1
|
Zhang ZL, Xu HN, Gong CM, Li YZ, Song XM, Li YM, Zhang DD, Wang R. Microorganism-Derived Bisindole Alkaloids With Anticancer Potential and Their Mechanisms: A Comprehensive Review. Chem Biodivers 2024:e202402398. [PMID: 39714457 DOI: 10.1002/cbdv.202402398] [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: 09/23/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Bisindole alkaloids constitute a significant class of natural compounds distinguished by their characteristic bisindole structure and renowned for their anticancer properties. Over the last six decades, researchers have isolated 425 microorganism-derived bisindole alkaloids (MDBAs). Among them, 187 MDBAs have demonstrated anticancer properties against various in vitro cancer cell lines, primarily by impeding the cell cycle, restraining cell proliferation, and inducing apoptosis and autophagy. These effects are mediated by regulating key targets and signaling pathways such as hypoxia-inducible factor (HIF)-1, MAPK, and phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR. This review provides a comprehensive examination of the sources, chemical diversity, and anticancer properties of these compounds. Furthermore, it summarizes the structure-activity relationship (SAR), druggability, and the mechanisms underlying MDBAs' anticancer effects. Ultimately, this article aims to furnish a thorough overview of the advancements in the investigation of microorganism-derived bisindole alkaloids for their continued development and utilization.
Collapse
Affiliation(s)
- Zi-Long Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Hao-Nan Xu
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Chuan-Ming Gong
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Yu-Ze Li
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Xiao-Mei Song
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Yi-Ming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Dong-Dong Zhang
- School of Pharmacy, Shaanxi Key Laboratory of Research and Application of "Taibai Qi Yao", Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| | - Rui Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
2
|
Xin DE, Liao Y, Rao R, Ogurek S, Sengupta S, Xin M, Bayat AE, Seibel WL, Graham RT, Koschmann C, Lu QR. Chaetocin-mediated SUV39H1 inhibition targets stemness and oncogenic networks of diffuse midline gliomas and synergizes with ONC201. Neuro Oncol 2024; 26:735-748. [PMID: 38011799 PMCID: PMC10995509 DOI: 10.1093/neuonc/noad222] [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: 05/22/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Diffuse intrinsic pontine gliomas (DIPG/DMG) are devastating pediatric brain tumors with extraordinarily limited treatment options and uniformly fatal prognosis. Histone H3K27M mutation is a common recurrent alteration in DIPG and disrupts epigenetic regulation. We hypothesize that genome-wide H3K27M-induced epigenetic dysregulation makes tumors vulnerable to epigenetic targeting. METHODS We performed a screen of compounds targeting epigenetic enzymes to identify potential inhibitors for the growth of patient-derived DIPG cells. We further carried out transcriptomic and genomic landscape profiling including RNA-seq and CUT&RUN-seq as well as shRNA-mediated knockdown to assess the effects of chaetocin and SUV39H1, a target of chaetocin, on DIPG growth. RESULTS High-throughput small-molecule screening identified an epigenetic compound chaetocin as a potent blocker of DIPG cell growth. Chaetocin treatment selectively decreased proliferation and increased apoptosis of DIPG cells and significantly extended survival in DIPG xenograft models, while restoring H3K27me3 levels. Moreover, the loss of H3K9 methyltransferase SUV39H1 inhibited DIPG cell growth. Transcriptomic and epigenomic profiling indicated that SUV39H1 loss or inhibition led to the downregulation of stemness and oncogenic networks including growth factor receptor signaling and stemness-related programs; however, D2 dopamine receptor (DRD2) signaling adaptively underwent compensatory upregulation conferring resistance. Consistently, a combination of chaetocin treatment with a DRD2 antagonist ONC201 synergistically increased the antitumor efficacy. CONCLUSIONS Our studies reveal a therapeutic vulnerability of DIPG cells through targeting the SUV39H1-H3K9me3 pathway and compensatory signaling loops for treating this devastating disease. Combining SUV39H1-targeting chaetocin with other agents such as ONC201 may offer a new strategy for effective DIPG treatment.
Collapse
Affiliation(s)
- Dazhuan Eric Xin
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Yunfei Liao
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rohit Rao
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sean Ogurek
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Mei Xin
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Arman Esshaghi Bayat
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - William L Seibel
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Richard T Graham
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Q Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati and Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
3
|
Goher SS, Abdrabo WS, Veerakanellore GB, Elgendy B. 2,5-Diketopiperazines (DKPs): Promising Scaffolds for Anticancer Agents. Curr Pharm Des 2024; 30:597-623. [PMID: 38343054 DOI: 10.2174/0113816128291798240201112916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/22/2024] [Indexed: 05/25/2024]
Abstract
2,5-Diketopiperazine (2,5-DKP) derivatives represent a family of secondary metabolites widely produced by bacteria, fungi, plants, animals, and marine organisms. Many natural products with DKP scaffolds exhibited various pharmacological activities such as antiviral, antifungal, antibacterial, and antitumor. 2,5-DKPs are recognized as privileged structures in medicinal chemistry, and compounds that incorporate the 2,5-DKP scaffold have been extensively investigated for their anticancer properties. This review is a thorough update on the anti-cancer activity of natural and synthesized 2,5-DKPs from 1997 to 2022. We have explored various aspects of 2,5-DKPs modifications and summarized their structure-activity relationships (SARs) to gain insight into their anticancer activities. We have also highlighted the novel approaches to enhance the specificity and pharmacokinetics of 2,5-DKP-based anticancer agents.
Collapse
Affiliation(s)
- Shaimaa S Goher
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
- Nanotechnology Research Centre (NTRC), The British University in Egypt (BUE), Suez Desert Road, El Sherouk City, Cairo 1183, Egypt
| | - Wessam S Abdrabo
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Giri Babu Veerakanellore
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Bahaa Elgendy
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
4
|
Wang L, Jiang Q, Chen S, Wang S, Lu J, Gao X, Zhang D, Jin X. Natural epidithiodiketopiperazine alkaloids as potential anticancer agents: Recent mechanisms of action, structural modification, and synthetic strategies. Bioorg Chem 2023; 137:106642. [PMID: 37276722 DOI: 10.1016/j.bioorg.2023.106642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/18/2023] [Accepted: 05/27/2023] [Indexed: 06/07/2023]
Abstract
Cancer has become a grave health crisis that threatens the lives of millions of people worldwide. Because of the drawbacks of the available anticancer drugs, the development of novel and efficient anticancer agents should be encouraged. Epidithiodiketopiperazine (ETP) alkaloids with a 2,5-diketopiperazine (DKP) ring equipped with transannular disulfide or polysulfide bridges or S-methyl moieties constitute a special subclass of fungal natural products. Owing to their privileged sulfur units and intriguing architectural structures, ETP alkaloids exhibit excellent anticancer activities by regulating multiple cancer proteins/signaling pathways, including HIF-1, NF-κB, NOTCH, Wnt, and PI3K/AKT/mTOR, or by inducing cell-cycle arrest, apoptosis, and autophagy. Furthermore, a series of ETP alkaloid derivatives obtained via structural modification showed more potent anticancer activity than natural ETP alkaloids. To solve supply difficulties from natural resources, the total synthetic routes for several ETP alkaloids have been designed. In this review, we summarized several ETP alkaloids with anticancer properties with particular emphasis on their underlying mechanisms of action, structural modifications, and synthetic strategies, which will offer guidance to design and innovate potential anticancer drugs.
