1
|
Sharma R, Yadav J, Bhat SA, Musayev A, Myrzagulova S, Sharma D, Padha N, Saini M, Tuli HS, Singh T. Emerging Trends in Neuroblastoma Diagnosis, Therapeutics, and Research. Mol Neurobiol 2025; 62:6423-6466. [PMID: 39804528 DOI: 10.1007/s12035-024-04680-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 12/20/2024] [Indexed: 03/29/2025]
Abstract
This review explores the current understanding and recent advancements in neuroblastoma, one of the most common extracranial solid pediatric cancers, accounting for ~ 15% of childhood cancer-related mortality. The hallmarks of NBL, including angiogenesis, metastasis, apoptosis resistance, cell cycle dysregulation, drug resistance, and responses to hypoxia and ROS, underscore its complex biology. The tumor microenvironment's significance in disease progression is acknowledged in this study, along with the pivotal role of cancer stem cells in sustaining tumor growth and heterogeneity. A number of molecular signatures are being studied in order to better understand the disease, with many of them serving as targets for the development of new therapeutics. This includes inhibitor therapies for NBL patients, which notably concentrate on ALK signaling, MDM2, PI3K/Akt/mTOR, Wnt, and RAS-MAPK pathways, along with regulators of epigenetic mechanisms. Additionally, this study offers an extensive understanding of the molecular therapies used, such as monoclonal antibodies and CAR-T therapy, focused on both preclinical and clinical studies. Radiation therapy's evolving role and the promise of stem cell transplantation-mediated interventions underscore the dynamic landscape of NBL treatment. This study has also emphasized the recent progress in the field of diagnosis, encompassing the adoption of artificial intelligence and liquid biopsy as a non-intrusive approach for early detection and ongoing monitoring of NBL. Furthermore, the integration of innovative treatment approaches such as CRISPR-Cas9, and cancer stem cell therapy has also been emphasized in this review.
Collapse
Affiliation(s)
- Rishabh Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Jaya Yadav
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Sajad Ahmad Bhat
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
- Department of Biochemistry, NIMS University, Rajasthan, Jaipur, 303121, India
| | - Abdugani Musayev
- Asfendiyarov Kazakh National Medical University, Almaty, 050000, Kazakhstan
| | | | - Deepika Sharma
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
| | - Nipun Padha
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Department of Zoology, Cluster University of Jammu, Jammu, 180001, India
| | - Manju Saini
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India
- Amity Stem Cell Institute, Amity Medical School, Amity University, Haryana, 122412, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana, 133207, India
| | - Tejveer Singh
- Translational Oncology Laboratory, Department of Zoology, Hansraj College, Delhi University, New Delhi, 110007, India.
- Division of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, (INMAS-DRDO), New Delhi, Delhi, 110054, India.
| |
Collapse
|
2
|
Xu L, Xuan H, Shi X. Dysregulation of the p300/CBP histone acetyltransferases in human cancer. Epigenomics 2025; 17:193-208. [PMID: 39929233 PMCID: PMC11812348 DOI: 10.1080/17501911.2024.2447807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
p300 (E1A binding protein 300) and CBP (CREB-binding protein) are critical regulators of chromatin dynamics and gene expression, playing essential roles in various cellular processes, including proliferation, differentiation, apoptosis, and immune responses. These homologous histone acetyltransferases (HATs) function as transcriptional co-activators by acetylating histones and non-histone proteins. p300/CBP is essential for development, and dysregulation of p300 and CBP has been implicated in several human diseases, particularly cancer. Somatic mutations that inactivate p300/CBP are frequently observed across various cancer types. Additionally, other mutations leading to translocations or truncations of p300/CBP can result in enhanced catalytic activity, potentially representing novel gain-of-function mutations that promote tumor progression. In this review, we discuss the mechanisms underlying the regulation of p300/CBP HAT activity, its dysregulation in cancer, and the development of p300/CBP inhibitors and their potential in cancer therapies.
Collapse
Affiliation(s)
- Longxia Xu
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Hongwen Xuan
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Xiaobing Shi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| |
Collapse
|
3
|
Gronkowska K, Robaszkiewicz A. Genetic dysregulation of EP300 in cancers in light of cancer epigenome control - targeting of p300-proficient and -deficient cancers. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200871. [PMID: 39351073 PMCID: PMC11440307 DOI: 10.1016/j.omton.2024.200871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Some cancer types including bladder, cervical, and uterine cancers are characterized by frequent mutations in EP300 that encode histone acetyltransferase p300. This enzyme can act both as a tumor suppressor and oncogene. In this review, we describe the role of p300 in cancer initiation and progression regarding EP300 aberrations that have been identified in TGCA Pan-Cancer Atlas studies and we also discuss possible anticancer strategies that target EP300 mutated cancers. Copy number alterations, truncating mutations, and abnormal EP300 transcriptions that affect p300 abundance and activity are associated with several pathological features such as tumor grading, metastases, and patient survival. Elevated EP300 correlates with a higher mRNA level of other epigenetic factors and chromatin remodeling enzymes that co-operate with p300 in creating permissive conditions for malignant transformation, tumor growth and metastases. The status of EP300 expression can be considered as a prognostic marker for anticancer immunotherapy efficacy, as EP300 mutations are followed by an increased expression of PDL-1.HAT activators such as CTB or YF2 can be applied for p300-deficient patients, whereas the natural and synthetic inhibitors of p300 activity, as well as dual HAT/bromodomain inhibitors and the PROTAC degradation of p300, may serve as strategies in the fight against p300-fueled cancers.
Collapse
Affiliation(s)
- Karolina Gronkowska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
- Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland
| | - Agnieszka Robaszkiewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
4
|
Feng T, Zhang H, Zhou Y, Zhu Y, Shi S, Li K, Lin P, Chen J. Roles of posttranslational modifications in lipid metabolism and cancer progression. Biomark Res 2024; 12:141. [PMID: 39551780 PMCID: PMC11571667 DOI: 10.1186/s40364-024-00681-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Lipid metabolism reprogramming has emerged as a hallmark of malignant tumors. Lipids represent a complex group of biomolecules that not only compose the essential components of biological membranes and act as an energy source, but also function as messengers to integrate various signaling pathways. In tumor cells, de novo lipogenesis plays a crucial role in acquiring lipids to meet the demands of rapid growth. Increasing evidence has suggested that dysregulated lipid metabolism serves as a driver of cancer progression. Posttranslational modifications (PTMs), which occurs in most eukaryotic proteins throughout their lifetimes, affect the activity, abundance, function, localization, and interactions of target proteins. PTMs of crucial molecules are potential intervention sites and are emerging as promising strategies for the cancer treatment. However, there is limited information available regarding the PTMs that occur in cancer lipid metabolism and the potential treatment strategies associated with these PTMs. Herein, we summarize current knowledge of the roles and regulatory mechanisms of PTMs in lipid metabolism. Understanding the roles of PTMs in lipid metabolism in cancer could provide valuable insights into tumorigenesis and progression. Moreover, targeting PTMs in cancer lipid metabolism might represent a promising novel therapeutic strategy.
Collapse
Affiliation(s)
- Tianyu Feng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - He Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Yanjie Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Yalan Zhu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Shiya Shi
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China
| | - Kai Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
| | - Ping Lin
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
- Sichuan Clinical Research Center for Laboratory Medicine, Chengdu, Sichuan Province, 610041, China.
- Clinical Laboratory Medicine Research Center of West China Hospital, #37, Guo Xue Lane, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
5
|
Pathikonda S, Amirmahani F, Mathew D, Muthukrishnan SD. Histone acetyltransferases as promising therapeutic targets in glioblastoma resistance. Cancer Lett 2024; 604:217269. [PMID: 39326554 DOI: 10.1016/j.canlet.2024.217269] [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: 06/17/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Glioblastoma (GBM) is a fatal adult brain tumor with an extremely poor prognosis. GBM poses significant challenges for targeted therapies due to its intra- and inter-tumoral heterogeneity, a highly immunosuppressive microenvironment, diffuse infiltration into normal brain parenchyma, protection by the blood-brain barrier and acquisition of therapeutic resistance. Recent studies have implicated epigenetic modifiers as key players driving tumorigenesis, resistance, and progression of GBM. While the vast majority of GBM research on epigenetic modifiers thus far has focused predominantly on elucidating the functional roles and targeting of DNA methyltransferases and histone deacetylases, emerging evidence indicates that histone acetyltransferases (HATs) also play a key role in mediating plasticity and therapeutic resistance in GBM. Here, we will provide an overview of HATs, their dual roles and functions in cancer as both tumor suppressors and oncogenes and focus specifically on their implications in GBM resistance. We also discuss the technical challenges in developing selective HAT inhibitors and highlight their promise as potential anti-cancer therapeutics for treating intractable cancers such as GBM.
Collapse
Affiliation(s)
- Spoorthy Pathikonda
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Farzaneh Amirmahani
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Diya Mathew
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| | - Sree Deepthi Muthukrishnan
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA.
| |
Collapse
|
6
|
Ma M, Fei X, Jiang D, Chen H, Xie X, Wang Z, Huang Q. Research Progress on the Mechanism of Histone Deacetylases in Ferroptosis of Glioma. Oncol Rev 2024; 18:1432131. [PMID: 39193375 PMCID: PMC11348391 DOI: 10.3389/or.2024.1432131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Glioma is the most prevalent primary malignant tumor of the central nervous system. While traditional treatment modalities such as surgical resection, radiotherapy, and chemotherapy have made significant advancements in glioma treatment, the prognosis for glioma patients remains often unsatisfactory. Ferroptosis, a novel form of programmed cell death, plays a crucial role in glioma and is considered to be the most functionally rich programmed cell death process. Histone deacetylases have emerged as a key focus in regulating ferroptosis in glioma. By inhibiting the activity of histone deacetylases, histone deacetylase inhibitors elevate acetylation levels of both histones and non-histone proteins, thereby influencing various cellular processes. Numerous studies have demonstrated that histone deacetylases are implicated in the development of glioma and hold promise for its treatment. This article provides an overview of research progress on the mechanism by which histone deacetylases contribute to ferroptosis in glioma.
