- Department
of Biomedical Engineering, Stony Brook University, Long Island, New York 11794, United States
| | - Sulan Xu
- Stony
Brook Cancer Center, Stony Brook, Long Island, New York 11794, United States
- Department
of Radiology, School of Medicine, Stony
Brook University, Long Island, New York 11794, United States
| | - Chun-Han Huang
- Stony
Brook Cancer Center, Stony Brook, Long Island, New York 11794, United States
- Department
of Radiology, School of Medicine, Stony
Brook University, Long Island, New York 11794, United States
- Department
of Biomedical Engineering, Stony Brook University, Long Island, New York 11794, United States
| | - Christopher Eyermann
- Department
of Radiology, School of Medicine, Stony
Brook University, Long Island, New York 11794, United States
- Department
of Surgery, School of Medicine, Stony Brook
University, Long Island, New York 11794, United States
| | - Julia Salino
- Stony
Brook Cancer Center, Stony Brook, Long Island, New York 11794, United States
- Department
of Radiology, School of Medicine, Stony
Brook University, Long Island, New York 11794, United States
| | - Palwasha Khan
- Stony
Brook Cancer Center, Stony Brook, Long Island, New York 11794, United States
- Department
of Radiology, School of Medicine, Stony
Brook University, Long Island, New York 11794, United States
| |
Collapse
|
5
|
Tien Anh D, Hai Nam N, Kircher B, Baecker D. The Impact of Fluorination on the Design of Histone Deacetylase Inhibitors. Molecules 2023; 28:molecules28041973. [PMID: 36838960 PMCID: PMC9965134 DOI: 10.3390/molecules28041973] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
In recent years, histone deacetylases (HDACs) have emerged as promising targets in the treatment of cancer. The approach is to inhibit HDACs with drugs known as HDAC inhibitors (HDACis). Such HDACis are broadly classified according to their chemical structure, e.g., hydroxamic acids, benzamides, thiols, short-chain fatty acids, and cyclic peptides. Fluorination plays an important role in the medicinal-chemical design of new active representatives. As a result of the introduction of fluorine into the chemical structure, parameters such as potency or selectivity towards isoforms of HDACs can be increased. However, the impact of fluorination cannot always be clearly deduced. Nevertheless, a change in lipophilicity and, hence, solubility, as well as permeability, can influence the potency. The selectivity towards certain HDACs isoforms can be explained by special interactions of fluorinated compounds with the structure of the slightly different enzymes. Another aspect is that for a more detailed investigation of newly synthesized fluorine-containing active compounds, fluorination is often used for the purpose of labeling. Aside from the isotope 19F, which can be detected by nuclear magnetic resonance spectroscopy, the positron emission tomography of 18F plays a major role. However, to our best knowledge, a survey of the general effects of fluorination on HDACis development is lacking in the literature to date. Therefore, the aim of this review is to highlight the introduction of fluorine in the course of chemical synthesis and the impact on biological activity, using selected examples of recently developed fluorinated HDACis.
Collapse
Affiliation(s)
- Duong Tien Anh
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 10000, Vietnam
| | - Nguyen Hai Nam
- Department of Pharmaceutical Chemistry, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 10000, Vietnam
| | - Brigitte Kircher
- Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology), Medical University Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria
- Tyrolean Cancer Research Institute, Innrain 66, 6020 Innsbruck, Austria
- Correspondence: (B.K.); (D.B.)
| | - Daniel Baecker
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany
- Correspondence: (B.K.); (D.B.)
| |
Collapse
|
6
|
Everix L, Seane EN, Ebenhan T, Goethals I, Bolcaen J. Introducing HDAC-Targeting Radiopharmaceuticals for Glioblastoma Imaging and Therapy. Pharmaceuticals (Basel) 2023; 16:227. [PMID: 37259375 PMCID: PMC9967489 DOI: 10.3390/ph16020227] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 09/29/2023] Open
Abstract
Despite recent advances in multimodality therapy for glioblastoma (GB) incorporating surgery, radiotherapy, chemotherapy and targeted therapy, the overall prognosis remains poor. One of the interesting targets for GB therapy is the histone deacetylase family (HDAC). Due to their pleiotropic effects on, e.g., DNA repair, cell proliferation, differentiation, apoptosis and cell cycle, HDAC inhibitors have gained a lot of attention in the last decade as anti-cancer agents. Despite their known underlying mechanism, their therapeutic activity is not well-defined. In this review, an extensive overview is given of the current status of HDAC inhibitors for GB therapy, followed by an overview of current HDAC-targeting radiopharmaceuticals. Imaging HDAC expression or activity could provide key insights regarding the role of HDAC enzymes in gliomagenesis, thus identifying patients likely to benefit from HDACi-targeted therapy.