Collapse
Affiliation(s)
- Lin Wang
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qinghua Jiang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Siyu Chen
- China Medical University-Queen's University of Belfast Joint College, China Medical University, Shenyang 110122, China
| | - Siyi Wang
- The 1st Clinical Department, China Medical University, Shenyang 110122, China
| | - Jingyi Lu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xun Gao
- Jiangsu Institute Marine Resources Development, Jiangsu Ocean University, Lianyungang 222005, China
| | - Dongfang Zhang
- School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
5
|
Agrawal-Singh S, Bagri J, Giotopoulos G, Azazi DMA, Horton SJ, Lopez CK, Anand S, Bach AS, Stedham F, Antrobus R, Houghton JW, Vassiliou GS, Sasca D, Yun H, Whetton AD, Huntly BJP. HOXA9 forms a repressive complex with nuclear matrix-associated protein SAFB to maintain acute myeloid leukemia. Blood 2023; 141:1737-1754. [PMID: 36577137 PMCID: PMC10113176 DOI: 10.1182/blood.2022016528] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 11/07/2022] [Accepted: 11/28/2022] [Indexed: 12/29/2022] Open
Abstract
HOXA9 is commonly upregulated in acute myeloid leukemia (AML), in which it confers a poor prognosis. Characterizing the protein interactome of endogenous HOXA9 in human AML, we identified a chromatin complex of HOXA9 with the nuclear matrix attachment protein SAFB. SAFB perturbation phenocopied HOXA9 knockout to decrease AML proliferation, increase differentiation and apoptosis in vitro, and prolong survival in vivo. Integrated genomic, transcriptomic, and proteomic analyses further demonstrated that the HOXA9-SAFB (H9SB)-chromatin complex associates with nucleosome remodeling and histone deacetylase (NuRD) and HP1γ to repress the expression of factors associated with differentiation and apoptosis, including NOTCH1, CEBPδ, S100A8, and CDKN1A. Chemical or genetic perturbation of NuRD and HP1γ-associated catalytic activity also triggered differentiation, apoptosis, and the induction of these tumor-suppressive genes. Importantly, this mechanism is operative in other HOXA9-dependent AML genotypes. This mechanistic insight demonstrates the active HOXA9-dependent differentiation block as a potent mechanism of disease maintenance in AML that may be amenable to therapeutic intervention by targeting the H9SB interface and/or NuRD and HP1γ activity.
Collapse
Affiliation(s)
- Shuchi Agrawal-Singh
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Jaana Bagri
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - George Giotopoulos
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Dhoyazan M A Azazi
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah J Horton
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Cecile K Lopez
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Shubha Anand
- Cancer Molecular Diagnostics Laboratory, Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Anne-Sophie Bach
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Frances Stedham
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Jack W Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - George S Vassiliou
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Daniel Sasca
- Department of Hematology, Oncology and Pneumology, University Medical Center Mainz, Mainz, Germany
| | - Haiyang Yun
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - Anthony D Whetton
- School of Veterinary Medicine, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, United Kingdom
| | - Brian J P Huntly
- Wellcome Trust-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Han L, Lee JB, Indermaur EW, Keung AJ. Chaetocin disrupts the SUV39H1-HP1 interaction independent of SUV39H1 methyltransferase activity. Biochem J 2023; 480:421-432. [PMID: 36896918 PMCID: PMC11127023 DOI: 10.1042/bcj20220528] [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: 10/19/2022] [Revised: 02/13/2023] [Accepted: 03/10/2023] [Indexed: 03/11/2023]
Abstract
Chemical tools to control the activities and interactions of chromatin components have broad impact on our understanding of cellular and disease processes. It is important to accurately identify their molecular effects to inform clinical efforts and interpretations of scientific studies. Chaetocin is a widely used chemical that decreases H3K9 methylation in cells. It is frequently attributed as a specific inhibitor of the histone methyltransferase activities of SUV39H1/SU(VAR)3-9, although prior observations showed chaetocin likely inhibits methyltransferase activity through covalent mechanisms involving its epipolythiodixopiperazine disulfide 'warhead' functionality. The continued use of chaetocin in scientific studies may derive from the net effect of reduced H3K9 methylation, irrespective of a direct or indirect mechanism. However, there may be other molecular impacts of chaetocin on SUV39H1 besides inhibition of H3K9 methylation levels that could confound the interpretation of past and future experimental studies. Here, we test a new hypothesis that chaetocin may have an additional downstream impact aside from inhibition of methyltransferase activity. Using a combination of truncation mutants, a yeast two-hybrid system, and direct in vitro binding assays, we show that the human SUV39H1 chromodomain (CD) and HP1 chromoshadow domain (CSD) directly interact. Chaetocin inhibits this binding interaction through its disulfide functionality with some specificity by covalently binding with the CD of SUV39H1, whereas the histone H3-HP1 interaction is not inhibited. Given the key role of HP1 dimers in driving a feedback cascade to recruit SUV39H1 and to establish and stabilize constitutive heterochromatin, this additional molecular consequence of chaetocin should be broadly considered.
Collapse
Affiliation(s)
- Linna Han
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695-7905, U.S.A
| | - Jessica B. Lee
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695-7905, U.S.A
| | - Elaine W. Indermaur
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695-7905, U.S.A
| | - Albert J. Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695-7905, U.S.A
| |
Collapse
|
7
|
Urwanisch L, Unger MS, Sieberer H, Dang HH, Neuper T, Regl C, Vetter J, Schaller S, Winkler SM, Kerschbamer E, Weichenberger CX, Krenn PW, Luciano M, Pleyer L, Greil R, Huber CG, Aberger F, Horejs-Hoeck J. The Class IIA Histone Deacetylase (HDAC) Inhibitor TMP269 Downregulates Ribosomal Proteins and Has Anti-Proliferative and Pro-Apoptotic Effects on AML Cells. Cancers (Basel) 2023; 15:cancers15041039. [PMID: 36831382 PMCID: PMC9953883 DOI: 10.3390/cancers15041039] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized by altered myeloid progenitor cell proliferation and differentiation. As in many other cancers, epigenetic transcriptional repressors such as histone deacetylases (HDACs) are dysregulated in AML. Here, we investigated (1) HDAC gene expression in AML patients and in different AML cell lines and (2) the effect of treating AML cells with the specific class IIA HDAC inhibitor TMP269, by applying proteomic and comparative bioinformatic analyses. We also analyzed cell proliferation, apoptosis, and the cell-killing capacities of TMP269 in combination with venetoclax compared to azacitidine plus venetoclax, by flow cytometry. Our results demonstrate significantly overexpressed class I and class II HDAC genes in AML patients, a phenotype which is conserved in AML cell lines. In AML MOLM-13 cells, TMP269 treatment downregulated a set of ribosomal proteins which are overexpressed in AML patients at the transcriptional level. TMP269 showed anti-proliferative effects and induced additive apoptotic effects in combination with venetoclax. We conclude that TMP269 exerts anti-leukemic activity when combined with venetoclax and has potential as a therapeutic drug in AML.