Collapse
Affiliation(s)
- Meng Ma
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Xifeng Fei
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Dongyi Jiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Hanchun Chen
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Xiangtong Xie
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou, China
| | - Zhimin Wang
- Department of Neurosurgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Qiang Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Marka S, Zografaki ME, Tsolomiti G, Kalliampakou KI, Tsolomitis A, Koumantou C, Smirlis D, Vassilaki N, Kintzios S. 2-(4-Nitrophenyl)isothiazol-3(2H)-one: A Promising Selective Agent against Hepatocellular Carcinoma Cells. Pharmaceuticals (Basel) 2024; 17:673. [PMID: 38931341 PMCID: PMC11206498 DOI: 10.3390/ph17060673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Liver cancer ranks among the most prevalent malignancies globally and stands as a leading cause of cancer-related mortality. Numerous isothiazolone derivatives and analogues have been synthesized and investigated for their potential as anticancer agents; however, limited data exist regarding their efficacy against liver cancer. In the present study, two nitrophenyl-isothiazolones, the 5-benzoyl-2-(4-nitrophenyl)isothiazol-3(2H)-one (IsoA) and the 2-(4-nitrophenyl)isothiazol-3(2H)-one (IsoB), were preliminarily investigated for their cytotoxicity against hepatoma human (Huh7) cells as a liver cancer model and Immortalized Human Hepatocytes (IHHs) as a model of non-cancerous hepatocytes. IsoB, derived from IsoA after removal of the benzoyl moiety, demonstrated the highest cytotoxic effect against Huh7 cells with CC50 values of 19.3 μΜ at 24 h, 16.4 μΜ at 48 h, and 16.2 μΜ at 72 h of incubation, respectively. IsoB also exhibited selective toxicity against the liver cancerous Huh7 cells compared to IHH cells, reinforcing its role as a potent and selective anticancer agent. Remarkably, the cytotoxicity of IsoB was higher when compared with the standard chemotherapeutical agent 5-fluorouracil (5-FU), which also failed to exhibit higher toxicity against the liver cancerous cell lines. Moreover, IsoB-treated Huh7 cells presented a noteworthy reduction in mitochondrial membrane potential (ΔΨm) after 48 and 72 h, while mitochondrial superoxide levels showed an increase after 24 h of incubation. The molecular mechanism of the IsoB cytotoxic effect was also investigated using RT-qPCR, revealing an apoptosis-mediated cell death along with tumor suppressor TP53 overexpression and key-oncogene MYCN downregulation.
Collapse
Affiliation(s)
- Sofia Marka
- Laboratory of Cell Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (G.T.); (C.K.)
| | - Maria-Eleftheria Zografaki
- Laboratory of Molecular Biology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (M.-E.Z.); (K.I.K.)
| | - Georgia Tsolomiti
- Laboratory of Cell Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (G.T.); (C.K.)
| | - Katerina I. Kalliampakou
- Laboratory of Molecular Biology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (M.-E.Z.); (K.I.K.)
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Athanasios Tsolomitis
- School of Chemical Engineering, National Technical University, 15772 Athens, Greece;
| | - Christina Koumantou
- Laboratory of Cell Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (G.T.); (C.K.)
| | - Despina Smirlis
- Molecular Parasitology Laboratory, Microbiology Department, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Niki Vassilaki
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece;
| | - Spyros Kintzios
- Laboratory of Cell Technology, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece; (G.T.); (C.K.)
| |
Collapse
|
8
|
Zohourian N, Brown JAL. Current trends in clinical trials and the development of small molecule epigenetic inhibitors as cancer therapeutics. Epigenomics 2024; 16:671-680. [PMID: 38639711 PMCID: PMC11233149 DOI: 10.2217/epi-2023-0443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
Epigenetic mechanisms control and regulate normal chromatin structure and gene expression patterns, with epigenetic dysregulation observed in many different cancer types. Importantly, epigenetic modifications are reversible, offering the potential to silence oncogenes and reactivate tumor suppressors. Small molecule drugs manipulating these epigenetic mechanisms are at the leading edge of new therapeutic options for cancer treatment. The clinical use of histone deacetyltransferases inhibitors (HDACi) demonstrates the effectiveness of targeting epigenetic mechanisms for cancer treatment. Notably, the development of new classes of inhibitors, including lysine acetyltransferase inhibitors (KATi), are the future of epigenetic-based therapeutics. We outline the progress of current classes of small molecule epigenetic drugs for use against cancer (preclinical and clinical) and highlight the potential market growth in epigenetic-based therapeutics.
Collapse
Affiliation(s)
- Nazanin Zohourian
- Department of Biological Science, University of Limerick, Limerick, V94 T9PX, Ireland
| | - James AL Brown
- Department of Biological Science, University of Limerick, Limerick, V94 T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC), University of Limerick, Limerick, Ireland
- Health Research Institute (HRI), University of Limerick, Limerick, Ireland
- Bernal Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
9
|
Runa F, Ortiz-Soto G, de Barros NR, Kelber JA. Targeting SMAD-Dependent Signaling: Considerations in Epithelial and Mesenchymal Solid Tumors. Pharmaceuticals (Basel) 2024; 17:326. [PMID: 38543112 PMCID: PMC10975212 DOI: 10.3390/ph17030326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024] Open
Abstract
SMADs are the canonical intracellular effector proteins of the TGF-β (transforming growth factor-β). SMADs translocate from plasma membrane receptors to the nucleus regulated by many SMAD-interacting proteins through phosphorylation and other post-translational modifications that govern their nucleocytoplasmic shuttling and subsequent transcriptional activity. The signaling pathway of TGF-β/SMAD exhibits both tumor-suppressing and tumor-promoting phenotypes in epithelial-derived solid tumors. Collectively, the pleiotropic nature of TGF-β/SMAD signaling presents significant challenges for the development of effective cancer therapies. Here, we review preclinical studies that evaluate the efficacy of inhibitors targeting major SMAD-regulating and/or -interacting proteins, particularly enzymes that may play important roles in epithelial or mesenchymal compartments within solid tumors.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| | | | | | - Jonathan A Kelber
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
- Department of Biology, Baylor University, Waco, TX 76706, USA
| |
Collapse
|
10
|
Ma Q, Zeng Q, Wang K, Qian M, Li J, Wang H, Zhang H, Jiang J, Chen Z, Huang W. Acetyltransferase P300 Regulates Glucose Metabolic Reprogramming through Catalyzing Succinylation in Lung Cancer. Int J Mol Sci 2024; 25:1057. [PMID: 38256128 PMCID: PMC10816063 DOI: 10.3390/ijms25021057] [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: 10/17/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Aberrant protein post-translational modification is a hallmark of malignant tumors. Lysine succinylation (Ksucc) plays a vital role in cell energy metabolism in various cancers. However, whether succinylation can be catalyzed by acetyltransferase p300 remains unclear. In this study, we unveiled that p300 is a "writer" for succinylation, and p300-mediated Ksucc promotes cell glycometabolism in lung adenocarcinoma (LUAD). Specifically, our succinylome data revealed that EP300 deficiency leads to the systemic reduction of Ksucc, and 79.55% of the p300-succinylated proteins were found in the cytoplasm, which were primarily enriched in the carbohydrate metabolism process. Interestingly, deleting EP300 led to a notable decrease in Ksucc levels on several glycolytic enzymes, especially Phosphoglycerate Kinase 1 (PGK1). Mutation of the succinylated site of PGK1 notably hindered cell glycolysis and lactic acid excretion. Metabolomics in vivo indicated that p300-caused metabolic reprogramming was mainly attributed to the altered carbohydrate metabolism. In addition, 89.35% of LUAD patients exhibited cytoplasmic localization of p300, with higher levels in tumor tissues than adjacent normal tissues. High levels of p300 correlated with advanced tumor stages and poor prognosis of LUAD patients. Briefly, we disclose the activity of p300 to catalyze succinylation, which contributes to cell glucose metabolic reprogramming and malignant progression of lung cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Wan Huang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
11
|
Veschi V, Durinck K, Thiele CJ, Speleman F. Neuroblastoma Epigenetic Landscape: Drugging Opportunities. PEDIATRIC ONCOLOGY 2024:71-95. [DOI: 10.1007/978-3-031-51292-6_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Chen G, Bao B, Cheng Y, Tian M, Song J, Zheng L, Tong Q. Acetyl-CoA metabolism as a therapeutic target for cancer. Biomed Pharmacother 2023; 168:115741. [PMID: 37864899 DOI: 10.1016/j.biopha.2023.115741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023] Open
Abstract
Acetyl-coenzyme A (acetyl-CoA), an essential metabolite, not only takes part in numerous intracellular metabolic processes, powers the tricarboxylic acid cycle, serves as a key hub for the biosynthesis of fatty acids and isoprenoids, but also serves as a signaling substrate for acetylation reactions in post-translational modification of proteins, which is crucial for the epigenetic inheritance of cells. Acetyl-CoA links lipid metabolism with histone acetylation to create a more intricate regulatory system that affects the growth, aggressiveness, and drug resistance of malignancies such as glioblastoma, breast cancer, and hepatocellular carcinoma. These fascinating advances in the knowledge of acetyl-CoA metabolism during carcinogenesis and normal physiology have raised interest regarding its modulation in malignancies. In this review, we provide an overview of the regulation and cancer relevance of main metabolic pathways in which acetyl-CoA participates. We also summarize the role of acetyl-CoA in the metabolic reprogramming and stress regulation of cancer cells, as well as medical application of inhibitors targeting its dysregulation in therapeutic intervention of cancers.
Collapse
Affiliation(s)
- Guo Chen
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Banghe Bao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Yang Cheng
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Minxiu Tian
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Jiyu Song
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China.
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China.
| |
Collapse
|
13
|
Beaudry A, Jacques-Ricard S, Darracq A, Sgarioto N, Garcia A, García TR, Lemieux W, Béland K, Haddad E, Cordeiro P, Duval M, McGraw S, Richer C, Caron M, Marois F, St-Onge P, Sinnett D, Banquy X, Raynal NJM. Repurposing disulfiram, an alcohol-abuse drug, in neuroblastoma causes KAT2A downregulation and in vivo activity with a water/oil emulsion. Sci Rep 2023; 13:16443. [PMID: 37777587 PMCID: PMC10543387 DOI: 10.1038/s41598-023-43219-2] [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: 03/09/2023] [Accepted: 09/21/2023] [Indexed: 10/02/2023] Open
Abstract
Neuroblastoma, the most common type of pediatric extracranial solid tumor, causes 10% of childhood cancer deaths. Despite intensive multimodal treatment, the outcomes of high-risk neuroblastoma remain poor. We urgently need to develop new therapies with safe long-term toxicity profiles for rapid testing in clinical trials. Drug repurposing is a promising approach to meet these needs. Here, we investigated disulfiram, a safe and successful chronic alcoholism treatment with known anticancer and epigenetic effects. Disulfiram efficiently induced cell cycle arrest and decreased the viability of six human neuroblastoma cell lines at half-maximal inhibitory concentrations up to 20 times lower than its peak clinical plasma level in patients treated for chronic alcoholism. Disulfiram shifted neuroblastoma transcriptome, decreasing MYCN levels and activating neuronal differentiation. Consistently, disulfiram significantly reduced the protein level of lysine acetyltransferase 2A (KAT2A), drastically reducing acetylation of its target residues on histone H3. To investigate disulfiram's anticancer effects in an in vivo model of high-risk neuroblastoma, we developed a disulfiram-loaded emulsion to deliver the highly liposoluble drug. Treatment with the emulsion significantly delayed neuroblastoma progression in mice. These results identify KAT2A as a novel target of disulfiram, which directly impacts neuroblastoma epigenetics and is a promising candidate for repurposing to treat pediatric neuroblastoma.