Collapse
Affiliation(s)
- Liesbeth Everix
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, 2610 Antwerpen, Belgium
| | - Elsie Neo Seane
- Department of Medical Imaging and Therapeutic Sciences, Cape Peninsula University of Technology, Cape Town 7530, South Africa
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility (PCIF), (NuMeRI) NPC, Pretoria 0001, South Africa
- Department of Science and Technology/Preclinical Drug Development Platform (PCDDP), North West University, Potchefstroom 2520, South Africa
- Nuclear Medicine, University of Pretoria, Pretoria 0001, South Africa
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Julie Bolcaen
- Radiation Biophysics Division, SSC laboratory, iThemba LABS, Cape Town 7131, South Africa
| |
Collapse
|
7
|
Travis CR, Francis DY, Williams DC, Waters ML. Evaluation of acyllysine isostere interactions with the aromatic pocket of the AF9 YEATS domain. Protein Sci 2023; 32:e4533. [PMID: 36482045 PMCID: PMC9793969 DOI: 10.1002/pro.4533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/21/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
Amide-π interactions, in which an amide interacts with an aromatic group, are ubiquitous in biology, yet remain understudied relative to other noncovalent interactions. Recently, we demonstrated that an electrostatically tunable amide-π interaction is key to recognition of histone acyllysine by the AF9 YEATS domain, a reader protein which has emerged as a therapeutic target due to its dysregulation in cancer. Amide isosteres are commonly employed in drug discovery, often to prevent degradation by proteases, and have proven valuable in achieving selectivity when targeting epigenetic proteins. However, like amide-π interactions, interactions of amide isosteres with aromatic rings have not been thoroughly studied despite widespread use. Herein, we evaluate the recognition of a series of amide isosteres by the AF9 YEATS domain using genetic code expansion to evaluate the amide isostere-π interaction. We show that compared to the amide-π interaction with the native ligand, each isostere exhibits similar electrostatic tunability with an aromatic residue in the binding pocket, demonstrating that the isosteres maintain similar interactions with the aromatic residue. We identify a urea-containing ligand that binds with enhanced affinity for the AF9 YEATS domain, offering a promising starting point for inhibitor development. Furthermore, we demonstrate that carbamate and urea isosteres of crotonyllysine are resistant to enzymatic removal by SIRT1, a protein that cleaves acyl post-translational modifications, further indicating the potential of amide isosteres in YEATS domain inhibitor development. These results also provide experimental precedent for interactions of these common drug discovery moieties with aromatic rings that can inform computational methods.
Collapse
Affiliation(s)
- Christopher R. Travis
- Department of Chemistry, CB 3290University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Denver Y. Francis
- Department of Chemistry, CB 3290University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - David C. Williams
- Department of Pathology and Laboratory Medicine, CB 7525University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| | - Marcey L. Waters
- Department of Chemistry, CB 3290University of North Carolina at Chapel HillChapel HillNorth CarolinaUSA
| |
Collapse
|
8
|
Perrine SA, Alsharif WF, Harutyunyan A, Kamal S, Viola NT, Gelovani JG. Low- and high-cocaine intake affects the spatial and temporal dynamics of class IIa HDAC expression-activity in the nucleus accumbens and hippocampus of male rats as measured by [18F]TFAHA PET/CT neuroimaging. ADDICTION NEUROSCIENCE 2022; 4:100046. [PMID: 36540409 PMCID: PMC9762729 DOI: 10.1016/j.addicn.2022.100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Repeated cocaine alters neuronal function in the nucleus accumbens (NAc), a brain region involved in cocaine taking, and in hippocampus (HC), known for contextual and associative learning. [18F]TFAHA is a histone deacetylase (HDAC) class IIa-specific radiotracer for positron emission tomography (PET)-imaging developed by our group to study epigenetic mechanisms. Here, [18F]TFAHA was used to conduct PET-imaging coupled with computed tomography (CT) of rat brains at baseline and after repeated cocaine intravenous self-administration (cocaine-IVSA) in low-intake versus high-intake cocaine groups. A 3 h-access FR1-schedule of cocaine-IVSA (0.5 mg/kg/infusion) for 12 continuous days was used with male Sprague Dawley rats following jugular vein catheterization. PET/CT neuroimaging with [18F]TFAHA was acquired in a dynamic mode over 40 min post-radiotracer administration at baseline and on day 12 of cocaine-IVSA using a longitudinal, repeated design. This study shows that high-cocaine intake significantly decreases class IIa HDAC expression-activity in NAc, while low-cocaine intake significantly decreases expression-activity in HC in male rats. These findings suggest the individual rats with low-cocaine intake had epigenetic changes in HC, where drug-associative changes occur. Alternatively, individuals with high-cocaine intake had robust epigenetic changes in NAc, where rewared-related behaviors originate. These findings are the first longitudinal data obtained in vivo to implicate class IIa HDACs in the persistent behavioral effects of cocaine. Furthermore, our results are consistent with published research implicating class IIa HDACs in cocaine-induced brain changes and studies suggesting a relationship between an individual's drug-taking behavior and regional pattern of epigenetic changes in the brain.