Collapse
Affiliation(s)
- Laura Urwanisch
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Michael Stefan Unger
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Helene Sieberer
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Hieu-Hoa Dang
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Christof Regl
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Julia Vetter
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Softwarepark 11, 4232 Hagenberg im Muehlkreis, Austria
| | - Susanne Schaller
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Softwarepark 11, 4232 Hagenberg im Muehlkreis, Austria
| | - Stephan M. Winkler
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Softwarepark 11, 4232 Hagenberg im Muehlkreis, Austria
| | - Emanuela Kerschbamer
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via A. Volta 21, 39100 Bolzano, Italy
| | - Christian X. Weichenberger
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Via A. Volta 21, 39100 Bolzano, Italy
| | - Peter W. Krenn
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Michela Luciano
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Lisa Pleyer
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- IIIrd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, 5020 Salzburg, Austria
| | - Richard Greil
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- IIIrd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Oncologic Center, Paracelsus Medical University, 5020 Salzburg, Austria
- Salzburg Cancer Research Institute with Laboratory of Immunological and Molecular Cancer Research and Center for Clinical Cancer and Immunology Trials, 5020 Salzburg, Austria
| | - Christian G. Huber
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Fritz Aberger
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, University of Salzburg, 5020 Salzburg, Austria
- Cancer Cluster Salzburg (CCS), 5020 Salzburg, Austria
- Correspondence: ; Tel.: +43-(0)662-8044-5709
| |
Collapse
|
8
|
Bouyahya A, El Omari N, Bakha M, Aanniz T, El Menyiy N, El Hachlafi N, El Baaboua A, El-Shazly M, Alshahrani MM, Al Awadh AA, Lee LH, Benali T, Mubarak MS. Pharmacological Properties of Trichostatin A, Focusing on the Anticancer Potential: A Comprehensive Review. Pharmaceuticals (Basel) 2022; 15:ph15101235. [PMID: 36297347 PMCID: PMC9612318 DOI: 10.3390/ph15101235] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/12/2022] [Accepted: 09/23/2022] [Indexed: 11/05/2022] Open
Abstract
Trichostatin A (TSA), a natural derivative of dienohydroxamic acid derived from a fungal metabolite, exhibits various biological activities. It exerts antidiabetic activity and reverses high glucose levels caused by the downregulation of brain-derived neurotrophic factor (BDNF) expression in Schwann cells, anti-inflammatory activity by suppressing the expression of various cytokines, and significant antioxidant activity by suppressing oxidative stress through multiple mechanisms. Most importantly, TSA exhibits potent inhibitory activity against different types of cancer through different pathways. The anticancer activity of TSA appeared in many in vitro and in vivo investigations that involved various cell lines and animal models. Indeed, TSA exhibits anticancer properties alone or in combination with other drugs used in chemotherapy. It induces sensitivity of some human cancers toward chemotherapeutical drugs. TSA also exhibits its action on epigenetic modulators involved in cell transformation, and therefore it is considered an epidrug candidate for cancer therapy. Accordingly, this work presents a comprehensive review of the most recent developments in utilizing this natural compound for the prevention, management, and treatment of various diseases, including cancer, along with the multiple mechanisms of action. In addition, this review summarizes the most recent and relevant literature that deals with the use of TSA as a therapeutic agent against various diseases, emphasizing its anticancer potential and the anticancer molecular mechanisms. Moreover, TSA has not been involved in toxicological effects on normal cells. Furthermore, this work highlights the potential utilization of TSA as a complementary or alternative medicine for preventing and treating cancer, alone or in combination with other anticancer drugs.
Collapse
Affiliation(s)
- Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
- Correspondence: (A.B.); (L.-H.L.); (M.S.M.)
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
| | - Mohamed Bakha
- Unit of Plant Biotechnology and Sustainable Development of Natural Resources “B2DRN”, Polydisciplinary Faculty of Beni Mellal, Sultan Moulay Slimane University, Mghila, P.O. Box 592, Beni Mellal 23000, Morocco
| | - Tarik Aanniz
- Medical Biotechnology Laboratory, Rabat Medical & Pharmacy School, Mohammed V University in Rabat, Rabat B.P. 6203, Morocco
| | - Naoual El Menyiy
- Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Naoufal El Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, Imouzzer Road Fez, Fez 30050, Morocco
| | - Aicha El Baaboua
- Biotechnology and Applied Microbiology Team, Department of Biology, Faculty of Sciences, Abdelmalek Essaadi University, Tetouan 93000, Morocco
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo 11566, Egypt
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ahmed Abdullah Al Awadh
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence: (A.B.); (L.-H.L.); (M.S.M.)
| | - Taoufiq Benali
- Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Sidi Bouzid B.P. 4162, Morocco
| | - Mohammad S. Mubarak
- Department of Chemistry, The University of Jordan, Amma 11942, Jordan
- Correspondence: (A.B.); (L.-H.L.); (M.S.M.)
| |
Collapse
|
9
|
Guerra MV, Cáceres MI, Herrera-Soto A, Arredondo SB, Varas-Godoy M, van Zundert B, Varela-Nallar L. H3K9 Methyltransferases Suv39h1 and Suv39h2 Control the Differentiation of Neural Progenitor Cells in the Adult Hippocampus. Front Cell Dev Biol 2022; 9:778345. [PMID: 35096813 PMCID: PMC8791356 DOI: 10.3389/fcell.2021.778345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022] Open
Abstract
In the dentate gyrus of the adult hippocampus new neurons are generated from neural precursor cells through different stages including proliferation and differentiation of neural progenitor cells and maturation of newborn neurons. These stages are controlled by the expression of specific transcription factors and epigenetic mechanisms, which together orchestrate the progression of the neurogenic process. However, little is known about the involvement of histone posttranslational modifications, a crucial epigenetic mechanism in embryonic neurogenesis that regulates fate commitment and neuronal differentiation. During embryonic development, the repressive modification trimethylation of histone H3 on lysine 9 (H3K9me3) contributes to the cellular identity of different cell-types. However, the role of this modification and its H3K9 methyltransferases has not been elucidated in adult hippocampal neurogenesis. We determined that during the stages of neurogenesis in the adult mouse dentate gyrus and in cultured adult hippocampal progenitors (AHPs), there was a dynamic change in the expression and distribution of H3K9me3, being enriched at early stages of the neurogenic process. A similar pattern was observed in the hippocampus for the dimethylation of histone H3 on lysine 9 (H3K9me2), another repressive modification. Among H3K9 methyltransferases, the enzymes Suv39h1 and Suv39h2 exhibited high levels of expression at early stages of neurogenesis and their expression decreased upon differentiation. Pharmacological inhibition of these enzymes by chaetocin in AHPs reduced H3K9me3 and concomitantly decreased neuronal differentiation while increasing proliferation. Moreover, Suv39h1 and Suv39h2 knockdown in newborn cells of the adult mouse dentate gyrus by retrovirus-mediated RNA interference impaired neuronal differentiation of progenitor cells. Our results indicate that H3K9me3 and H3K9 methyltransferases Suv39h1 and Suv39h2 are critically involved in the regulation of adult hippocampal neurogenesis by controlling the differentiation of neural progenitor cells.
Collapse
Affiliation(s)
- Miguel V Guerra
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Matías I Cáceres
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Andrea Herrera-Soto
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Sebastián B Arredondo
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Manuel Varas-Godoy
- Cancer Cell Biology Lab, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Brigitte van Zundert
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Lorena Varela-Nallar
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
10
|
Jiang H, Li Y, Xiang X, Tang Z, Liu K, Su Q, Zhang X, Li L. Chaetocin: A review of its anticancer potentials and mechanisms. Eur J Pharmacol 2021; 910:174459. [PMID: 34464601 DOI: 10.1016/j.ejphar.2021.174459] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/17/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Chaetocin is a natural metabolite product with various biological activities and pharmacological functions isolated from Chaetomium species fungi belonging to the thiodiketopyrazines. Numerous studies have demonstrated a wide range of antitumor activities of chaetocin in vitro and in vivo. Several studies have demonstrated that chaetocin suppresses the growth and proliferation of various tumour cells by regulating multiple signalling pathways related to tumour initiation and progression, inducing cancer cell apoptosis (intrinsic and extrinsic), enhancing autophagy, inducing cell cycle arrest, and inhibiting tumour angiogenesis, invasion, and migration. The antitumor effects and molecular mechanisms of chaetocin are reviewed and analysed in this paper, and the prospective applications of chaetocin in cancer prevention and therapy are also discussed. This review aimed to summarize the recent advances in the antitumor activity of chaetocin and to provide a rationale for further exploring the potential application of chaetocin in overcoming cancer in the future.