Collapse
Affiliation(s)
- Annie Beaudry
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Simon Jacques-Ricard
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Pharmacologie et de Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Anaïs Darracq
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Pharmacologie et de Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Nicolas Sgarioto
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Araceli Garcia
- Faculté de Pharmacie, Université de Montréal, Montreal, QC, Canada
| | | | - William Lemieux
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Kathie Béland
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Elie Haddad
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Paulo Cordeiro
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Michel Duval
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Serge McGraw
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Chantal Richer
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Maxime Caron
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - François Marois
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Pascal St-Onge
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Daniel Sinnett
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Pédiatrie, Université de Montréal, Montreal, QC, Canada
| | - Xavier Banquy
- Faculté de Pharmacie, Université de Montréal, Montreal, QC, Canada
| | - Noël J-M Raynal
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
- Département de Pharmacologie et de Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
14
|
Zhang Y, Gao Y, Ding Y, Jiang Y, Chen H, Zhan Z, Liu X. Targeting KAT2A inhibits inflammatory macrophage activation and rheumatoid arthritis through epigenetic and metabolic reprogramming. MedComm (Beijing) 2023; 4:e306. [PMID: 37313329 PMCID: PMC10258526 DOI: 10.1002/mco2.306] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/15/2023] Open
Abstract
Epigenetic regulation of inflammatory macrophages governs inflammation initiation and resolution in the pathogenesis of rheumatoid arthritis (RA). Nevertheless, the mechanisms underlying macrophage-mediated arthritis injuries remain largely obscure. Here, we found that increased expression of lysine acetyltransferase 2A (KAT2A) in synovial tissues was closely correlated with inflammatory joint immunopathology in both RA patients and experimental arthritis mice. Administration of MB-3, the KAT2A-specific chemical inhibitor, significantly ameliorated the synovitis and bone destruction in collagen-induced arthritis model. Both pharmacological inhibition and siRNA silencing of KAT2A, not only suppressed innate stimuli-triggered proinflammatory gene (such as Il1b and Nlrp3) transcription but also impaired NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in vivo and in vitro. Mechanistically, KAT2A facilitated macrophage glycolysis reprogramming through suppressing nuclear factor-erythroid 2-related factor 2 (NRF2) activity as well as downstream antioxidant molecules, which supported histone 3 lysine 9 acetylation (H3K9ac) and limited NRF2-mediated transcriptional repression of proinflammatory genes. Our study proves that acetyltransferase KAT2A licenses metabolic and epigenetic reprogramming for NLRP3 inflammasome activation in inflammatory macrophages, thereby targeting KAT2A represents a potential therapeutic approach for patients suffering from RA and relevant inflammatory diseases.
Collapse
Affiliation(s)
- Yunkai Zhang
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
- National Key Laboratory of Immunity & InflammationNaval Medical UniversityShanghaiChina
| | - Ying Gao
- Department of RheumatologyChanghai Hospital, Naval Medical UniversityShanghaiChina
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Yingying Ding
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| | - Yuyu Jiang
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| | - Huiying Chen
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| | - Zhenzhen Zhan
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaShanghai East Hospital, Tongji University School of MedicineShanghaiChina
- Department of Liver Surgery, Shanghai Institute of TransplantationRenji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xingguang Liu
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
- National Key Laboratory of Immunity & InflammationNaval Medical UniversityShanghaiChina
| |
Collapse
|
15
|
Ramakrishnan J, Magudeeswaran S, Suresh S, Poomani K. Investigation of intermolecular interactions and binding mechanism of PU139 and PU141 molecules with p300 HAT enzyme via molecular docking, molecular dynamics simulations and binding free energy analysis. J Biomol Struct Dyn 2023; 41:1351-1365. [PMID: 34974819 DOI: 10.1080/07391102.2021.2020164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The p300 histone acetyltransferase (HAT) enzyme acetylates the lysine residue of histone promotes the transcription reaction. The abnormal function of p300 HAT enzyme causes various diseases such as Cancer, Asthma, Alzheimer, Diabetics, and AIDS. In the recent years, several studies have been conducted to design potential drug to inhibit this enzyme. Recently, an in vitro study has been performed on the synthetic molecules PU139 and PU141 to inhibit the p300 HAT enzyme. The present study aims to understand the binding affinity, intermolecular interactions, conformational stability and binding energy of PU139 and PU141 molecules in the active site of p300 HAT enzyme from the in silico studies. The molecular docking and molecular dynamics (MD) simulations were carried out for both ligands with the p300 HAT enzyme. The molecular docking and MD simulations reveals that both molecules forms expected interactions with the catalytic site key residues of p300 enzyme. The MD simulation shows the maximum RMSD value for the PU141 is 2.3 Å, whereas for PU139 is 3.3 Å; these low RMSD values indicate that both molecules are highly stable in the active site of p300. The calculated binding free energy of PU141 (-20.62 kcal/mol) is higher than the molecule PU139 (-17.67 kcal/mol). Among the results, PU141 shows the high binding affinity with p300 while comparing with PU139. The results of this in-silico study coupled with the findings reported in the in vitro study confirm that PU141 may be suitable for clinical study.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jaganathan Ramakrishnan
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| | - Sivanandam Magudeeswaran
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| | - Suganya Suresh
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| | - Kumaradhas Poomani
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, India
| |
Collapse
|
16
|
Effects of the Acetyltransferase p300 on Tumour Regulation from the Novel Perspective of Posttranslational Protein Modification. Biomolecules 2023; 13:biom13030417. [PMID: 36979352 PMCID: PMC10046601 DOI: 10.3390/biom13030417] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
p300 acts as a transcription coactivator and an acetyltransferase that plays an important role in tumourigenesis and progression. In previous studies, it has been confirmed that p300 is an important regulator in regulating the evolution of malignant tumours and it also has extensive functions. From the perspective of non-posttranslational modification, it has been proven that p300 can participate in regulating many pathophysiological processes, such as activating oncogene transcription, promoting tumour cell growth, inducing apoptosis, regulating immune function and affecting embryo development. In recent years, p300 has been found to act as an acetyltransferase that catalyses a variety of protein modification types, such as acetylation, propanylation, butyylation, 2-hydroxyisobutyration, and lactylation. Under the catalysis of this acetyltransferase, it plays its crucial tumourigenic driving role in many malignant tumours. Therefore, the function of p300 acetyltransferase has gradually become a research hotspot. From a posttranslational modification perspective, p300 is involved in the activation of multiple transcription factors and additional processes that promote malignant biological behaviours, such as tumour cell proliferation, migration, and invasion, as well as tumour cell apoptosis, drug resistance, and metabolism. Inhibitors of p300 have been developed and are expected to become novel anticancer drugs for several malignancies. We review the characteristics of the p300 protein and its functional role in tumour from the posttranslational modification perspective, as well as the current status of p300-related inhibitor research, with a view to gaining a comprehensive understanding of p300.
Collapse
|
17
|
Gaál Z. Targeted Epigenetic Interventions in Cancer with an Emphasis on Pediatric Malignancies. Biomolecules 2022; 13:61. [PMID: 36671446 PMCID: PMC9855367 DOI: 10.3390/biom13010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Over the past two decades, novel hallmarks of cancer have been described, including the altered epigenetic landscape of malignant diseases. In addition to the methylation and hyd-roxymethylation of DNA, numerous novel forms of histone modifications and nucleosome remodeling have been discovered, giving rise to a wide variety of targeted therapeutic interventions. DNA hypomethylating drugs, histone deacetylase inhibitors and agents targeting histone methylation machinery are of distinguished clinical significance. The major focus of this review is placed on targeted epigenetic interventions in the most common pediatric malignancies, including acute leukemias, brain and kidney tumors, neuroblastoma and soft tissue sarcomas. Upcoming novel challenges include specificity and potential undesirable side effects. Different epigenetic patterns of pediatric and adult cancers should be noted. Biological significance of epigenetic alterations highly depends on the tissue microenvironment and widespread interactions. An individualized treatment approach requires detailed genetic, epigenetic and metabolomic evaluation of cancer. Advances in molecular technologies and clinical translation may contribute to the development of novel pediatric anticancer treatment strategies, aiming for improved survival and better patient quality of life.
Collapse
Affiliation(s)
- Zsuzsanna Gaál
- Department of Pediatric Hematology-Oncology, Institute of Pediatrics, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
18
|
Garcia K, Gingras AC, Harvey KF, Tanas MR. TAZ/YAP fusion proteins: mechanistic insights and therapeutic opportunities. Trends Cancer 2022; 8:1033-1045. [PMID: 36096997 PMCID: PMC9671862 DOI: 10.1016/j.trecan.2022.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022]
Abstract
The Hippo pathway is dysregulated in many different cancers, but point mutations in the pathway are rare. Transcriptional co-activator with PDZ-binding motif (TAZ) and Yes-associated protein (YAP) fusion proteins have emerged in almost all major cancer types and represent the most common genetic mechanism by which the two transcriptional co-activators are activated. Given that the N termini of TAZ or YAP are fused to the C terminus of another transcriptional regulator, the resultant fusion proteins hyperactivate a TEAD transcription factor-based transcriptome. Recent advances show that the C-terminal fusion partners confer oncogenic properties to TAZ/YAP fusion proteins by recruiting epigenetic modifiers that promote a hybrid TEAD-based transcriptome. Elucidating these cooperating epigenetic complexes represents a strategy to identify new therapeutic approaches for a pathway that has been recalcitrant to medical therapy.
Collapse
Affiliation(s)
- Keith Garcia
- Department of Pathology, University of Iowa, Iowa City, IA, USA; Cancer Biology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Munir R Tanas
- Department of Pathology, University of Iowa, Iowa City, IA, USA; Cancer Biology Graduate Program, University of Iowa, Iowa City, IA, USA; Pathology and Laboratory Medicine, Veterans Affairs Medical Center, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
19
|
Hao Y, Yi Q, XiaoWu X, WeiBo C, GuangChen Z, XueMin C. Acetyl-CoA: An interplay between metabolism and epigenetics in cancer. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:1044585. [PMID: 39086974 PMCID: PMC11285595 DOI: 10.3389/fmmed.2022.1044585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/02/2022] [Indexed: 08/02/2024]
Abstract
Due to its high mortality and severe economic burden, cancer has become one of the most difficult medical problems to solve today. As a key node in metabolism and the main producer of energy, acetyl-coenzyme A (acetyl-CoA) plays an important role in the invasion and migration of cancer. In this review, we discuss metabolic pathways involving acetyl-CoA, the targeted therapy of cancer through acetyl-CoA metabolic pathways and the roles of epigenetic modifications in cancer. In particular, we emphasize that the metabolic pathway of acetyl-CoA exerts a great impact in cancer; this process is very different from normal cells due to the "Warburg effect". The concentration of acetyl-CoA is increased in the mitochondria of cancer cells to provide ATP for survival, hindering the growth of normal cells. Therefore, it may be possible to explore new feasible and more effective treatments through the acetyl-CoA metabolic pathway. In addition, a growing number of studies have shown that abnormal epigenetic modifications have been shown to play contributing roles in cancer formation and development. In most cancers, acetyl-CoA mediated acetylation promotes the growth of cancer cells. Thus, acetylation biomarkers can also be detected and serve as potential cancer prediction and prognostic markers.