Collapse
Affiliation(s)
- Shane A. Perrine
- Psychiatry and Behavioral Neurosciences, Wayne State University, 6135 Woodward Avenue, Suite 3119, Detroit, MI, USA
- Research Services, John D. Dingell VAMC, Detroit, MI, USA
| | | | - Arman Harutyunyan
- Psychiatry and Behavioral Neurosciences, Wayne State University, 6135 Woodward Avenue, Suite 3119, Detroit, MI, USA
| | - Swatabdi Kamal
- Biomedical Engineering, Wayne State University, Detroit, MI, USA
| | - Nerissa T. Viola
- Oncology, Wayne State University, Detroit, MI, USA
- Karmanos Cancer Institute, Detroit, MI, USA
| | - Juri G. Gelovani
- Biomedical Engineering, Wayne State University, Detroit, MI, USA
- Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
9
|
Miller GM, Flynn EM, Tom J, Song A, Cochran AG. Trifluoroacetyl Lysine as a Bromodomain Binding Mimic of Lysine Acetylation. ACS Chem Biol 2022; 17:1022-1029. [PMID: 35467836 DOI: 10.1021/acschembio.2c00016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Genetic code expansion has proven invaluable to the elucidation of functions of defined protein modifications through the site-specific incorporation of noncanonical amino acids. The use of nonhydrolyzable derivatives of post-translational modifications can greatly increase site stoichiometry and half-life. Investigating acetyllysine reader domain (bromodomain) interactions with acetylated nonhistone proteins is challenging due to the limited tools available and dynamic nature of this post-translational modification. Here, we demonstrate that bromodomains bind acetyllysine peptides and those substituted with an acetyllysine derivative, trifluoroacetyllysine, with similar affinity and selectivity. Importantly, both trifluoroacetyllysine and acetyllysine can be site-specifically incorporated into proteins expressed in bacterial and mammalian cells, and the strong electron-withdrawing trifluoro substituent makes the latter resistant to deacetylation by sirtuins (SIRTs). The controlled expression of SIRT-resistant, site-specifically acetylated transcription factors expands the set of available tools for determining the function of acetylation, and it serves as a template for investigating bromodomain interactions with acetylated transcription factors.
Collapse
Affiliation(s)
- Gregory M. Miller
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco 94080, California, United States
| | - E. Megan Flynn
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco 94080, California, United States
| | - Jeffrey Tom
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco 94080, California, United States
| | - Aimin Song
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco 94080, California, United States
| | - Andrea G. Cochran
- Department of Early Discovery Biochemistry, Genentech, 1 DNA Way, South San Francisco 94080, California, United States
| |
Collapse
|
10
|
Kamal SR, Potukutchi S, Gelovani DJ, Bonomi RE, Kallakuri S, Cavanaugh JM, Mangner T, Conti A, Liu RS, Pasqualini R, Arap W, Sidman RL, Perrine SA, Gelovani JG. Spatial and temporal dynamics of HDACs class IIa following mild traumatic brain injury in adult rats. Mol Psychiatry 2022; 27:1683-1693. [PMID: 35027678 PMCID: PMC11629393 DOI: 10.1038/s41380-021-01369-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/28/2021] [Accepted: 10/15/2021] [Indexed: 11/09/2022]
Abstract
The fundamental role of epigenetic regulatory mechanisms involved in neuroplasticity and adaptive responses to traumatic brain injury (TBI) is gaining increased recognition. TBI-induced neurodegeneration is associated with several changes in the expression-activity of various epigenetic regulatory enzymes, including histone deacetylases (HDACs). In this study, PET/CT with 6-([18F]trifluoroacetamido)-1- hexanoicanilide ([18F]TFAHA) to image spatial and temporal dynamics of HDACs class IIa expression-activity in brains of adult rats subjected to a weight drop model of diffuse, non-penetrating, mild traumatic brain injury (mTBI). The mTBI model was validated by histopathological and immunohistochemical analyses of brain tissue sections for localization and magnitude of expression of heat-shock protein-70 kDa (HSP70), amyloid precursor protein (APP), cannabinoid receptor-2 (CB2), ionized calcium-binding adapter protein-1 (IBA1), histone deacetylase-4 and -5 (HDAC4 and HDAC5). In comparison to baseline, the expression-activities of HDAC4 and HDAC5 were downregulated in the hippocampus, nucleus accumbens, peri-3rd ventricular part of the thalamus, and substantia nigra at 1-3 days post mTBI, and remained low at 7-8 days post mTBI. Reduced levels of HDAC4 and HDAC5 expression observed in neurons of these brain regions post mTBI were associated with the reduced nuclear and neuropil levels of HDAC4 and HDAC5 with the shift to perinuclear localization of these enzymes. These results support the rationale for the development of therapeutic strategies to upregulate expression-activity of HDACs class IIa post-TBI. PET/CT (MRI) with [18F]TFAHA can facilitate the development and clinical translation of unique therapeutic approaches to upregulate the expression and activity of HDACs class IIa enzymes in the brain after TBI.