Collapse
Affiliation(s)
- Hangyu Jiang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Yuqi Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; School of Pharmacy, North Sichuan Medical College, Nanchong, China
| | - Xiaocong Xiang
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Zhili Tang
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Qiang Su
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Xiaofen Zhang
- Department of Urology, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China.
| | - Lin Li
- Department of Pharmacy, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China; College of Bioengineering, Chongqing University, Chongqing, China.
| |
Collapse
|
11
|
Saha N, Muntean AG. Insight into the multi-faceted role of the SUV family of H3K9 methyltransferases in carcinogenesis and cancer progression. Biochim Biophys Acta Rev Cancer 2020; 1875:188498. [PMID: 33373647 DOI: 10.1016/j.bbcan.2020.188498] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022]
Abstract
Growing evidence implicates histone H3 lysine 9 methylation in tumorigenesis. The SUV family of H3K9 methyltransferases, which include G9a, GLP, SETDB1, SETDB2, SUV39H1 and SUV39H2 deposit H3K9me1/2/3 marks at euchromatic and heterochromatic regions, catalyzed by their conserved SET domain. In cancer, this family of enzymes can be deregulated by genomic alterations and transcriptional mis-expression leading to alteration of transcriptional programs. In solid and hematological malignancies, studies have uncovered pro-oncogenic roles for several H3K9 methyltransferases and accordingly, small molecule inhibitors are being tested as potential therapies. However, emerging evidence demonstrate onco-suppressive roles for these enzymes in cancer development as well. Here, we review the role H3K9 methyltransferases play in tumorigenesis focusing on gene targets and biological pathways affected due to misregulation of these enzymes. We also discuss molecular mechanisms regulating H3K9 methyltransferases and their influence on cancer. Finally, we describe the impact of H3K9 methylation on therapy induced resistance in carcinoma. Converging evidence point to multi-faceted roles for H3K9 methyltransferases in development and cancer that encourages a deeper understanding of these enzymes to inform novel therapy.
Collapse
Affiliation(s)
- Nirmalya Saha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America
| | - Andrew G Muntean
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America.
| |
Collapse
|
12
|
Damasceno Teixeira TV, Fry RC, McKinnon A, Fry KL, Kelly JM, Verma PJ, Burden C, Salamone DF, Gambini A. Targeting epigenetic nuclear reprogramming in aggregated cloned equine embryos. Reprod Fertil Dev 2020; 31:1885-1893. [PMID: 31581975 DOI: 10.1071/rd19239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/10/2019] [Indexed: 12/16/2022] Open
Abstract
Epigenetic perturbations during the reprogramming process have been described as the primary cause of the low efficiency of somatic cell nuclear transfer (SCNT). In this study, we tested three strategies targeting nuclear reprogramming to investigate effects on equine SCNT. First, we evaluated the effect of treating somatic cells with chetomin, a fungal secondary metabolite reported to inhibit the trimethylation on histone 3 lysine 9 (H3K9 me3). Second, caffeine was added to the culture medium during the enucleation of oocytes and before activation of reconstructed embryos as a protein phosphatase inhibitor to improve nuclear reprogramming. Third, we tested the effects of the histone deacetylase inhibitor trichostatin A (TSA) added during both activation and early embryo culture. Although none of these treatments significantly improved the developmental rates of the invitro aggregated cloned equine embryos, the first equine cloned foal born in Australia was produced with somatic cells treated with chetomin. The present study describes the use of chetomin, caffeine and TSA for the first time in horses, serving as a starting point for the establishment of future protocols to target epigenetic reprogramming for improving the efficiency of equine cloning. Cloning is an expensive and inefficient process, but has gained particular interest in the equine industry. In this study we explored different strategies to improve cloning efficiency and produced the first cloned foal born in Australia. Our data serve as a starting point for the establishment of future protocols for improving equine cloning efficiency.
Collapse
Affiliation(s)
- Thiago V Damasceno Teixeira
- Laboratory of Animal and Meat Sciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Grattan Street, Parkville, Victoria, 3010, Australia
| | - Richard C Fry
- Laboratory of Animal and Meat Sciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Grattan Street, Parkville, Victoria, 3010, Australia
| | - Angus McKinnon
- Goulburn Valley Equine Hospital, 905 Goulburn Valley Highway, Congupna, Victoria 3633, Australia
| | - Kerri L Fry
- Laboratory of Animal and Meat Sciences, Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Grattan Street, Parkville, Victoria, 3010, Australia
| | - Jennifer M Kelly
- South Australian Research and Development Institute (SARDI), Turretfield Research Centre, Holland Road, Rosedale, 5350, South Australia, Australia
| | - Paul J Verma
- South Australian Research and Development Institute (SARDI), Turretfield Research Centre, Holland Road, Rosedale, 5350, South Australia, Australia
| | - Chelsie Burden
- Goulburn Valley Equine Hospital, 905 Goulburn Valley Highway, Congupna, Victoria 3633, Australia
| | - Daniel F Salamone
- Laboratorio de Biotecnología Animal, Facultad de Agronomia, Universidad de Buenos Aires, Av. San Martin 4453, C1417DSE, Ciudad Autónoma de Buenos Aires, Argentina; and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, C1425FQB, Ciudad Autónoma de Buenos Aires, Argentina
| | - Andrés Gambini
- Laboratorio de Biotecnología Animal, Facultad de Agronomia, Universidad de Buenos Aires, Av. San Martin 4453, C1417DSE, Ciudad Autónoma de Buenos Aires, Argentina; and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, C1425FQB, Ciudad Autónoma de Buenos Aires, Argentina; and Corresponding author.
| |
Collapse
|
13
|
Jeong PS, Sim BW, Park SH, Kim MJ, Kang HG, Nanjidsuren T, Lee S, Song BS, Koo DB, Kim SU. Chaetocin Improves Pig Cloning Efficiency by Enhancing Epigenetic Reprogramming and Autophagic Activity. Int J Mol Sci 2020; 21:ijms21144836. [PMID: 32650566 PMCID: PMC7402317 DOI: 10.3390/ijms21144836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Efficient epigenetic reprogramming is crucial for the in vitro development of mammalian somatic cell nuclear transfer (SCNT) embryos. The aberrant levels of histone H3 lysine 9 trimethylation (H3K9me3) is an epigenetic barrier. In this study, we evaluated the effects of chaetocin, an H3K9me3-specific methyltransferase inhibitor, on the epigenetic reprogramming and developmental competence of porcine SCNT embryos. The SCNT embryos showed abnormal levels of H3K9me3 at the pronuclear, two-cell, and four-cell stages compared to in vitro fertilized embryos. Moreover, the expression levels of H3K9me3-specific methyltransferases (suv39h1 and suv39h2) and DNA methyltransferases (DNMT1, DNMT3a, and DNMT3b) were higher in SCNT embryos. Treatment with 0.5 nM chaetocin for 24 h after activation significantly increased the developmental competence of SCNT embryos in terms of the cleavage rate, blastocyst formation rate, hatching rate, cell number, expression of pluripotency-related genes, and cell survival rate. In particular, chaetocin enhanced epigenetic reprogramming by reducing the H3K9me3 and 5-methylcytosine levels and restoring the abnormal expression of H3K9me3-specific methyltransferases and DNA methyltransferases. Chaetocin induced autophagic activity, leading to a significant reduction in maternal mRNA levels in embryos at the pronuclear and two-cell stages. These findings revealed that chaetocin enhanced the developmental competence of porcine SCNT embryos by regulating epigenetic reprogramming and autophagic activity and so could be used to enhance the production of transgenic pigs for biomedical research.