Collapse
Affiliation(s)
- Yang Hao
- Changzhou First People’s Hospital, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| | - Qin Yi
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xu XiaoWu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen WeiBo
- Changzhou First People’s Hospital, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| | - Zu GuangChen
- Changzhou First People’s Hospital, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| | - Chen XueMin
- Changzhou First People’s Hospital, The Third Affiliated Hospital of Suzhou University, Changzhou, China
| |
Collapse
|
20
|
Muthukrishnan SD, Kawaguchi R, Nair P, Prasad R, Qin Y, Johnson M, Wang Q, VanderVeer-Harris N, Pham A, Alvarado AG, Condro MC, Gao F, Gau R, Castro MG, Lowenstein PR, Deb A, Hinman JD, Pajonk F, Burns TC, Goldman SA, Geschwind DH, Kornblum HI. P300 promotes tumor recurrence by regulating radiation-induced conversion of glioma stem cells to vascular-like cells. Nat Commun 2022; 13:6202. [PMID: 36261421 PMCID: PMC9582000 DOI: 10.1038/s41467-022-33943-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 10/07/2022] [Indexed: 12/24/2022] Open
Abstract
Glioma stem cells (GSC) exhibit plasticity in response to environmental and therapeutic stress leading to tumor recurrence, but the underlying mechanisms remain largely unknown. Here, we employ single-cell and whole transcriptomic analyses to uncover that radiation induces a dynamic shift in functional states of glioma cells allowing for acquisition of vascular endothelial-like and pericyte-like cell phenotypes. These vascular-like cells provide trophic support to promote proliferation of tumor cells, and their selective depletion results in reduced tumor growth post-treatment in vivo. Mechanistically, the acquisition of vascular-like phenotype is driven by increased chromatin accessibility and H3K27 acetylation in specific vascular genes allowing for their increased expression post-treatment. Blocking P300 histone acetyltransferase activity reverses the epigenetic changes induced by radiation and inhibits the adaptive conversion of GSC into vascular-like cells and tumor growth. Our findings highlight a role for P300 in radiation-induced stress response, suggesting a therapeutic approach to prevent glioma recurrence.
Collapse
Affiliation(s)
- Sree Deepthi Muthukrishnan
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Riki Kawaguchi
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Pooja Nair
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Rachna Prasad
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Yue Qin
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Maverick Johnson
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Qing Wang
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Nathan VanderVeer-Harris
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Amy Pham
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Alvaro G Alvarado
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Michael C Condro
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Fuying Gao
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Raymond Gau
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Maria G Castro
- Department of Neurosurgery, and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Terry C Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Translational Neuromedicine, University of Coppenhagen School of Medicine, Coppenhagen, Denmark
| | - Daniel H Geschwind
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Harley I Kornblum
- The UCLA Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Yang J, Song C, Zhan X. The role of protein acetylation in carcinogenesis and targeted drug discovery. Front Endocrinol (Lausanne) 2022; 13:972312. [PMID: 36171897 PMCID: PMC9510633 DOI: 10.3389/fendo.2022.972312] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/23/2022] [Indexed: 12/01/2022] Open
Abstract
Protein acetylation is a reversible post-translational modification, and is involved in many biological processes in cells, such as transcriptional regulation, DNA damage repair, and energy metabolism, which is an important molecular event and is associated with a wide range of diseases such as cancers. Protein acetylation is dynamically regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs) in homeostasis. The abnormal acetylation level might lead to the occurrence and deterioration of a cancer, and is closely related to various pathophysiological characteristics of a cancer, such as malignant phenotypes, and promotes cancer cells to adapt to tumor microenvironment. Therapeutic modalities targeting protein acetylation are a potential therapeutic strategy. This article discussed the roles of protein acetylation in tumor pathology and therapeutic drugs targeting protein acetylation, which offers the contributions of protein acetylation in clarification of carcinogenesis, and discovery of therapeutic drugs for cancers, and lays the foundation for precision medicine in oncology.
Collapse
Affiliation(s)
- Jingru Yang
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Cong Song
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| |
Collapse
|
22
|
Xia JK, Qin XQ, Zhang L, Liu SJ, Shi XL, Ren HZ. Roles and regulation of histone acetylation in hepatocellular carcinoma. Front Genet 2022; 13:982222. [PMID: 36092874 PMCID: PMC9452893 DOI: 10.3389/fgene.2022.982222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Hepatocellular Carcinoma (HCC) is the most frequent malignant tumor of the liver, but its prognosis is poor. Histone acetylation is an important epigenetic regulatory mode that modulates chromatin structure and transcriptional status to control gene expression in eukaryotic cells. Generally, histone acetylation and deacetylation processes are controlled by the opposing activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs). Dysregulation of histone modification is reported to drive aberrant transcriptional programmes that facilitate liver cancer onset and progression. Emerging studies have demonstrated that several HDAC inhibitors exert tumor-suppressive properties via activation of various cell death molecular pathways in HCC. However, the complexity involved in the epigenetic transcription modifications and non-epigenetic cellular signaling processes limit their potential clinical applications. This review brings an in-depth view of the oncogenic mechanisms reported to be related to aberrant HCC-associated histone acetylation, which might provide new insights into the effective therapeutic strategies to prevent and treat HCC.
Collapse
Affiliation(s)
- Jin-kun Xia
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| | - Xue-qian Qin
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lu Zhang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shu-jun Liu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiao-lei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| | - Hao-zhen Ren
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Hepatobiliary Institute Nanjing University, Nanjing, China
| |
Collapse
|
23
|
Busto N, Leitão-Castro J, García-Sosa AT, Cadete F, Marques CS, Freitas R, Burke AJ. N-1,2,3-Triazole-isatin derivatives: anti-proliferation effects and target identification in solid tumour cell lines. RSC Med Chem 2022; 13:970-977. [PMID: 36092141 PMCID: PMC9384811 DOI: 10.1039/d2md00044j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/30/2022] [Indexed: 09/01/2023] Open
Abstract
Molecular hybridization approaches have become an important strategy in medicinal chemistry, and to this end, we have developed a series of novel N-1,2,3-triazole-isatin hybrids that are promising as tumour anti-proliferative agents. Our isatin hybrids presented high cytotoxic activity against colon cancer cell line SW480, lung adenocarcinoma cell line A549, as well as breast cancer cell lines MCF7 and MDA-MB-231. All tested compounds demonstrated better anti-proliferation (to 1-order of magnitude) than the cis-platin (CDDP) benchmark. In order to explore potential biological targets for these compounds, we used information from previous screenings and identified as putative targets the histone acetyltransferase P-300 (EP300) and the acyl-protein thioesterase 2 (LYPLA2), both known to be involved in epigenetic regulation. Advantageous pharmacological properties were predicted for these compounds such as good total surface area of binding to aromatic and hydrophobic units in the enzyme active site. In addition, we found down-regulation of LYPLA2 and EP300 in both the MCF7 and MDA-MB-231 breast cancer cells treated with our inhibitors, but no significant effect was detected in normal breast cells MCF10A. We also observed upregulation of EP300 mRNA expression in the MCF10A cell line for some of these compounds and the same effect for LYPLA2 mRNA in MCF7 for one of our compounds. These results suggest an effect at the transcriptional regulation level and associated with oncological contexts.
Collapse
Affiliation(s)
- Natalia Busto
- Departamento de Ciencias de la Salud, Facultad de Ciencias de la Salud, Universidad de Burgos Paseo de los Comendadores, s/n 09001 Burgos Spain
| | - Joana Leitão-Castro
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto Rua Alfredo Allen, 208 4200-135 Porto Portugal
| | | | - Francisco Cadete
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto Rua Alfredo Allen, 208 4200-135 Porto Portugal
| | - Carolina S Marques
- LAQV-REQUIMTE, Institute for Research and Advanced Studies, University of Évora Rua Romão Ramalho, 59 7000-671 Évora Portugal
| | - Renata Freitas
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto Rua Alfredo Allen, 208 4200-135 Porto Portugal
| | - Anthony J Burke
- LAQV-REQUIMTE, Institute for Research and Advanced Studies, University of Évora Rua Romão Ramalho, 59 7000-671 Évora Portugal
- Chemistry and Biochemistry Department, School of Science and Technology, University of Évora Rua Romão Ramalho 59 7000-671 Évora Portugal
| |
Collapse
|
24
|
Jin Y, Liu T, Luo H, Liu Y, Liu D. Targeting Epigenetic Regulatory Enzymes for Cancer Therapeutics: Novel Small-Molecule Epidrug Development. Front Oncol 2022; 12:848221. [PMID: 35419278 PMCID: PMC8995554 DOI: 10.3389/fonc.2022.848221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of the epigenetic enzyme-mediated transcription of oncogenes or tumor suppressor genes is closely associated with the occurrence, progression, and prognosis of tumors. Based on the reversibility of epigenetic mechanisms, small-molecule compounds that target epigenetic regulation have become promising therapeutics. These compounds target epigenetic regulatory enzymes, including DNA methylases, histone modifiers (methylation and acetylation), enzymes that specifically recognize post-translational modifications, chromatin-remodeling enzymes, and post-transcriptional regulators. Few compounds have been used in clinical trials and exhibit certain therapeutic effects. Herein, we summarize the classification and therapeutic roles of compounds that target epigenetic regulatory enzymes in cancer treatment. Finally, we highlight how the natural compounds berberine and ginsenosides can target epigenetic regulatory enzymes to treat cancer.