Collapse
Affiliation(s)
- Swatabdi R Kamal
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Shreya Potukutchi
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - David J Gelovani
- School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Robin E Bonomi
- School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Srinivasu Kallakuri
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - John M Cavanaugh
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Thomas Mangner
- Cyclotron-Radiochemistry Facility, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alana Conti
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Departments of Neurosurgery and Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Ren-Shyan Liu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Nuclear Medicine, Cheng-Hsin General Hospital, Taipei, 112, Taiwan
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Renata Pasqualini
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers Cancer Institute of New Jersey, Newark, NJ, 07103, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, 07103, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA.
- Molecular Imaging Program, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA.
- College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
11
|
Daśko M, de Pascual-Teresa B, Ortín I, Ramos A. HDAC Inhibitors: Innovative Strategies for Their Design and Applications. Molecules 2022; 27:molecules27030715. [PMID: 35163980 PMCID: PMC8837987 DOI: 10.3390/molecules27030715] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Histone deacetylases (HDACs) are a large family of epigenetic metalloenzymes that are involved in gene transcription and regulation, cell proliferation, differentiation, migration, and death, as well as angiogenesis. Particularly, disorders of the HDACs expression are linked to the development of many types of cancer and neurodegenerative diseases, making them interesting molecular targets for the design of new efficient drugs and imaging agents that facilitate an early diagnosis of these diseases. Thus, their selective inhibition or degradation are the basis for new therapies. This is supported by the fact that many HDAC inhibitors (HDACis) are currently under clinical research for cancer therapy, and the Food and Drug Administration (FDA) has already approved some of them. In this review, we will focus on the recent advances and latest discoveries of innovative strategies in the development and applications of compounds that demonstrate inhibitory or degradation activity against HDACs, such as PROteolysis-TArgeting Chimeras (PROTACs), tumor-targeted HDACis (e.g., folate conjugates and nanoparticles), and imaging probes (positron emission tomography (PET) and fluorescent ligands).
Collapse
Affiliation(s)
- Mateusz Daśko
- Department of Inorganic Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Narutowicza 11/12, 80-233 Gdańsk, Poland;
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28925 Alcorcón, Spain;
| | - Beatriz de Pascual-Teresa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28925 Alcorcón, Spain;
| | - Irene Ortín
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28925 Alcorcón, Spain;
- Correspondence: (I.O.); (A.R.)
| | - Ana Ramos
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28925 Alcorcón, Spain;
- Correspondence: (I.O.); (A.R.)
| |
Collapse
|
12
|
Turkman N, Liu D, Pirola I. Design, synthesis, biochemical evaluation, radiolabeling and in vivo imaging with high affinity class-IIa histone deacetylase inhibitor for molecular imaging and targeted therapy. Eur J Med Chem 2022; 228:114011. [PMID: 34875522 PMCID: PMC8919062 DOI: 10.1016/j.ejmech.2021.114011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 11/17/2022]
Abstract
Herein, we describe the design, synthesis and deciphering of the key characteristics of the structure activity relationship (SAR) of trifluoromethyloxadiazole (TFMO) bearing class-IIa HDAC inhibitors. Our medicinal chemistry campaign of 23 compounds identified compound 1 as a highly potent inhibitor with sub nM affinity to class-IIa HDAC4 isoform. Therefore, We radiolabeled compound 1 (named thereafter as NT160) with [18F]fluoride thus producing the identical [18F]-NT160 as a diagnostic tool for positron emission tomography (PET). [18F]-NT160 was produced in high radiochemical purity (>95%), moderate radiochemical yield (2−5%) and moderate molar activity in the range of 0.30−0.85 GBq/umol (8.0−23.0 mCi/umol). We also established that [18F]-NT160 can cross the blood brain barrier and bind to class-IIa HDACs in vivo. The combination of [18F]-NT160 and 1 represent a novel theranostic pair using the same molecule to enable diagnostic PET imaging with [18F]-NT160 followed by targeted therapy with NT160.