Collapse
Affiliation(s)
- Pil-Soo Jeong
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
- Department of Biotechnology, Daegu University, Gyeongsangbuk-do 38453, Korea
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Soo-Hyun Park
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Min Ju Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Hyo-Gu Kang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Tsevelmaa Nanjidsuren
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Sanghoon Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
| | - Deog-Bon Koo
- Department of Biotechnology, Daegu University, Gyeongsangbuk-do 38453, Korea
- Correspondence: (D.-B.K.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Chungcheongbuk-do 28116, Korea; (P.-S.J.); (B.-W.S.); (S.-H.P.); (M.J.K.); (H.-G.K.); (T.N.); (S.L.); (B.-S.S.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (D.-B.K.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| |
Collapse
|
14
|
Yang Z, Wang H, Zhang N, Xing T, Zhang W, Wang G, Li C, Yu C. Chaetocin Abrogates the Self-Renewal of Bladder Cancer Stem Cells via the Suppression of the KMT1A-GATA3-STAT3 Circuit. Front Cell Dev Biol 2020; 8:424. [PMID: 32626701 PMCID: PMC7311639 DOI: 10.3389/fcell.2020.00424] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Bladder cancer stem cells (BCSCs) have the abilities of self-renewal, differentiation, and metastasis; confer drug resistance; and exhibit high tumorigenicity. We previously identified that the KMT1A–GATA3–STAT3 axis drives the self-renewal of BCSCs. However, the therapeutic effect of targeting KMT1A in BCSCs remains unknown. In this study, we confirmed that the expression of KMT1A was remarkably higher in BCSCs (3–5-fold) than those in bladder cancer non-stem cells or normal bladder epithelial cells. Among the six KMT1A inhibitors, chaetocin significantly suppressed the cell propagation (inhibition ratio: 65%–88%, IC50 = 24.4–32.5 nM), induced apoptosis (2–5-fold), and caused G1 phase cell cycle arrest (68.9 vs 55.5%) of bladder cancer (BC) cells, without influencing normal bladder epithelial cells. More importantly, chaetocin abrogated the self-renewal of BCSCs (inhibition ratio: 80.1%) via the suppression of the KMT1A–GATA3–STAT3 circuit and other stemness-related pathways. Finally, intravesical instillation of chaetocin remarkably inhibited the growth of xenograft tumors (inhibition ratio: 71–82%) and prolonged the survival of tumor-bearing mice (70 vs 53 days). In sum, chaetocin abrogated the stemness maintenance and tumor growth of BCSCs via the suppression of the KMT1A–GATA3–STAT3 circuit. Chaetocin is an effective inhibitor targeting KMT1A in BCSCs and could be a promising therapeutic strategy for BC.
Collapse
Affiliation(s)
- Zhao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Haifeng Wang
- Department of Urology, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Nan Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Tianying Xing
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Urology, Affiliated Hospital of Hebei University, Baoding, China
| | - Guoqing Wang
- Department of Pathogenobiology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Chong Li
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
15
|
Ozyerli-Goknar E, Sur-Erdem I, Seker F, Cingöz A, Kayabolen A, Kahya-Yesil Z, Uyulur F, Gezen M, Tolay N, Erman B, Gönen M, Dunford J, Oppermann U, Bagci-Onder T. The fungal metabolite chaetocin is a sensitizer for pro-apoptotic therapies in glioblastoma. Cell Death Dis 2019; 10:894. [PMID: 31772153 PMCID: PMC6879621 DOI: 10.1038/s41419-019-2107-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/12/2019] [Accepted: 10/16/2019] [Indexed: 01/19/2023]
Abstract
Glioblastoma Multiforme (GBM) is the most common and aggressive primary brain tumor. Despite recent developments in surgery, chemo- and radio-therapy, a currently poor prognosis of GBM patients highlights an urgent need for novel treatment strategies. TRAIL (TNF Related Apoptosis Inducing Ligand) is a potent anti-cancer agent that can induce apoptosis selectively in cancer cells. GBM cells frequently develop resistance to TRAIL which renders clinical application of TRAIL therapeutics inefficient. In this study, we undertook a chemical screening approach using a library of epigenetic modifier drugs to identify compounds that could augment TRAIL response. We identified the fungal metabolite chaetocin, an inhibitor of histone methyl transferase SUV39H1, as a novel TRAIL sensitizer. Combining low subtoxic doses of chaetocin and TRAIL resulted in very potent and rapid apoptosis of GBM cells. Chaetocin also effectively sensitized GBM cells to further pro-apoptotic agents, such as FasL and BH3 mimetics. Chaetocin mediated apoptosis sensitization was achieved through ROS generation and consequent DNA damage induction that involved P53 activity. Chaetocin induced transcriptomic changes showed induction of antioxidant defense mechanisms and DNA damage response pathways. Heme Oxygenase 1 (HMOX1) was among the top upregulated genes, whose induction was ROS-dependent and HMOX1 depletion enhanced chaetocin mediated TRAIL sensitization. Finally, chaetocin and TRAIL combination treatment revealed efficacy in vivo. Taken together, our results provide a novel role for chaetocin as an apoptosis priming agent and its combination with pro-apoptotic therapies might offer new therapeutic approaches for GBMs.
Collapse
Affiliation(s)
- Ezgi Ozyerli-Goknar
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Ilknur Sur-Erdem
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Fidan Seker
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Ahmet Cingöz
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Alisan Kayabolen
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Zeynep Kahya-Yesil
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey
| | - Fırat Uyulur
- Department of Computational Biology, Koç University, 34450, Istanbul, Turkey
| | - Melike Gezen
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Nazife Tolay
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Batu Erman
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Mehmet Gönen
- Department of Industrial Engineering, College of Engineering, Koç University, İstanbul, Turkey
| | - James Dunford
- Botnar Research Centre, NIHR Biomedical Research Centre Oxford, University of Oxford, Oxford, OX3 7LD, UK
| | - Udo Oppermann
- Botnar Research Centre, NIHR Biomedical Research Centre Oxford, University of Oxford, Oxford, OX3 7LD, UK
- Structural Genomics Consortium, University of Oxford, Oxford, OX3 7DQ, UK
- FRIAS, Freiburg Institute of Advanced Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University School of Medicine, 34450, Istanbul, Turkey.
| |
Collapse
|
16
|
San José-Enériz E, Gimenez-Camino N, Agirre X, Prosper F. HDAC Inhibitors in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11111794. [PMID: 31739588 PMCID: PMC6896008 DOI: 10.3390/cancers11111794] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/05/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy characterized by uncontrolled proliferation, differentiation arrest, and accumulation of immature myeloid progenitors. Although clinical advances in AML have been made, especially in young patients, long-term disease-free survival remains poor, making this disease an unmet therapeutic challenge. Epigenetic alterations and mutations in epigenetic regulators contribute to the pathogenesis of AML, supporting the rationale for the use of epigenetic drugs in patients with AML. While hypomethylating agents have already been approved in AML, the use of other epigenetic inhibitors, such as histone deacetylases (HDAC) inhibitors (HDACi), is under clinical development. HDACi such as Panobinostat, Vorinostat, and Tricostatin A have been shown to promote cell death, autophagy, apoptosis, or growth arrest in preclinical AML models, yet these inhibitors do not seem to be effective as monotherapies, but rather in combination with other drugs. In this review, we discuss the rationale for the use of different HDACi in patients with AML, the results of preclinical studies, and the results obtained in clinical trials. Although so far the results with HDACi in clinical trials in AML have been modest, there are some encouraging data from treatment with the HDACi Pracinostat in combination with DNA demethylating agents.