Collapse
Affiliation(s)
- Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Tianjia Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Haoming Luo
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Yangyang Liu
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
25
|
Abbas EMH, Farghaly TA, Sabour R, Shaaban MR, Abdallah ZA. Design, synthesis, cytotoxicity, and molecular docking studies of novel thiazolyl-hydrazone derivatives as histone lysine acetyl-transferase inhibitors and apoptosis inducers. Arch Pharm (Weinheim) 2022; 355:e2200076. [PMID: 35393652 DOI: 10.1002/ardp.202200076] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 01/30/2023]
Abstract
Compounds containing both thiazole and arylsulfone moieties are recognized for their high biological activity and ability to fight a variety of ailments. Thus, in this context, new derivatives of (thiazol-2-yl)hydrazone with an arylsulfone moiety were synthesized as CPTH2 analogs with potent anti-histone lysine acetyl-transferase activity. Compounds 3, 4, 10b, and 11b showed an excellent inhibitory effect on P300 (E1A-associated protein p300), compared to CPTH2. Among all the tested derivatives, compound 10b revealed the highest activity against both P300 and pCAF. In addition, the new hits were tested for anticancer efficacy against two leukemia cell lines. Most of them showed a moderate to potent antitumor effect on the k562 and CCRF-CEM cell lines. Interestingly, the activity of compound 10b against the k562 cell line was found to be higher than that of CPTH2. Furthermore, it showed a good safety profile, better than CPTH2 on normal cells. Molecular docking analysis was carried out to reveal the crucial binding contacts in the inhibition of the P300 and pCAF enzymes.
Collapse
Affiliation(s)
- Eman M H Abbas
- Department of Chemistry, Natural and Microbial Products, National Research Center, Dokki, Cairo, Egypt
| | - Thoraya A Farghaly
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Rehab Sabour
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Mohamed R Shaaban
- Department of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah, Almukkarramah, Saudi Arabia
| | - Zeinab A Abdallah
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
26
|
Ciaccio R, De Rosa P, Aloisi S, Viggiano M, Cimadom L, Zadran SK, Perini G, Milazzo G. Targeting Oncogenic Transcriptional Networks in Neuroblastoma: From N-Myc to Epigenetic Drugs. Int J Mol Sci 2021; 22:12883. [PMID: 34884690 PMCID: PMC8657550 DOI: 10.3390/ijms222312883] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma (NB) is one of the most frequently occurring neurogenic extracranial solid cancers in childhood and infancy. Over the years, many pieces of evidence suggested that NB development is controlled by gene expression dysregulation. These unleashed programs that outline NB cancer cells make them highly dependent on specific tuning of gene expression, which can act co-operatively to define the differentiation state, cell identity, and specialized functions. The peculiar regulation is mainly caused by genetic and epigenetic alterations, resulting in the dependency on a small set of key master transcriptional regulators as the convergence point of multiple signalling pathways. In this review, we provide a comprehensive blueprint of transcriptional regulation bearing NB initiation and progression, unveiling the complexity of novel oncogenic and tumour suppressive regulatory networks of this pathology. Furthermore, we underline the significance of multi-target therapies against these hallmarks, showing how novel approaches, together with chemotherapy, surgery, or radiotherapy, can have substantial antineoplastic effects, disrupting a wide variety of tumorigenic pathways through combinations of different treatments.
Collapse
|
27
|
Cai Q, Gan C, Tang C, Wu H, Gao J. Mechanism and Therapeutic Opportunities of Histone Modifications in Chronic Liver Disease. Front Pharmacol 2021; 12:784591. [PMID: 34887768 PMCID: PMC8650224 DOI: 10.3389/fphar.2021.784591] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
Chronic liver disease (CLD) represents a global health problem, accounting for the heavy burden of disability and increased health care utilization. Epigenome alterations play an important role in the occurrence and progression of CLD. Histone modifications, which include acetylation, methylation, and phosphorylation, represent an essential part of epigenetic modifications that affect the transcriptional activity of genes. Different from genetic mutations, histone modifications are plastic and reversible. They can be modulated pharmacologically without changing the DNA sequence. Thus, there might be chances to establish interventional solutions by targeting histone modifications to reverse CLD. Here we summarized the roles of histone modifications in the context of alcoholic liver disease (ALD), metabolic associated fatty liver disease (MAFLD), viral hepatitis, autoimmune liver disease, drug-induced liver injury (DILI), and liver fibrosis or cirrhosis. The potential targets of histone modifications for translation into therapeutics were also investigated. In prospect, high efficacy and low toxicity drugs that are selectively targeting histone modifications are required to completely reverse CLD and prevent the development of liver cirrhosis and malignancy.
Collapse
Affiliation(s)
- Qiuyu Cai
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Can Gan
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Chengwei Tang
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Wu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinhang Gao
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Han P, Wang Y, Luo W, Lu Y, Zhou X, Yang Y, Zheng Q, Li D, Wu S, Li L, Zhang H, Zhao J, Zhang Z, Matskova L, Li P, Zhou X. Epigenetic inactivation of hydroxymethylglutaryl CoA synthase reduces ketogenesis and facilitates tumor cell motility in clear cell renal carcinoma. Pathol Res Pract 2021; 227:153622. [PMID: 34624592 DOI: 10.1016/j.prp.2021.153622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Previously, we have reported that the dysregulation of ketogenesis plays an important role in the carcinogenesis of clear cell renal cell carcinoma (ccRCC). Here, we demonstrate decreased expression of the HMGCS2 gene in ccRCC, a critical enzyme for the synthesis of the ketone body β-hydroxybutyrate (β-OHB). We found that the reduced transcription of the HMGCS2 gene in ccRCC cells was significantly correlated to a higher relative methylation rate in its promotor region. The higher methylation rate in the region of the transcription start site and 1st exon of the HMGCS2 gene was, in turn, correlated with a worse clinical outcome for patients. The transcription of HMGCS2 was possible to restore by treatment with 5-aza-2'-deoxycytidine and with the histone deacetylase inhibitor β-OHB. Therefore, the low levels of the HMGCS2 enzyme in ccRCC may be the consequence of hypermethylation of the HMGCS2 promotor. The ensuing reduction in the ketone body levels further suppresses the transcription of HMGCS2 via a feedback loop. Ectopic expression of HMGCS2 attenuates the migration and invasion of ccRCC but does not affect the proliferative capacity of ccRCC cells in vitro. In addition, we showed that ectopic expression of HMGCS2 boosts the intracellular levels of β-OHB and that exogenously applied β-OHB suppresses the motility and invasion of ccRCC. Our study reveals crosstalk between genes that regulate metabolism and their metabolites, thus providing a better understanding of the epigenetic mechanism involved in ccRCC carcinogenesis and suggesting opportunities for metabolic therapy of tumors. Initially, we suggest that the mRNA level of HMGCS2 could serve as a potentially valuable diagnostic (AUC = 0.918, p < 0.001) and prognostic biomarker.
Collapse
Affiliation(s)
- Peipei Han
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China; Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yifang Wang
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Wenqi Luo
- Department of Pathology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yunliang Lu
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Yanping Yang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Qian Zheng
- Life Science Institute, Guangxi Medical University, Nanning, China
| | - Danping Li
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Shu Wu
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Limei Li
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Haishan Zhang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Jun Zhao
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Zhe Zhang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Liudmila Matskova
- Institute of Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Ping Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China; Department of Pathology, College & Hospital of Stomatology Guangxi Medical University, Nanning, China.
| | - Xiaoying Zhou
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China; Life Science Institute, Guangxi Medical University, Nanning, China.
| |
Collapse
|
29
|
The GCN5: its biological functions and therapeutic potentials. Clin Sci (Lond) 2021; 135:231-257. [PMID: 33443284 DOI: 10.1042/cs20200986] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
General control non-depressible 5 (GCN5) or lysine acetyltransferase 2A (KAT2A) is one of the most highly studied histone acetyltransferases. It acts as both histone acetyltransferase (HAT) and lysine acetyltransferase (KAT). As an HAT it plays a pivotal role in the epigenetic landscape and chromatin modification. Besides, GCN5 regulates a wide range of biological events such as gene regulation, cellular proliferation, metabolism and inflammation. Imbalance in the GCN5 activity has been reported in many disorders such as cancer, metabolic disorders, autoimmune disorders and neurological disorders. Therefore, unravelling the role of GCN5 in different diseases progression is a prerequisite for both understanding and developing novel therapeutic agents of these diseases. In this review, we have discussed the structural features, the biological function of GCN5 and the mechanical link with the diseases associated with its imbalance. Moreover, the present GCN5 modulators and their limitations will be presented in a medicinal chemistry perspective.
Collapse
|
30
|
Wang Y, Xie Q, Tan H, Liao M, Zhu S, Zheng LL, Huang H, Liu B. Targeting cancer epigenetic pathways with small-molecule compounds: Therapeutic efficacy and combination therapies. Pharmacol Res 2021; 173:105702. [PMID: 34102228 DOI: 10.1016/j.phrs.2021.105702] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/07/2021] [Accepted: 05/29/2021] [Indexed: 02/08/2023]
Abstract
Epigenetics mainly refers to covalent modifications to DNA or histones without affecting genomes, which ultimately lead to phenotypic changes in cells or organisms. Given the abundance of regulatory targets in epigenetic pathways and their pivotal roles in tumorigenesis and drug resistance, the development of epigenetic drugs holds a great promise for the current cancer therapy. However, lack of potent, selective, and clinically tractable small-molecule compounds makes the strategy to target cancer epigenetic pathways still challenging. Therefore, this review focuses on epigenetic pathways, small molecule inhibitors targeting DNA methyltransferase (DNMT) and small molecule inhibitors targeting histone modification (the main regulatory targets are histone acetyltransferases (HAT), histone deacetylases (HDACs) and histone methyltransferases (HMTS)), as well as the combination strategies of the existing epigenetic therapeutic drugs and more new therapies to improve the efficacy, which will shed light on a new clue on discovery of more small-molecule drugs targeting cancer epigenetic pathways as promising strategies in the future.
Collapse
Affiliation(s)
- Yi Wang
- Health Management Center, Sichuan Provincial People' Hospital, University of Electronic Science and Technology of China, Chengdu 610072, PR China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, PR China
| | - Qiang Xie
- Department of Stomatology, Sichuan Provincial People' Hospital, University of Electronic Science and Technology of China, Chengdu 610072, PR China
| | - Huidan Tan
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Minru Liao
- Department of Stomatology, Sichuan Provincial People' Hospital, University of Electronic Science and Technology of China, Chengdu 610072, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Shiou Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ling-Li Zheng
- Department of Pharmacy, The First Affiliated Hospital of Chengdu Medical College, No. 278, Baoguang Rd, Xindu Region, Chengdu 610500, PR China.
| | - Haixia Huang
- Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, PR China; Department of Prosthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, 646000, PR China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
31
|
Alsamri H, Hasasna HE, Baby B, Alneyadi A, Dhaheri YA, Ayoub MA, Eid AH, Vijayan R, Iratni R. Carnosol Is a Novel Inhibitor of p300 Acetyltransferase in Breast Cancer. Front Oncol 2021; 11:664403. [PMID: 34055630 PMCID: PMC8155611 DOI: 10.3389/fonc.2021.664403] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/20/2021] [Indexed: 12/21/2022] Open
Abstract
Carnosol, a natural polyphenol abundant in edible plants such as sage, rosemary, and oregano, has shown promising anticancer activity against various types of cancers. Nonetheless, very little is known about its molecular mechanism of action or its downstream target(s). We have previously shown that carnosol inhibits cellular proliferation, migration, invasion, and metastasis as well as triggers autophagy and apoptosis in the highly invasive MDA-MB-231 breast cancer cells. Here, we report that carnosol induces histone hypoacetylation in MDA-MB-231 and Hs578T breast cancer cells. We show that, while carnosol does not affect HDACs, it promotes a ROS-dependent proteasome degradation of p300 and PCAF histone acetyl transferases (HATs) without affecting other HATs such as GCN5 and hMOF. Carnosol-induced histone hypoacetylation remains persistent even when p300 and PCAF protein levels were rescued from degradation by (i) the inhibition of the proteasome activity by the proteasome inhibitors MG-132 and bortezomib, and (ii) the inhibition of ROS accumulation by the ROS scavenger, N-acetylcysteine. In addition, we report that, in a cell-free system, carnosol efficiently inhibits histone acetyltransferase activity of recombinant p300 but not that of PCAF or GCN5. Molecular docking studies reveal that carnosol inhibits p300 HAT activity by blocking the entry of the acetyl-CoA binding pocket of the catalytic domain. The superimposition of the docked conformation of the p300 HAT domain in complex with carnosol shows a similar orientation as the p300 structure with acetyl-CoA. Carnosol occupies the region where the pantetheine arm of the acetyl-CoA is bound. This study further confirms carnosol as a promising anti-breast cancer therapeutic compound and identifies it as a novel natural p300 inhibitor that could be added to the existing panel of inhibitors.