Collapse
Affiliation(s)
- Nashaat Turkman
- Stony Brook Cancer Center, Stony Brook, Long Island, NY, USA; Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY, USA.
| | - Daxing Liu
- Stony Brook Cancer Center, Stony Brook, Long Island, NY, USA; Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY, USA
| | - Isabella Pirola
- Stony Brook Cancer Center, Stony Brook, Long Island, NY, USA; Department of Radiology, School of Medicine, Stony Brook University, Long Island, NY, USA
| |
Collapse
|
13
|
Evaluation of Class IIa Histone Deacetylases Expression and In Vivo Epigenetic Imaging in a Transgenic Mouse Model of Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22168633. [PMID: 34445342 PMCID: PMC8395513 DOI: 10.3390/ijms22168633] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022] Open
Abstract
Epigenetic regulation by histone deacetylase (HDAC) is associated with synaptic plasticity and memory formation, and its aberrant expression has been linked to cognitive disorders, including Alzheimer's disease (AD). This study aimed to investigate the role of class IIa HDAC expression in AD and monitor it in vivo using a novel radiotracer, 6-(tri-fluoroacetamido)-1-hexanoicanilide ([18F]TFAHA). A human neural cell culture model with familial AD (FAD) mutations was established and used for in vitro assays. Positron emission tomography (PET) imaging with [18F]TFAHA was performed in a 3xTg AD mouse model for in vivo evaluation. The results showed a significant increase in HDAC4 expression in response to amyloid-β (Aβ) deposition in the cell model. Moreover, treatment with an HDAC4 selective inhibitor significantly upregulated the expression of neuronal memory-/synaptic plasticity-related genes. In [18F]TFAHA-PET imaging, whole brain or regional uptake was significantly higher in 3xTg AD mice compared with WT mice at 8 and 11 months of age. Our study demonstrated a correlation between class IIa HDACs and Aβs, the therapeutic benefit of a selective inhibitor, and the potential of using [18F]TFAHA as an epigenetic radiotracer for AD, which might facilitate the development of AD-related neuroimaging approaches and therapies.
Collapse
|
14
|
Laws MT, Bonomi RE, Gelovani DJ, Llaniguez J, Lu X, Mangner T, Gelovani JG. Noninvasive quantification of SIRT1 expression-activity and pharmacologic inhibition in a rat model of intracerebral glioma using 2-[ 18F]BzAHA PET/CT/MRI. Neurooncol Adv 2020; 2:vdaa006. [PMID: 32118205 PMCID: PMC7034639 DOI: 10.1093/noajnl/vdaa006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Several studies demonstrated that glioblastoma multiforme progression and recurrence is linked to epigenetic regulatory mechanisms. Sirtuin 1 (SIRT1) plays an important role in glioma progression, invasion, and treatment response and is a potential therapeutic target. The aim of this study is to test the feasibility of 2-[18F]BzAHA for quantitative imaging of SIRT1 expression–activity and monitoring pharmacologic inhibition in a rat model of intracerebral glioma. Methods Sprague Dawley rats bearing 9L (N = 12) intracerebral gliomas were injected with 2-[18F]BzAHA (300–500 µCi/animal i.v.) and dynamic positron-emission tomography (PET) imaging was performed for 60 min. Then, SIRT1 expression in 9L tumors (N = 6) was studied by immunofluorescence microscopy (IF). Two days later, rats with 9L gliomas were treated either with SIRT1 specific inhibitor EX-527 (5 mg/kg, i.p.; N = 3) or with histone deacetylases class IIa specific inhibitor MC1568 (30 mg/kg, i.p.; N = 3) and 30 min later were injected i.v. with 2-[18F]BzAHA. PET-computerized tomography-magnetic resonance (PET/CT/MR) images acquired after EX-527 and MC1568 treatments were co-registered with baseline images. Results Standard uptake values (SUVs) of 2-[18F]BzAHA in 9L tumors measured at 20 min post-radiotracer administration were 1.11 ± 0.058 and had a tumor-to-brainstem SUV ratio of 2.73 ± 0.141. IF of 9L gliomas revealed heterogeneous upregulation of SIRT1, especially in hypoxic and peri-necrotic regions. Significant reduction in 2-[18F]BzAHA SUV and distribution volume in 9L tumors was observed after administration of EX-527, but not MC1568. Conclusions PET/CT/MRI with 2-[18F]BzAHA can facilitate studies to elucidate the roles of SIRT1 in gliomagenesis and progression, as well as to optimize therapeutic doses of novel SIRT1 inhibitors.