Collapse
Affiliation(s)
- Edurne San José-Enériz
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Naroa Gimenez-Camino
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Xabier Agirre
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (X.A.); (F.P.); Tel.: +34-948-194700 (ext. 1002) (X.A.); +34-948-255400 (ext. 5807) (F.P.)
| | - Felipe Prosper
- Área de Hemato-Oncología, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra (IDISNA), Universidad de Navarra, 31008 Pamplona, Spain; (E.S.J.-E.); (N.G.-C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Departamento de Hematología, Clínica Universidad de Navarra, Universidad de Navarra, 31008 Pamplona, Spain
- Correspondence: (X.A.); (F.P.); Tel.: +34-948-194700 (ext. 1002) (X.A.); +34-948-255400 (ext. 5807) (F.P.)
| |
Collapse
|
17
|
Gomes NGM, Pereira RB, Andrade PB, Valentão P. Double the Chemistry, Double the Fun: Structural Diversity and Biological Activity of Marine-Derived Diketopiperazine Dimers. Mar Drugs 2019; 17:md17100551. [PMID: 31569621 PMCID: PMC6835637 DOI: 10.3390/md17100551] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/22/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022] Open
Abstract
While several marine natural products bearing the 2,5-diketopiperazine ring have been reported to date, the unique chemistry of dimeric frameworks appears to remain neglected. Frequently reported from marine-derived strains of fungi, many naturally occurring diketopiperazine dimers have been shown to display a wide spectrum of pharmacological properties, particularly within the field of cancer and antimicrobial therapy. While their structures illustrate the unmatched power of marine biosynthetic machinery, often exhibiting unsymmetrical connections with rare linkage frameworks, enhanced binding ability to a variety of pharmacologically relevant receptors has been also witnessed. The existence of a bifunctional linker to anchor two substrates, resulting in a higher concentration of pharmacophores in proximity to recognition sites of several receptors involved in human diseases, portrays this group of metabolites as privileged lead structures for advanced pre-clinical and clinical studies. Despite the structural novelty of various marine diketopiperazine dimers and their relevant bioactive properties in several models of disease, to our knowledge, this attractive subclass of compounds is reviewed here for the first time.
Collapse
Affiliation(s)
- Nelson G M Gomes
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, Porto 4050-313, Portugal.
| | - Renato B Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, Porto 4050-313, Portugal.
| | - Paula B Andrade
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, Porto 4050-313, Portugal.
| | - Patrícia Valentão
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, nº 228, Porto 4050-313, Portugal.
| |
Collapse
|
18
|
Xie L, Lin L, Huang S, Yang T, Shi D, Li X. Inhibition of Suv39H1 enhances transgenic IFNα-2b gene expression in Bcap-37 cells. Anim Biotechnol 2018; 30:358-365. [PMID: 30179066 DOI: 10.1080/10495398.2018.1500373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The low expression of exogenous transferred gene limited the application of transgenic animal technology. Suppressor of variegation 3 ∼ 9 homolog 1(SUV39H1) gene plays a prominent role on repressive heterochromatin and transcription. To understand if exogenous transgenic gene expression was affected by SUV39H1 epigenetic modification, in this paper, the effective shRNA fragments targeting SUV39H1 gene were first screened, their roles on expression of exogenous transgenic genes were determined by using Bcap-37 cell line with stable expressing IFNα-2b gene as a model, the preliminary regulation mechanism of SUV39H1 gene was investigated. The results showed that the designed shRNA1/2 fragments of SUV39H1 gene had an obvious inhibition effect on the expression of SUV39H1 gene, reached 53.07 and 31.28%, respectively by qRT-PCR analysis. Compared with the control group, the expression of IFNα-2b gene in transgenic Bcap-37 cells infected with shRNA1 and 2 viruses significantly increased by 96.25 and 121.08%, respectively (p < 0.05). In addition, the expression of DNMT1, HDAC1 and G9a gene in the shRNA infected cells reduced significantly, and the expression of the HAT1 gene increased significantly (p < 0.05). The above results indicated that the expression of exogenous transgenic gene could be promoted by suppressing SUV39H1 gene at the cell level.
Collapse
Affiliation(s)
- Liangliang Xie
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University , Guangxi , China
| | - Lang Lin
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University , Guangxi , China
| | - Shihai Huang
- College of Life Science and Technology, Guangxi University , Guangxi , China
| | - Ting Yang
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University , Guangxi , China
| | - Deshun Shi
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University , Guangxi , China
| | - Xiangping Li
- State Key Laboratory of Subtropical Bioresource Conservation and Utilization at Guangxi University , Guangxi , China
| |
Collapse
|
19
|
Zhang YM, Gao EE, Wang QQ, Tian H, Hou J. Effects of histone methyltransferase inhibitor chaetocin on histone H3K9 methylation of cultured ovine somatic cells and development of preimplantation cloned embryos. Reprod Toxicol 2018; 79:124-131. [PMID: 29909068 DOI: 10.1016/j.reprotox.2018.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 01/24/2023]
Abstract
Aberrant hypermethylation of histone H3 lysine 9 (H3K9) is a key barrier to the development of cloned embryos by somatic cell nuclear transfer (SCNT). The objective of this study was to assess the effects of chaetocin, an inhibitor of H3K9 methyltransferase SUV39 H, in regulating the H3K9 methylation in ovine SCNT embryos. Treatment of sheep fetal fibroblast cells with chaetocin specifically decreased the levels of H3K9 di-and trimethylation, and down-regulated the expression of H3K9 methyltransferases, SUV39H1/2 and G9A. Cloned embryos from chaetocin-treated cells could develop to the blastocyst stage at a similar rate to those derived from non-treated cells. However, direct treatment of SCNT or in vitro fertilized embryos with chaetocin impaired the embryonic development. These results suggest that although chaetocin is a potential agent for modulating H3K9 methylation in cells, it may have an adverse effect on the development of embryos.
Collapse
Affiliation(s)
- Yu-Mei Zhang
- State Key Laboratory of Agrobiotechnology and College of Biological Science, China Agricultural University, Beijing, China
| | - En-En Gao
- State Key Laboratory of Agrobiotechnology and College of Biological Science, China Agricultural University, Beijing, China
| | - Qian-Qian Wang
- State Key Laboratory of Agrobiotechnology and College of Biological Science, China Agricultural University, Beijing, China
| | - Hao Tian
- State Key Laboratory of Agrobiotechnology and College of Biological Science, China Agricultural University, Beijing, China
| | - Jian Hou
- State Key Laboratory of Agrobiotechnology and College of Biological Science, China Agricultural University, Beijing, China.
| |
Collapse
|
20
|
Structure based design, synthesis and activity studies of small hybrid molecules as HDAC and G9a dual inhibitors. Oncotarget 2017; 8:63187-63207. [PMID: 28968981 PMCID: PMC5609913 DOI: 10.18632/oncotarget.18730] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022] Open
Abstract
Aberrant enzymatic activities or expression profiles of epigenetic regulations are therapeutic targets for cancers. Among these, histone 3 lysine 9 methylation (H3K9Me2) and global de-acetylation on histone proteins are associated with multiple cancer phenotypes including leukemia, prostatic carcinoma, hepatocellular carcinoma and pulmonary carcinoma. Here, we report the discovery of the first small molecule capable of acting as a dual inhibitor targeting both G9a and HDAC. Our structure based design, synthesis, and screening for the dual activity of the small molecules led to the discovery of compound 14 which displays promising inhibition of both G9a and HDAC in low micro-molar range in cell based assays.