Collapse
Affiliation(s)
- Halima Alsamri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Hussain El Hasasna
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bincy Baby
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Aysha Alneyadi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed Akli Ayoub
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
32
|
Mitsiogianni M, Anestopoulos I, Kyriakou S, Trafalis DT, Franco R, Pappa A, Panayiotidis MI. Benzyl and phenethyl isothiocyanates as promising epigenetic drug compounds by modulating histone acetylation and methylation marks in malignant melanoma. Invest New Drugs 2021; 39:1460-1468. [PMID: 33963962 DOI: 10.1007/s10637-021-01127-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/04/2021] [Indexed: 11/30/2022]
Abstract
Melanoma is an aggressive skin cancer with increasing incidence rates globally. On the other hand, isothiocyanates are derived from cruciferous vegetables and are known to exert a wide range of anti-cancer activities including, among others, their ability to interact with the epigenome in order to supress cancer progression. The aim of this study was to determine the role of phenethyl and benzyl isothiocyanates in modulating histone acetylation and methylation as a potential epigenetic therapeutic strategy in an in vitro model of malignant melanoma. We report that both isothiocyanates induced cytotoxicity and influenced acetylation and methylation status of specific lysine residues on histones H3 and H4 by modulating the expression of various histone acetyltransferases, deacetylases and methyltransferases in malignant melanoma cells. Our data highlight novel insights on the interaction of isothiocyanates with components of the histone regulatory machinery in order to exert their anti-cancer action in malignant melanoma.
Collapse
Affiliation(s)
- Melina Mitsiogianni
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK
| | - Ioannis Anestopoulos
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Sotiris Kyriakou
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus.,The Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Clinical Pharmacology Unit, Medical School, National & Kapodistrian University of Athens, Athens, Greece
| | - Rodrigo Franco
- Redox Biology Centre, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.,School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Mihalis I Panayiotidis
- Department of Applied Sciences, Northumbria University, Newcastle Upon Tyne, UK. .,Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia, Cyprus. .,The Cyprus School of Molecular Medicine, Nicosia, Cyprus.
| |
Collapse
|
33
|
Krstic A, Konietzny A, Halasz M, Cain P, Oppermann U, Kolch W, Duffy DJ. A Chemo-Genomic Approach Identifies Diverse Epigenetic Therapeutic Vulnerabilities in MYCN-Amplified Neuroblastoma. Front Cell Dev Biol 2021; 9:612518. [PMID: 33968920 PMCID: PMC8097097 DOI: 10.3389/fcell.2021.612518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
Although a rare disease, neuroblastoma accounts for the highest proportion of childhood cancer deaths. There is a lack of recurrent somatic mutations in neuroblastoma embryonal tumours, suggesting a possible role for epigenetic alterations in driving this cancer. While an increasing number of reports suggest an association of MYCN with epigenetic machinery, the mechanisms of these interactions are poorly understood in the neuroblastoma setting. Utilising chemo-genomic approaches we revealed global MYCN-epigenetic interactions and identified numerous epigenetic proteins as MYCN targets. The epigenetic regulators HDAC2, CBX8 and CBP (CREBBP) were all MYCN target genes and also putative MYCN interactors. MYCN-related epigenetic genes included SMARCs, HDACs, SMYDs, BRDs and CREBBP. Expression levels of the majority of MYCN-related epigenetic genes showed predictive ability for neuroblastoma patient outcome. Furthermore, a compound library screen targeting epigenetic proteins revealed broad susceptibility of neuroblastoma cells to all classes of epigenetic regulators, belonging to families of bromodomains, HDACs, HATs, histone methyltransferases, DNA methyltransferases and lysin demethylases. Ninety-six percent of the compounds reduced MYCN-amplified neuroblastoma cell viability. We show that the C646 (CBP-bromodomain targeting compound) exhibits switch-like temporal and dose response behaviour and is effective at reducing neuroblastoma viability. Responsiveness correlates with MYCN expression, with MYCN-amplified cells being more susceptible to C646 treatment. Thus, exploiting the broad vulnerability of neuroblastoma cells to epigenetic targeting compounds represents an exciting strategy in neuroblastoma treatment, particularly for high-risk MYCN-amplified tumours.
Collapse
Affiliation(s)
- Aleksandar Krstic
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Anja Konietzny
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Centre for Molecular Neurobiology Hamburg (ZMNH), Emmy-Noether Group "Neuronal Protein Transport", University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melinda Halasz
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland
| | - Peter Cain
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, Institute of Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Udo Oppermann
- Botnar Research Centre, NIHR Oxford Biomedical Research Unit, Institute of Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Centre for Medicines Discovery, University of Oxford, Oxford, United Kingdom
| | - Walter Kolch
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - David J Duffy
- Systems Biology Ireland and Precision Oncology Ireland, School of Medicine, University College Dublin, Dublin, Ireland.,The Whitney Laboratory for Marine Bioscience and Sea Turtle Hospital, University of Florida, St. Augustine, FL, United States.,Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
34
|
Effects of Environmental Conditions on Nephron Number: Modeling Maternal Disease and Epigenetic Regulation in Renal Development. Int J Mol Sci 2021; 22:ijms22084157. [PMID: 33923831 PMCID: PMC8073167 DOI: 10.3390/ijms22084157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 12/14/2022] Open
Abstract
A growing body of evidence suggests that low nephron numbers at birth can increase the risk of chronic kidney disease or hypertension later in life. Environmental stressors, such as maternal malnutrition, medication and smoking, can influence renal size at birth. Using metanephric organ cultures to model single-variable environmental conditions, models of maternal disease were evaluated for patterns of developmental impairment. While hyperthermia had limited effects on renal development, fetal iron deficiency was associated with severe impairment of renal growth and nephrogenesis with an all-proximal phenotype. Culturing kidney explants under high glucose conditions led to cellular and transcriptomic changes resembling human diabetic nephropathy. Short-term high glucose culture conditions were sufficient for long-term alterations in DNA methylation-associated epigenetic memory. Finally, the role of epigenetic modifiers in renal development was tested using a small compound library. Among the selected epigenetic inhibitors, various compounds elicited an effect on renal growth, such as HDAC (entinostat, TH39), histone demethylase (deferasirox, deferoxamine) and histone methyltransferase (cyproheptadine) inhibitors. Thus, metanephric organ cultures provide a valuable system for studying metabolic conditions and a tool for screening for epigenetic modifiers in renal development.
Collapse
|
35
|
Gene Transactivation and Transrepression in MYC-Driven Cancers. Int J Mol Sci 2021; 22:ijms22073458. [PMID: 33801599 PMCID: PMC8037706 DOI: 10.3390/ijms22073458] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
MYC is a proto-oncogene regulating a large number of genes involved in a plethora of cellular functions. Its deregulation results in activation of MYC gene expression and/or an increase in MYC protein stability. MYC overexpression is a hallmark of malignant growth, inducing self-renewal of stem cells and blocking senescence and cell differentiation. This review summarizes the latest advances in our understanding of MYC-mediated molecular mechanisms responsible for its oncogenic activity. Several recent findings indicate that MYC is a regulator of cancer genome and epigenome: MYC modulates expression of target genes in a site-specific manner, by recruiting chromatin remodeling co-factors at promoter regions, and at genome-wide level, by regulating the expression of several epigenetic modifiers that alter the entire chromatin structure. We also discuss novel emerging therapeutic strategies based on both direct modulation of MYC and its epigenetic cofactors.
Collapse
|
36
|
Abstract
Neuroblastoma (NB) is a pediatric cancer of the sympathetic nervous system and one of the most common solid tumors in infancy. Amplification of MYCN, copy number alterations, numerical and segmental chromosomal aberrations, mutations, and rearrangements on a handful of genes, such as ALK, ATRX, TP53, RAS/MAPK pathway genes, and TERT, are attributed as underlying causes that give rise to NB. However, the heterogeneous nature of the disease-along with the relative paucity of recurrent somatic mutations-reinforces the need to understand the interplay of genetic factors and epigenetic alterations in the context of NB. Epigenetic mechanisms tightly control gene expression, embryogenesis, imprinting, chromosomal stability, and tumorigenesis, thereby playing a pivotal role in physio- and pathological settings. The main epigenetic alterations include aberrant DNA methylation, disrupted patterns of posttranslational histone modifications, alterations in chromatin composition and/or architecture, and aberrant expression of non-coding RNAs. DNA methylation and demethylation are mediated by DNA methyltransferases (DNMTs) and ten-eleven translocation (TET) proteins, respectively, while histone modifications are coordinated by histone acetyltransferases and deacetylases (HATs, HDACs), and histone methyltransferases and demethylases (HMTs, HDMs). This article focuses predominately on the crosstalk between the epigenome and NB, and the implications it has on disease diagnosis and treatment.
Collapse
Affiliation(s)
- Irfete S Fetahu
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090, Vienna, Austria.
| |
Collapse
|
37
|
He ZX, Wei BF, Zhang X, Gong YP, Ma LY, Zhao W. Current development of CBP/p300 inhibitors in the last decade. Eur J Med Chem 2021; 209:112861. [PMID: 33045661 DOI: 10.1016/j.ejmech.2020.112861] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 01/10/2023]
Abstract
CBP/p300, functioning as histone acetyltransferases and transcriptional co-factors, represents an attractive target for various diseases, including malignant tumor. The development of small-molecule inhibitors targeting the bromodomain and HAT domains of CBP/p300 has aroused broad interests of medicinal chemist in expectation of providing new hope for anti-cancer treatment. In particular, the CBP/p300 bromodomain inhibitor CCS1477, identified by CellCentric, is currently undergone clinical evaluation for the treatment of haematological malignancies and prostate cancer. In this review, we depict the development of CBP/p300 inhibitors reported from 2010 to 2020 and particularly highlight their structure-activity relationships (SARs), binding modes, selectivity and pharmacological functions with the aim to facilitate rational design and development of CBP/p300 inhibitors.