Collapse
Affiliation(s)
- Maxwell T Laws
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Robin E Bonomi
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - David J Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Jeremy Llaniguez
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Xin Lu
- Positron Emission Tomography Center, Wayne State University, Detroit, Michigan, USA
| | - Thomas Mangner
- Positron Emission Tomography Center, Wayne State University, Detroit, Michigan, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, Michigan, USA.,Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA.,Molecular Imaging Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
15
|
Molecular imaging HDACs class IIa expression-activity and pharmacologic inhibition in intracerebral glioma models in rats using PET/CT/(MRI) with [ 18F]TFAHA. Sci Rep 2019; 9:3595. [PMID: 30837601 PMCID: PMC6401080 DOI: 10.1038/s41598-019-40054-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022] Open
Abstract
HDAC class IIa enzymes (HDAC4, 5, 7, 9) are important for glioma progression, invasion, responses to TMZ and radiotherapy, and prognosis. In this study, we demonstrated the efficacy of PET/CT/(MRI) with [18F]TFAHA for non-invasive and quantitative imaging of HDAC class IIa expression-activity in intracerebral 9L and U87-MG gliomas in rats. Increased accumulation of [18F]TFAHA in 9L and U87-MG tumors was observed at 20 min post radiotracer administration with SUV of 1.45 ± 0.05 and 1.08 ± 0.05, respectively, and tumor-to-cortex SUV ratios of 1.74 ± 0.07 and 1.44 ± 0.03, respectively. [18F]TFAHA accumulation was also observed in normal brain structures known to overexpress HDACs class IIa: hippocampus, n.accumbens, PAG, and cerebellum. These results were confirmed by immunohistochemical staining of brain tissue sections revealing the upregulation of HDACs 4, 5, and 9, and HIF-1α, hypoacetylation of H2AK5ac, H2BK5ac, H3K9ac, H4K8ac, and downregulation of KLF4. Significant reduction in [18F]TFAHA accumulation in 9L tumors was observed after administration of HDACs class IIa specific inhibitor MC1568, but not the SIRT1 specific inhibitor EX-527. Thus, PET/CT/(MRI) with [18F]TFAHA can facilitate studies to elucidate the roles of HDAC class IIa enzymes in gliomagenesis and progression and to optimize therapeutic doses of novel HDACs class IIa inhibitors in gliomas.
Collapse
|
16
|
A Novel Substrate Radiotracer for Molecular Imaging of SIRT2 Expression and Activity with Positron Emission Tomography. Mol Imaging Biol 2019; 20:594-604. [PMID: 29423902 PMCID: PMC6816246 DOI: 10.1007/s11307-017-1149-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE The purpose of this study was to develop a SIRT2-specific substrate-type radiotracer for non-invasive PET imaging of epigenetic regulatory processes mediated by SIRT2 in normal and disease tissues. PROCEDURES A library of compounds containing tert-butyloxycarbonyl-lysine-aminomethylcoumarin backbone was derivatized with fluoroalkyl chains 3-16 carbons in length. SIRT2 most efficiently cleaved the myristoyl, followed by 12-fluorododecanoic and 10-fluorodecanoic groups (Kcat/Km 716.5 ± 72.8, 615.4 ± 50.5, 269.5 ± 52.1/s mol, respectively). Radiosynthesis of 12- [18F]fluorododecanoic aminohexanoicanilide (12-[18F]DDAHA) was achieved by nucleophilic radiofluorination of 12-iododecanoic-AHA precursor. RESULTS A significantly higher accumulation of 12-[18F]DDAHA was observed in MCF-7 and MDA-MB-435 cells in vitro as compared to U87, MiaPaCa, and MCF10A, which was consistent with levels of SIRT2 expression. Initial in vivo studies using 12-[18F]DDAHA conducted in a 9L glioma-bearing rats were discouraging, due to rapid defluorination of this radiotracer upon intravenous administration, as evidenced by significant accumulation of F-18 radioactivity in the skull and other bones, which confounded the interpretation of images of radiotracer accumulation within the tumor and other regions of the brain. CONCLUSIONS The next generation of SIRT2-specific radiotracers resistant to systemic defluorination should be developed using alternative sites of radiofluorination on the aliphatic chain of DDAHA. A SIRT2-selective radiotracer may provide information about SIRT2 expression and activity in tumors and normal organs and tissues, which may help to better understand the roles of SIRT2 in different diseases.