Collapse
|
21
|
Zuma AA, Santos JDO, Mendes I, de Souza W, Machado CR, Motta MCM. Chaetocin-A histone methyltransferase inhibitor-Impairs proliferation, arrests cell cycle and induces nucleolar disassembly in Trypanosoma cruzi. Acta Trop 2017; 170:149-160. [PMID: 28185826 DOI: 10.1016/j.actatropica.2017.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 01/12/2017] [Accepted: 02/06/2017] [Indexed: 12/15/2022]
Abstract
The Trypanosomatidae family includes pathogenic species of medical and veterinary interest. Chagas disease is endemic in Latin America, and about 8 million people are infected worldwide. There is a need for more effective drugs for the acute, undetermined and chronic phases of the disease that, in addition, do not cause side effects, stimulating the search for identification of new drug targets, as well as new chemotherapeutic targets. Trypanosomatids contain characteristic structures, such as the nucleus that undergoes a closed mitosis without chromosome formation and variations of chromatin packing in the different protozoa developmental stages. The nuclear DNA is condensed by histones that suffer post-translational modifications, such as addition of methyl groups by histone methyltransferases (MHT) and addition of acetyl groups by acetyltransferases. These processes modulate gene expression and chromatin organization, which are crucial to transcription, replication, repair and recombination. In the present study, the effects of chaetocin, a HMT inhibitor, on T. cruzi epimastigote proliferation, viability, ultrastructure and cell cycle were investigated. Results indicate that chaetocin promoted irreversible inhibition of protozoa growth, evident unpacking of nuclear heterochromatin and intense nucleolus fragmentation, which is associated with parasite cell cycle arrest and RNA transcription blockage. Taken together, data obtained with chaetocin treatment stimulate the use of histone methyltransferase inhibitors against pathogenic trypanosomatids.
Collapse
|
22
|
Tran TQ, Lowman XH, Kong M. Molecular Pathways: Metabolic Control of Histone Methylation and Gene Expression in Cancer. Clin Cancer Res 2017; 23:4004-4009. [PMID: 28404599 DOI: 10.1158/1078-0432.ccr-16-2506] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/19/2022]
Abstract
Epigenetic alterations contribute to tumor development, progression, and therapeutic response. Many epigenetic enzymes use metabolic intermediates as cofactors to modify chromatin structure. Emerging evidence suggests that fluctuation in metabolite levels may regulate activities of these chromatin-modifying enzymes. Here, we summarize recent progress in understanding the cross-talk between metabolism and epigenetic control of gene expression in cancer. We focus on how metabolic changes, due to diet, genetic mutations, or tumor microenvironment, regulate histone methylation status and, consequently, affect gene expression profiles to promote tumorigenesis. Importantly, we also suggest some potential therapeutic approaches to target the oncogenic role of metabolic alterations and epigenetic modifications in cancer. Clin Cancer Res; 23(15); 4004-9. ©2017 AACR.
Collapse
Affiliation(s)
- Thai Q Tran
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California
| | - Xazmin H Lowman
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California
| | - Mei Kong
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, California.
| |
Collapse
|
23
|
Gaál Z, Oláh É, Rejtő L, Erdődi F, Csernoch L. Strong Correlation between the Expression Levels of HDAC4 and SIRT6 in Hematological Malignancies of the Adults. Pathol Oncol Res 2016; 23:493-504. [DOI: 10.1007/s12253-016-0139-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 10/13/2016] [Indexed: 12/18/2022]
|
24
|
Li C, Tao Y, Li C, Liu B, Liu J, Wang G, Liu H. PU.1-Bim axis is involved in Trichostatin A-induced apoptosis in murine pro-B lymphoma FL5.12 cells. Acta Biochim Biophys Sin (Shanghai) 2016; 48:850-5. [PMID: 27451443 DOI: 10.1093/abbs/gmw067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/29/2016] [Indexed: 11/13/2022] Open
Abstract
Trichostatin A (TSA) is a well-known histone deacetylases (HDACs) inhibitor that has been reported to show potent anti-tumor capabilities in some types of cancer cell lines. However, detailed mechanism of TSA action on lymphoma remains to be described. In the present study, anti-proliferative effects of TSA were investigated using a murine pro-B lymphoma cell line FL5.12. MTT assay revealed that TSA potently inhibited the proliferation of FL5.12 cells in a time- and dose-dependent manner. Bright-field microscopy of FL5.12 cells showed apoptotic morphology at 24 h after TSA treatment. Consistently, TSA treatment led to DNA fragmentation and increased the protein levels of cleaved caspase 3 and PARP as revealed by western blot analysis. To explore the underlying mechanism of TSA-induced apoptosis of FL5.12 cells, we further analyzed the hematopoietic transcription factor Purine Rich Box-1 (PU.1) by western blot analysis. TSA treatment resulted in the inhibition of PU.1 in FL5.12 cells. In contrast, apoptotic protein Bim was induced by TSA, which was inversely correlated with the survival of FL5.12 cells. These results suggest the possible mechanism of TSA-induced apoptosis in murine pro-B lymphoma FL5.12 cells via the PU.1-Bim axis.
Collapse
Affiliation(s)
- Chao Li
- Infection and Immunity Laboratory, Kunming National High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Yufen Tao
- Infection and Immunity Laboratory, Kunming National High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Chao Li
- Infection and Immunity Laboratory, Kunming National High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Bo Liu
- Infection and Immunity Laboratory, Kunming National High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Jiansheng Liu
- Infection and Immunity Laboratory, Kunming National High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Guanlin Wang
- Kunming University of Science and Technology, Kunming 650118, China
| | - Hongqi Liu
- Infection and Immunity Laboratory, Kunming National High-level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
25
|
Ramachandran S, Ient J, Göttgens EL, Krieg AJ, Hammond EM. Epigenetic Therapy for Solid Tumors: Highlighting the Impact of Tumor Hypoxia. Genes (Basel) 2015; 6:935-56. [PMID: 26426056 PMCID: PMC4690023 DOI: 10.3390/genes6040935] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/18/2015] [Accepted: 09/22/2015] [Indexed: 12/14/2022] Open
Abstract
In the last few decades, epigenetics has emerged as an exciting new field in development and disease, with a more recent focus towards cancer. Epigenetics has classically referred to heritable patterns of gene expression, primarily mediated through DNA methylation patterns. More recently, it has come to include the reversible chemical modification of histones and DNA that dictate gene expression patterns. Both the epigenetic up-regulation of oncogenes and downregulation of tumor suppressors have been shown to drive tumor development. Current clinical trials for cancer therapy include pharmacological inhibition of DNA methylation and histone deacetylation, with the aim of reversing these cancer-promoting epigenetic changes. However, the DNA methyltransferase and histone deacetylase inhibitors have met with less than promising results in the treatment of solid tumors. Regions of hypoxia are a common occurrence in solid tumors. Tumor hypoxia is associated with increased aggressiveness and therapy resistance, and importantly, hypoxic tumor cells have a distinct epigenetic profile. In this review, we provide a summary of the recent clinical trials using epigenetic drugs in solid tumors, discuss the hypoxia-induced epigenetic changes and highlight the importance of testing the epigenetic drugs for efficacy against the most aggressive hypoxic fraction of the tumor in future preclinical testing.