Collapse
Affiliation(s)
- Zhang-Xu He
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bing-Fei Wei
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xin Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yun-Peng Gong
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Li-Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
38
|
Wu D, Qiu Y, Jiao Y, Qiu Z, Liu D. Small Molecules Targeting HATs, HDACs, and BRDs in Cancer Therapy. Front Oncol 2020; 10:560487. [PMID: 33262941 PMCID: PMC7686570 DOI: 10.3389/fonc.2020.560487] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Evidence for research over the past decade shows that epigenetic regulation mechanisms run through the development and prognosis of tumors. Therefore, small molecular compounds targeting epigenetic regulation have become a research hotspot in the development of cancer therapeutic drugs. According to the obvious abnormality of histone acetylation when tumors occur, it suggests that histone acetylation modification plays an important role in the process of tumorigenesis. Currently, as a new potential anti-cancer therapeutic drugs, many active small molecules that target histone acetylation regulatory enzymes or proteins such as histone deacetylases (HDACs), histone acetyltransferase (HATs) and bromodomains (BRDs) have been developed to restore abnormal histone acetylation levels to normal. In this review, we will focus on summarizing the changes of histone acetylation levels during tumorigenesis, as well as the possible pharmacological mechanisms of small molecules that target histone acetylation in cancer treatment.
Collapse
Affiliation(s)
- Donglu Wu
- School of Clinical Medical, Changchun University of Chinese Medicine, Changchun, China.,Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Ye Qiu
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China.,School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Yunshuang Jiao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Zhidong Qiu
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China.,School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Da Liu
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China.,School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
39
|
Zafar A, Wang W, Liu G, Wang X, Xian W, McKeon F, Foster J, Zhou J, Zhang R. Molecular targeting therapies for neuroblastoma: Progress and challenges. Med Res Rev 2020; 41:961-1021. [PMID: 33155698 PMCID: PMC7906923 DOI: 10.1002/med.21750] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 01/09/2023]
Abstract
There is an urgent need to identify novel therapies for childhood cancers. Neuroblastoma is the most common pediatric solid tumor, and accounts for ~15% of childhood cancer‐related mortality. Neuroblastomas exhibit genetic, morphological and clinical heterogeneity, which limits the efficacy of existing treatment modalities. Gaining detailed knowledge of the molecular signatures and genetic variations involved in the pathogenesis of neuroblastoma is necessary to develop safer and more effective treatments for this devastating disease. Recent studies with advanced high‐throughput “omics” techniques have revealed numerous genetic/genomic alterations and dysfunctional pathways that drive the onset, growth, progression, and resistance of neuroblastoma to therapy. A variety of molecular signatures are being evaluated to better understand the disease, with many of them being used as targets to develop new treatments for neuroblastoma patients. In this review, we have summarized the contemporary understanding of the molecular pathways and genetic aberrations, such as those in MYCN, BIRC5, PHOX2B, and LIN28B, involved in the pathogenesis of neuroblastoma, and provide a comprehensive overview of the molecular targeted therapies under preclinical and clinical investigations, particularly those targeting ALK signaling, MDM2, PI3K/Akt/mTOR and RAS‐MAPK pathways, as well as epigenetic regulators. We also give insights on the use of combination therapies involving novel agents that target various pathways. Further, we discuss the future directions that would help identify novel targets and therapeutics and improve the currently available therapies, enhancing the treatment outcomes and survival of patients with neuroblastoma.
Collapse
Affiliation(s)
- Atif Zafar
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA.,Drug Discovery Institute, University of Houston, Houston, Texas, USA
| | - Gang Liu
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Xinjie Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Wa Xian
- Department of Biology and Biochemistry, Stem Cell Center, University of Houston, Houston, Texas, USA
| | - Frank McKeon
- Department of Biology and Biochemistry, Stem Cell Center, University of Houston, Houston, Texas, USA
| | - Jennifer Foster
- Department of Pediatrics, Texas Children's Hospital, Section of Hematology-Oncology Baylor College of Medicine, Houston, Texas, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Chemical Biology Program, University of Texas Medical Branch, Galveston, Texas, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA.,Drug Discovery Institute, University of Houston, Houston, Texas, USA
| |
Collapse
|
40
|
Xiong Y, Zhang M, Li Y. Recent Advances in the Development of CBP/p300 Bromodomain Inhibitors. Curr Med Chem 2020; 27:5583-5598. [DOI: 10.2174/0929867326666190731141055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022]
Abstract
CBP and p300 are two closely related Histone Acetyltransferases (HATs) that interact
with numerous transcription factors and act to increase the expression of their target genes. Both
proteins contain a bromodomain flanking the HAT catalytic domain that is important in binding of
CBP/p300 to chromatin, which offers an opportunity to develop protein-protein interaction inhibitors.
Since their discovery in 2006, CBP/p300 bromodomains have attracted much interest as promising
new epigenetic targets for diverse human diseases, including inflammation, cancer, autoimmune
disorders, and cardiovascular disease. Herein, we present a comprehensive review of the
structure, function, and inhibitors of CBP/p300 bromodomains developed in the last several years,
which is expected to be beneficial to relevant studies.
Collapse
Affiliation(s)
- Ying Xiong
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Mingming Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yingxia Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
41
|
Abstract
Acetylation was initially discovered as a post-translational modification (PTM) on the unstructured, highly basic N-terminal tails of eukaryotic histones in the 1960s. Histone acetylation constitutes part of the "histone code", which regulates chromosome compaction and various DNA processes such as gene expression, recombination, and DNA replication. In bacteria, nucleoid-associated proteins (NAPs) are responsible these functions in that they organize and compact the chromosome and regulate some DNA processes. The highly conserved DNABII family of proteins are considered functional homologues of eukaryotic histones despite having no sequence or structural conservation. Within the past decade, a growing interest in Nε-lysine acetylation led to the discovery that hundreds of bacterial proteins are acetylated with diverse cellular functions, in direct contrast to the original thought that this was a rare phenomenon. Similarly, other previously undiscovered bacterial PTMs, like serine, threonine, and tyrosine phosphorylation, have also been characterized. In this review, the various PTMs that were discovered among DNABII family proteins, specifically histone-like protein (HU) orthologues, from large-scale proteomic studies are discussed. The functional significance of these modifications and the enzymes involved are also addressed. The discovery of novel PTMs on these proteins begs this question: is there a histone-like code in bacteria?
Collapse
Affiliation(s)
- Valerie J Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, New Jersey 08103, United States
| |
Collapse
|
42
|
Shanmugam MK, Dharmarajan A, Warrier S, Bishayee A, Kumar AP, Sethi G, Ahn KS. Role of histone acetyltransferase inhibitors in cancer therapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 125:149-191. [PMID: 33931138 DOI: 10.1016/bs.apcsb.2020.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of cancer is a complex phenomenon driven by various extrinsic as well as intrinsic risk factors including epigenetic modifications. These post-translational modifications are encountered in diverse cancer cells and appear for a relatively short span of time. These changes can significantly affect various oncogenic genes and proteins involved in cancer initiation and progression. Histone lysine acetylation and deacetylation processes are controlled by two opposing classes of enzymes that modulate gene regulation either by adding an acetyl moiety on a histone lysine residue by histone lysine acetyltransferases (KATs) or via removing it by histone deacetylases (KDACs). Deregulated KAT activity has been implicated in the development of several diseases including cancer and can be targeted for the development of anti-neoplastic drugs. Here, we describe the predominant epigenetic changes that can affect key KAT superfamily members during carcinogenesis and briefly highlight the pharmacological potential of employing lysine acetyltransferase inhibitors (KATi) for cancer therapy.
Collapse
Affiliation(s)
- Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arunasalam Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences Technology and Research, Sri Ramachandra Institute of Higher Education & Research, Chennai, India
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal University, Bangalore, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov 2020; 19:776-800. [PMID: 32929243 DOI: 10.1038/s41573-020-0077-5] [Citation(s) in RCA: 368] [Impact Index Per Article: 73.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2020] [Indexed: 01/10/2023]
Abstract
Dysregulation of the epigenome drives aberrant transcriptional programmes that promote cancer onset and progression. Although defective gene regulation often affects oncogenic and tumour-suppressor networks, tumour immunogenicity and immune cells involved in antitumour responses may also be affected by epigenomic alterations. This could have important implications for the development and application of both epigenetic therapies and cancer immunotherapies, and combinations thereof. Here, we review the role of key aberrant epigenetic processes - DNA methylation and post-translational modification of histones - in tumour immunogenicity, as well as the effects of epigenetic modulation on antitumour immune cell function. We emphasize opportunities for small-molecule inhibitors of epigenetic regulators to enhance antitumour immune responses, and discuss the challenges of exploiting the complex interplay between cancer epigenetics and cancer immunology to develop treatment regimens combining epigenetic therapies with immunotherapies.
Collapse
|
44
|
Gujral P, Mahajan V, Lissaman AC, Ponnampalam AP. Histone acetylation and the role of histone deacetylases in normal cyclic endometrium. Reprod Biol Endocrinol 2020; 18:84. [PMID: 32791974 PMCID: PMC7425564 DOI: 10.1186/s12958-020-00637-5] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022] Open
Abstract
Histone acetylation is a critical epigenetic modification that changes chromatin architecture and regulates gene expression by opening or closing the chromatin structure. It plays an essential role in cell cycle progression and differentiation. The human endometrium goes through cycles of regeneration, proliferation, differentiation, and degradation each month; each phase requiring strict epigenetic regulation for the proper functioning of the endometrium. Aberrant histone acetylation and alterations in levels of two acetylation modulators - histone acetylases (HATs) and histone deacetylases (HDACs) - have been associated with endometrial pathologies such as endometrial cancer, implantation failures, and endometriosis. Thus, histone acetylation is likely to have an essential role in the regulation of endometrial remodelling throughout the menstrual cycle.
Collapse
Affiliation(s)
- Palak Gujral
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Vishakha Mahajan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Abbey C Lissaman
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Anna P Ponnampalam
- The Liggins Institute, The University of Auckland, Auckland, New Zealand.