Collapse
|
17
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
18
|
Bonomi R, Popov V, Laws MT, Gelovani D, Majhi A, Shavrin A, Lu X, Muzik O, Turkman N, Liu R, Mangner T, Gelovani JG. Molecular Imaging of Sirtuin1 Expression-Activity in Rat Brain Using Positron-Emission Tomography-Magnetic-Resonance Imaging with [ 18F]-2-Fluorobenzoylaminohexanoicanilide. J Med Chem 2018; 61:7116-7130. [PMID: 30052441 DOI: 10.1021/acs.jmedchem.8b00253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sirtuin 1 (SIRT1) is a class III histone deacetylase that plays significant roles in the regulation of lifespan, metabolism, memory, and circadian rhythms and in the mechanisms of many diseases. However, methods of monitoring the pharmacodynamics of SIRT1-targeted drugs are limited to blood sampling because of the invasive nature of biopsies. For the noninvasive monitoring of the spatial and temporal dynamics of SIRT1 expression-activity in vivo by PET-CT-MRI, we developed a novel substrate-type radiotracer, [18F]-2-fluorobenzoylaminohexanoicanilide (2-[18F]BzAHA). PET-CT-MRI studies in rats demonstrated increased accumulation of 2-[18F]BzAHA-derived radioactivity in the hypothalamus, hippocampus, nucleus accumbens, and locus coeruleus, consistent with autoradiographic and immunofluorescent (IMF) analyses of brain-tissue sections. Pretreatment with the SIRT1 specific inhibitor, EX-527 (5 mg/kg, ip), resulted in about a 20% reduction of 2-[18F]BzAHA-derived-radioactivity accumulation in these structures. In vivo imaging of SIRT1 expression-activity should facilitate studies that improve the understanding of SIRT1-mediated regulation in the brain and aid in the development and clinical translation of SIRT1-targeted therapies.
Collapse
Affiliation(s)
- Robin Bonomi
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | - Vadim Popov
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | - Maxwell T Laws
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | - David Gelovani
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | - Anjoy Majhi
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | - Aleksandr Shavrin
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | | | | | - Nashaat Turkman
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| | - Renshyan Liu
- National Taiwan University , Taipei City 10617 , Taiwan
| | | | - Juri G Gelovani
- Karmanos Cancer Institute , Detroit , Michigan 48202 , United States
| |
Collapse
|
19
|
Abstract
Hydrolytic enzymes are a large class of biological catalysts that play a vital role in a plethora of critical biochemical processes required to maintain human health. However, the expression and/or activity of these important enzymes can change in many different diseases and therefore represent exciting targets for the development of positron emission tomography (PET) and single-photon emission computed tomography (SPECT) radiotracers. This review focuses on recently reported radiolabeled substrates, reversible inhibitors, and irreversible inhibitors investigated as PET and SPECT tracers for imaging hydrolytic enzymes. By learning from the most successful examples of tracer development for hydrolytic enzymes, it appears that an early focus on careful enzyme kinetics and cell-based studies are key factors for identifying potentially useful new molecular imaging agents.
Collapse
Affiliation(s)
- Brian P Rempel
- 1 Department of Science, Augustana Faculty, University of Alberta, Edmonton, Alberta, Canada
| | - Eric W Price
- 2 Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Christopher P Phenix
- 2 Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,3 Biomarker Discovery, Thunder Bay Regional Health Research Institute, Thunder Bay, Ontario, Canada
| |
Collapse
|
20
|
Kim GS, Smith AK, Nievergelt CM, Uddin M. Neuroepigenetics of Post-Traumatic Stress Disorder. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 158:227-253. [PMID: 30072055 PMCID: PMC6474244 DOI: 10.1016/bs.pmbts.2018.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While diagnosis of PTSD is based on behavioral symptom clusters that are most directly associated with brain function, epigenetic studies of PTSD in humans to date have been limited to peripheral tissues. Animal models of PTSD have been key for understanding the epigenetic alterations in the brain most directly relevant to endophenotypes of PTSD, in particular those pertaining to fear memory and stress response. This chapter provides an overview of neuroepigenetic studies based on animal models of PTSD, with an emphasis on the effect of stress on fear memory. Where relevant, we also describe human-based studies with relevance to neuroepigenetic insights gleaned from animal work and suggest promising directions for future studies of PTSD neuroepigenetics in living humans that combine peripheral epigenetic measures with measures of central nervous system activity, structure and function.