Collapse
Affiliation(s)
- Shaliny Ramachandran
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK.
| | - Jonathan Ient
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK.
| | - Eva-Leonne Göttgens
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK.
| | - Adam J Krieg
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Ester M Hammond
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK.
| |
Collapse
|
26
|
Lai YS, Chen JY, Tsai HJ, Chen TY, Hung WC. The SUV39H1 inhibitor chaetocin induces differentiation and shows synergistic cytotoxicity with other epigenetic drugs in acute myeloid leukemia cells. Blood Cancer J 2015; 5:e313. [PMID: 25978433 PMCID: PMC4476016 DOI: 10.1038/bcj.2015.37] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/14/2015] [Indexed: 12/26/2022] Open
Abstract
Epigenetic modifying enzymes have a crucial role in the pathogenesis of acute myeloid leukemia (AML). Methylation of lysine 9 on histone H3 by the methyltransferase G9a and SUV39H1 is associated with inhibition of tumor suppressor genes. We studied the effect of G9a and SUV39H1 inhibitors on viability and differentiation of AML cells and tested the cytotoxicity induced by combination of G9a and SUV39H1 inhibitors and various epigenetic drugs. The SUV39H1 inhibitor (chaetocin) and the G9a inhibitor (UNC0638) caused cell death in AML cells at high concentrations. However, only chaetocin-induced CD11b expression and differentiation of AML cells at non-cytotoxic concentration. HL-60 and KG-1a cells were more sensitive to chaetocin than U937 cells. Long-term incubation of chaetocin led to downregulation of SUV39H1 and reduction of H3K9 tri-methylation in HL-60 and KG-1a cells. Combination of chaetocin with suberoylanilide hydroxamic acid (SAHA, a histone deacetylase inhibitor) or JQ (a BET (bromodomain extra terminal) bromodomain inhibitor) showed synergistic cytotoxicity. Conversely, no synergism was found by combining chaetocin and UNC0638. More importantly, chaetocin-induced differentiation and combined cytotoxicity were also found in the primary cells of AML patients. Collectively, the SUV39H1 inhibitor chaetocin alone or in combination with other epigenetic drugs may be effective for the treatment of AML.
Collapse
Affiliation(s)
- Y-S Lai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - J-Y Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - H-J Tsai
- 1] National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan [2] Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - T-Y Chen
- Division of Hematology/Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - W-C Hung
- 1] National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
27
|
Baumann M, Dieskau AP, Loertscher BM, Walton MC, Nam S, Xie J, Horne D, Overman LE. Tricyclic Analogues of Epidithiodioxopiperazine Alkaloids with Promising In Vitro and In Vivo Antitumor Activity. Chem Sci 2015; 6:4451-4457. [PMID: 26301062 PMCID: PMC4540405 DOI: 10.1039/c5sc01536g] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A short synthesis of 1,4-dioxohexahydro-6H-3,8a-epidithiopyrrolo[1,2-a]pyrazines will enable future mechanistic and translational studies of these structurally novel and promising clinical antitumor candidates.
Epipolythiodioxopiperazine (ETP) alkaloids are structurally elaborate alkaloids that show potent antitumor activity. However, their high toxicity and demonstrated interactions with various biological receptors compromises their therapeutic potential. In an effort to mitigate these disadvantages, a short stereocontrolled construction of tricyclic analogues of epidithiodioxopiperazine alkaloids was developed. Evaluation of a small library of such structures against two invasive cancer cell lines defined initial structure–activity relationships (SAR), which identified 1,4-dioxohexahydro-6H-3,8a-epidithiopyrrolo[1,2-a]pyrazine 3c and related structures as particularly promising antitumor agents. ETP alkaloid analogue 3c exhibits low nanomolar activity against both solid and blood tumors in vitro. In addition, 3c significantly suppresses tumor growth in mouse xenograft models of melanoma and lung cancer, without obvious signs of toxicity, following either intraperitoneal (IP) or oral administration. The short synthesis of molecules in this series will enable future mechanistic and translational studies of these structurally novel and highly promising clinical antitumor candidates.
Collapse
Affiliation(s)
- Marcus Baumann
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, California 92697-2025
| | - André P Dieskau
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, California 92697-2025
| | - Brad M Loertscher
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, California 92697-2025
| | - Mary C Walton
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, California 92697-2025
| | - Sangkil Nam
- Department of Molecular Medicine, Beckman Research Institute of City Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, 1500 E. Duarte Road, Duarte, California 91010
| | - Jun Xie
- Department of Molecular Medicine, Beckman Research Institute of City Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, 1500 E. Duarte Road, Duarte, California 91010
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City Hope Comprehensive Cancer Center, Beckman Research Institute, Department of Molecular Medicine, 1500 E. Duarte Road, Duarte, California 91010
| | - Larry E Overman
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, California 92697-2025
| |
Collapse
|
28
|
Chiba T, Saito T, Yuki K, Zen Y, Koide S, Kanogawa N, Motoyama T, Ogasawara S, Suzuki E, Ooka Y, Tawada A, Otsuka M, Miyazaki M, Iwama A, Yokosuka O. Histone lysine methyltransferase SUV39H1 is a potent target for epigenetic therapy of hepatocellular carcinoma. Int J Cancer 2014; 136:289-98. [PMID: 24844570 DOI: 10.1002/ijc.28985] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 05/04/2014] [Accepted: 05/09/2014] [Indexed: 12/14/2022]
Abstract
Histone H3 lysine 9 trimethylation (H3K9me3) is associated with transcriptional repression and regulated by histone lysine methyltransferases such as SUV39H1 and ESET. However, the functional roles of these enzymes in hepatocellular carcinoma (HCC) remain uncertain. In this study, we conducted loss-of-function assays for HCC cells. SUV39H1 knockdown but not ESET knockdown reduced H3K9me3 levels and impaired HCC cell growth and sphere formation. The pharmacological inhibition of SUV39H1 by chaetocin resulted in cell growth inhibition and inducing cellular apoptosis in culture and xenograft subcutaneous tumors. Real-time polymerase chain reaction analysis indicated high levels of SUV39H1 expression in 24 of 42 (57.1%) HCC surgical samples compared with corresponding nontumor tissues. Immunohistochemistry identified high levels of H3K9me3 and ESET proteins in 23 (54.8%) and 29 (69.0%) tumor tissues, respectively. However, these proteins' expressions were only observed in biliary epithelial cells and periportal hepatocytes of nontumor tissues. Expression levels of SUV39H1 but not those of ESET were significantly correlated with H3K9me3 levels. The cumulative HCC recurrence rate was significantly higher for patients with elevated SUV39H1 expression and H3K9me3 levels. In conclusion, our data indicate that elevated SUV39H1 expression and high levels of H3K9me3 have important roles in HCC development and progression. Therefore, the pharmacological inhibition of SUV39H1 may be a promising therapeutic approach for HCC treatment.
Collapse
Affiliation(s)
- Tetsuhiro Chiba
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan; Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|