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
45
|
Idrissou M, Sanchez A, Penault-Llorca F, Bignon YJ, Bernard-Gallon D. Epi-drugs as triple-negative breast cancer treatment. Epigenomics 2020; 12:725-742. [PMID: 32396394 DOI: 10.2217/epi-2019-0312] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Triple-negative breast cancer (TNBC) types with poor prognosis are due to the absence of estrogen receptors, progesterone receptors and HEGFR-2. The lack of suitable therapy for TNBC has led the research community to turn toward epigenetic regulation and its protagonists that can modulate certain oncogenes and tumor suppressors. This has opened an important new field of therapy using epi-drugs, in preclinical and clinical trials. The epi-drugs are natural or synthetic molecules capable of inhibiting or modulating the activity of epigenetic proteins such as DNA methyltransferases, modulating the expression of interferon microRNAs, as well as histone methyltransferases, demethylases, acetyltransferases and deacetylases. This review investigated the epi-drugs used in the treatment of TNBC.
Collapse
Affiliation(s)
- Mouhamed Idrissou
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri-Dunant, Clermont-Ferrand 63001, France.,INSERM U 1240 Molecular Imagery & Theranostic Strategies (IMoST), 58 Rue Montalembert, Clermont-Ferrand 63005, France
| | - Anna Sanchez
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri-Dunant, Clermont-Ferrand 63001, France.,INSERM U 1240 Molecular Imagery & Theranostic Strategies (IMoST), 58 Rue Montalembert, Clermont-Ferrand 63005, France
| | - Frédérique Penault-Llorca
- INSERM U 1240 Molecular Imagery & Theranostic Strategies (IMoST), 58 Rue Montalembert, Clermont-Ferrand 63005, France.,Department of Biopathology, Centre Jean Perrin, 58 Rue Montalembert, Clermont-Ferrand 63011, France
| | - Yves-Jean Bignon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri-Dunant, Clermont-Ferrand 63001, France.,INSERM U 1240 Molecular Imagery & Theranostic Strategies (IMoST), 58 Rue Montalembert, Clermont-Ferrand 63005, France
| | - Dominique Bernard-Gallon
- Department of Oncogenetics, Centre Jean Perrin, CBRV, 28 place Henri-Dunant, Clermont-Ferrand 63001, France.,INSERM U 1240 Molecular Imagery & Theranostic Strategies (IMoST), 58 Rue Montalembert, Clermont-Ferrand 63005, France
| |
Collapse
|
46
|
Kozako T, Itoh Y, Honda SI, Suzuki T. Epigenetic Control Using Small Molecules in Cancer. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/978-3-030-32857-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
47
|
Westphal M, Sant P, Hauser AT, Jung M, Driever W. Chemical Genetics Screen Identifies Epigenetic Mechanisms Involved in Dopaminergic and Noradrenergic Neurogenesis in Zebrafish. Front Genet 2020; 11:80. [PMID: 32158467 PMCID: PMC7052299 DOI: 10.3389/fgene.2020.00080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/24/2020] [Indexed: 12/17/2022] Open
Abstract
The cell type diversity and complexity of the nervous system is generated by a network of signaling events, transcription factors, and epigenetic regulators. Signaling and transcriptional control have been easily amenable to forward genetic screens in model organisms like zebrafish. In contrast, epigenetic mechanisms have been somewhat elusive in genetic screens, likely caused by broad action in multiple developmental pathways that masks specific phenotypes, but also by genetic redundancies of epigenetic factors. Here, we performed a screen using small molecule inhibitors of epigenetic mechanisms to reveal contributions to specific aspects of neurogenesis in zebrafish. We chose development of dopaminergic and noradrenergic neurons from neural progenitors as target of epigenetic regulation. We performed the screen in two phases: First, we tested a small molecule inhibitor library that targets a broad range of epigenetic protein classes and mechanisms, using expression of the dopaminergic and noradrenergic marker tyrosine hydroxylase as readout. We identified 10 compounds, including HDAC, Bromodomain and HAT inhibitors, which interfered with dopaminergic and noradrenergic development in larval zebrafish. In the second screening phase, we aimed to identify neurogenesis stages affected by these 10 inhibitors. We analyzed treated embryos for effects on neural stem cells, growth progression of the retina, and apoptosis in neural tissues. In addition, we analyzed effects on islet1 expressing neuronal populations to determine potential selectivity of compounds for transmitter phenotypes. In summary, our targeted screen of epigenetic inhibitors identified specific compounds, which reveal chromatin regulator classes that contribute to dopaminergic and noradrenergic neurogenesis in vivo.
Collapse
Affiliation(s)
- Markus Westphal
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS and BIOSS—Centres for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Pooja Sant
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Alexander-Thomas Hauser
- Chemical Epigenetics Group, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Manfred Jung
- Chemical Epigenetics Group, Institute of Pharmaceutical Sciences, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS—Centre for Integrative Biological SignallingStudies, University of Freiburg, Freiburg, Germany
| | - Wolfgang Driever
- Developmental Biology, Faculty of Biology, Institute Biology 1, Albert Ludwigs University Freiburg, Freiburg, Germany
- CIBSS and BIOSS—Centres for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Mustachio LM, Roszik J, Farria A, Dent SYR. Targeting the SAGA and ATAC Transcriptional Coactivator Complexes in MYC-Driven Cancers. Cancer Res 2020; 80:1905-1911. [PMID: 32094302 DOI: 10.1158/0008-5472.can-19-3652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/28/2020] [Accepted: 02/19/2020] [Indexed: 12/26/2022]
Abstract
Targeting epigenetic regulators, such as histone-modifying enzymes, provides novel strategies for cancer therapy. The GCN5 lysine acetyltransferase (KAT) functions together with MYC both during normal development and in oncogenesis. As transcription factors, MYC family members are difficult to target with small-molecule inhibitors, but the acetyltransferase domain and the bromodomain in GCN5 might provide alternative targets for disruption of MYC-driven functions. GCN5 is part of two distinct multiprotein histone-modifying complexes, SAGA and ATAC. This review summarizes key findings on the roles of SAGA and ATAC in embryo development and in cancer to better understand the functional relationships of these complexes with MYC family members, as well as their future potential as therapeutic targets.
Collapse
Affiliation(s)
- Lisa Maria Mustachio
- Departments of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason Roszik
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aimee Farria
- Departments of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sharon Y R Dent
- Departments of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Center for Cancer Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
49
|
Yang Y, Zhang R, Li Z, Mei L, Wan S, Ding H, Chen Z, Xing J, Feng H, Han J, Jiang H, Zheng M, Luo C, Zhou B. Discovery of Highly Potent, Selective, and Orally Efficacious p300/CBP Histone Acetyltransferases Inhibitors. J Med Chem 2020; 63:1337-1360. [PMID: 31910017 DOI: 10.1021/acs.jmedchem.9b01721] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
p300 and CREB-binding protein (CBP) are ubiquitously expressed pleiotropic lysine acetyltransferases and play a key role as transcriptional co-activators that are essential for a multitude of cellular processes. Despite great importance, there is a lack of highly selective, potent, druglike p300/CBP inhibitors. Through the artificial-intelligence-assisted drug discovery pipeline and further optimization, we reported the discovery of novel, highly selective, potent small-molecule inhibitors of p300/CBP histone acetyltransferases (HAT) with desired druglike properties, exemplified by B026. Our data demonstrated that B026, with half maximal inhibitory concentration (IC50) values of 1.8 nM to p300 and 9.5 nM to CBP enzyme inhibitory activity, is the most potent, selective p300/CBP HAT inhibitor. Moreover, B026 achieves significant and dose-dependent tumor growth inhibition in an animal model of human cancer, suggesting that B026 is a highly promising p300/CBP HAT inhibitor and warrants extensive preclinical investigation as a potential clinical development candidate.
Collapse
Affiliation(s)
- Yaxi Yang
- Department of Medicinal Chemistry, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Rukang Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Zhaojun Li
- School of Information Management , Dezhou University , No. 566 University Road West , Dezhou 253023 , Shandong , China
| | - Lianghe Mei
- Suzhou Institute of Drug Innovation , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 108 Yuxin Road , Suzhou 213000 , Jiangsu , China
| | - Shili Wan
- Department of Medicinal Chemistry, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Hong Ding
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Zhifeng Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Jing Xing
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Huijin Feng
- Department of Medicinal Chemistry, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Jie Han
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Hualiang Jiang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Mingyue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
- University of Chinese Academy of Sciences , 19 Yuquan Road , Beijing 100049 , China
| | - Cheng Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
- University of Chinese Academy of Sciences , 19 Yuquan Road , Beijing 100049 , China
| | - Bing Zhou
- Department of Medicinal Chemistry, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zuchongzhi Road , Shanghai 201203 , China
- University of Chinese Academy of Sciences , 19 Yuquan Road , Beijing 100049 , China
- Hangzhou Institute for Advanced Study , University of Chinese Academy of Sciences , Hangzhou 310024 , China
| |
Collapse
|
50
|
Zhang L, Wang W, Zhang S, Wang Y, Guo W, Liu Y, Wang Y, Zhang Y. Identification of lysine acetylome in cervical cancer by label-free quantitative proteomics. Cancer Cell Int 2020; 20:182. [PMID: 32489318 PMCID: PMC7247262 DOI: 10.1186/s12935-020-01266-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lysine acetylation is a post-translational modification that regulates a diversity of biological processes, including cancer development. METHODS Here, we performed the quantitative acetylproteomic analysis of three primary cervical cancer tissues and corresponding adjacent normal tissues by using the label-free proteomics approach. RESULTS We identified a total of 928 lysine acetylation sites from 1547 proteins, in which 495 lysine acetylation sites corresponding to 296 proteins were quantified. Further, 41 differentially expressed lysine acetylation sites corresponding to 30 proteins were obtained in cervical cancer tissues compared with adjacent normal tissues (Fold change > 2 and P < 0.05), of which 1 was downregulated, 40 were upregulated. Moreover, 75 lysine acetylation sites corresponding to 58 proteins were specifically detected in cancer tissues or normal adjacent tissues. Motif-X analysis showed that kxxxkxxxk, GkL, AxxEk, kLxE, and kkxxxk are the most enriched motifs with over four-fold increases when compared with the background matches. KEGG analysis showed that proteins identified from differently and specifically expressed peptides may influence key pathways, such as Notch signaling pathway, viral carcinogenesis, RNA transport, and Jak-STAT, which play an important role in tumor progression. Furthermore, the acetylated levels of CREBBP and S100A9 in cervical cancer tissues were confirmed by immunoprecipitation (IP) and Western blot analysis. CONCLUSIONS Taken together, our data provide novel insights into the role of protein lysine acetylation in cervical carcinogenesis.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Wanyue Wang
- School of Basic Medical Sciences, Qiqihar Medical University, Qiqihar, 161006 Heilongjiang China
| | - Shanqiang Zhang
- Medical Research Center, Yue Bei People’s Hospital Affiliated to Shantou University Medical College, Shaoguan, 512025 Guangdong China
| | - Yuxin Wang
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Weikang Guo
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yunduo Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yaoxian Wang
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yunyan Zhang
- Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| |
Collapse
|