Collapse
Affiliation(s)
- Grace S Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Medical Scholars Program, University of Illinois College of Medicine, Urbana, IL, United States
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, United States; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, United States
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States
| | - Monica Uddin
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Department of Psychology, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
21
|
Kuchar M, Neuber C, Belter B, Bergmann R, Lenk J, Wodtke R, Kniess T, Steinbach J, Pietzsch J, Löser R. Evaluation of Fluorine-18-Labeled α1(I)-N-Telopeptide Analogs as Substrate-Based Radiotracers for PET Imaging of Melanoma-Associated Lysyl Oxidase. Front Chem 2018; 6:121. [PMID: 29755969 PMCID: PMC5932954 DOI: 10.3389/fchem.2018.00121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/30/2018] [Indexed: 12/28/2022] Open
Abstract
Accumulating evidence suggests an unequivocal role of lysyl oxidases as key players of tumor progression and metastasis, which renders this enzyme family highly attractive for targeted non-invasive functional imaging of tumors. Considering their function in matrix remodeling, malignant melanoma appears as particularly interesting neoplasia in this respect. For the development of radiotracers that enable PET imaging of the melanoma-associated lysyl oxidase activity, substrates derived from the type I collagen α1 N-telopeptide were labeled with fluorine-18 using N-succinimidyl 4-[18F]fluorobenzoate ([18F]SFB) as prosthetic reagent. With regards to potential crosslinking to tumor-associated collagen in vivo, their interaction with triple-helical type I collagen was studied by SPR. A mouse model of human melanoma was established on the basis of the A375 cell line, for which the expression of the oncologically relevant lysyl oxidase isoforms LOX and LOXL2 was demonstrated in Western blot and immunohistochemical experiments. The radiopharmacological profiles of the peptidic radiotracers were evaluated in normal rats and A375 melanoma-bearing mice by ex vivo metabolite analysis, whole-body biodistribution studies and dynamic PET imaging. Out of three 18F-labeled telopeptide analogs, the one with the most favorable substrate properties has shown favorable tumor uptake and tumor-to-muscle ratio. Lysyl oxidase-mediated tumor uptake was proven by pharmacological inhibition using β-aminopropionitrile and by employing negative-control analogs of impeded or abolished targeting capability. The latter were obtained by substituting the lysine residue by ornithine and norleucine, respectively. Comparing the tumor uptake of the lysine-containing peptide with that of the non-functional analogs indicate the feasibility of lysyl oxidase imaging in melanoma using substrate-based radiotracers.
Collapse
Affiliation(s)
- Manuela Kuchar
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Birgit Belter
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jens Lenk
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Robert Wodtke
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Torsten Kniess
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| |
Collapse
|
22
|
Tago T, Toyohara J. Advances in the Development of PET Ligands Targeting Histone Deacetylases for the Assessment of Neurodegenerative Diseases. Molecules 2018; 23:E300. [PMID: 29385079 PMCID: PMC6017260 DOI: 10.3390/molecules23020300] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 12/05/2022] Open
Abstract
Epigenetic alterations of gene expression have emerged as a key factor in several neurodegenerative diseases. In particular, inhibitors targeting histone deacetylases (HDACs), which are enzymes responsible for deacetylation of histones and other proteins, show therapeutic effects in animal neurodegenerative disease models. However, the details of the interaction between changes in HDAC levels in the brain and disease progression remain unknown. In this review, we focus on recent advances in development of radioligands for HDAC imaging in the brain with positron emission tomography (PET). We summarize the results of radiosynthesis and biological evaluation of the HDAC ligands to identify their successful results and challenges. Since 2006, several small molecules that are radiolabeled with a radioisotope such as carbon-11 or fluorine-18 have been developed and evaluated using various assays including in vitro HDAC binding assays and PET imaging in rodents and non-human primates. Although most compounds do not readily cross the blood-brain barrier, adamantane-conjugated radioligands tend to show good brain uptake. Until now, only one HDAC radioligand has been tested clinically in a brain PET study. Further PET imaging studies to clarify age-related and disease-related changes in HDACs in disease models and humans will increase our understanding of the roles of HDACs in neurodegenerative diseases.
Collapse
Affiliation(s)
- Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan.
| | - Jun Toyohara
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo 173-0015, Japan.
| |
Collapse
|