1
|
Li B, Chen Y, Wang S, Jin B, Yang J, Niu Q, Hao G, Wang N, Zhang W, Zhao L, Wen J, Liu D. Discovery of 4,5-dihydro-benzo[g]indazole-based hydroxamic acids as HDAC3/BRD4 dual inhibitors and anti-tumor agents. Eur J Med Chem 2025; 285:117230. [PMID: 39764880 DOI: 10.1016/j.ejmech.2024.117230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 02/04/2025]
Abstract
Concurrent inhibition of HDAC and BRD4, two well-established epigenetic targets for anti-tumor therapy, demonstrates the potential to enhance anti-tumor effects synergistically. The present study involves the development of a series of novel HDAC3/BRD4 dual inhibitors, followed by evaluation of their antitumor efficacy against several tumor models. Guided by scaffold hopping strategy, key pharmacophore of BRD4 inhibitor I-BET-151 was incorporated into an in-house developed HDAC3-selective inhibitor 17h. A set of twenty-two compounds was synthesized and characterized. Most of these compounds demonstrated significant potency in inhibiting HDAC3 and exhibited selectivity over its closely-related isoform, HDAC1. The potent BRD4 inhibition of these compounds has been further confirmed through HTFR and thermal shift assays. Of which, compounds 26b and 26n demonstrated potent dual inhibition against HDAC3 and BRD4. Compound 26n demonstrated potent antiproliferative effects against a panel of cancer cells, with human pancreatic cancer cell line Capan-1 displaying the highest susceptibility. Compound 26n exhibited significant upregulation of Ac-H3 and downregulation of c-Myc at concentrations as low as 1 μM, suggesting proper target engagement in Capan-1 cells. Compound 26n demonstrated significant antitumor efficacy in Capan-1 CDX model, with a tumor growth inhibition rate of 71 % under the given dosing regimen. In summary, this research highlights the promising therapeutic potential of benzodihydroindazole derivatives as HDAC3/BRD4 dual inhibitors, warranting further investigation.
Collapse
Affiliation(s)
- Bo Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yibing Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Siyuan Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bo Jin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jinyu Yang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qun Niu
- Institute of Shandong Xinhua Pharmaceutical, Shandong Xinhua Pharmaceutical Co., Ltd., Zibo, 255000, China
| | - Guizhou Hao
- Shandong Engineering Research Center of Complex Injectables, Shangdong New Time Pharmaceutical Co., Ltd., Linyi, 273400, China
| | - Ning Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wenchao Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jiachen Wen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
2
|
Kao WH, Chiu KY, Tsai SCS, Teng CLJ, Oner M, Lai CH, Hsieh JT, Lin CC, Wang HY, Chen MC, Lin H. PI3K/Akt inhibition promotes AR activity and prostate cancer cell proliferation through p35-CDK5 modulation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167568. [PMID: 39536992 DOI: 10.1016/j.bbadis.2024.167568] [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: 04/01/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Aberrant PI3K/Akt activation is linked to prostate cancer (PCa) malignancy, while androgen receptor (AR) is critical in early-stage PCa development. Investigating the interaction between these pathways is crucial for PCa malignancy. Our previous study demonstrated that p35-CDK5 mediates post-translational modifications of AR, STAT3, and p21CIP1, eventually promoting PCa cell growth. This study revealed the role of p35-CDK5 in between PI3K/Akt and AR by utilizing LNCaP and 22Rv1 cells. Through the TCGA database analysis, we observed a positive correlation between PTEN and p35 expression, implying a potential negative correlation between PI3K/Akt activation and p35-CDK5. Inhibiting PI3K/Akt with LY294002, Capivasertib (AZD5363), or using an inactive Akt mutant significantly increased p35 expression and subsequently enhanced AR stability and activation in PCa cells. On the other hand, CDK5-knockdown reversed these effects. The involvement of the β-catenin/Egr1-axis was observed in regulating PI3K/Akt inhibition and p35-CDK5 activation, implying a possible mechanistic connection. Importantly, CDK5 knockdown further reduced PI3K/Akt-inhibition-induced AR and cell viability maintenance, suggesting a compensatory role for CDK5-AR in maintaining cell viability under Akt inhibition. In conclusion, PI3K/Akt inhibition could trigger p35-CDK5-dependent AR activation and cell viability, highlighting p35-CDK5 as a critical link connecting PI3K/Akt inhibition to AR activation and pivotal in PCa cell resistance to PI3K/Akt blockade.
Collapse
Affiliation(s)
- Wei-Hsiang Kao
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; Translational Cell Therapy Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Stella Chin-Shaw Tsai
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan; Superintendent Office, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan; College of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chieh-Lin Jerry Teng
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan; Division of Hematology/Medical Oncology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan.
| | - Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, TX75390, USA.
| | - Chi-Chien Lin
- Institute of Biomedical Science, National Chung Hsing University, Taichung 40227, Taiwan
| | - Hsin-Yi Wang
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan.
| | - Mei-Chih Chen
- Translational Cell Therapy Center, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
3
|
Wang C, Ma X. The role of acetylation and deacetylation in cancer metabolism. Clin Transl Med 2025; 15:e70145. [PMID: 39778006 PMCID: PMC11706801 DOI: 10.1002/ctm2.70145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
As a hallmark of cancer, metabolic reprogramming adjusts macromolecular synthesis, energy metabolism and redox homeostasis processes to adapt to and promote the complex biological processes of abnormal growth and proliferation. The complexity of metabolic reprogramming lies in its precise regulation by multiple levels and factors, including the interplay of multiple signalling pathways, precise regulation of transcription factors and dynamic adjustments in metabolic enzyme activity. In this complex regulatory network, acetylation and deacetylation, which are important post-translational modifications, regulate key molecules and processes related to metabolic reprogramming by affecting protein function and stability. Dysregulation of acetylation and deacetylation may alter cancer cell metabolic patterns by affecting signalling pathways, transcription factors and metabolic enzyme activity related to metabolic reprogramming, increasing the susceptibility to rapid proliferation and survival. In this review, we focus on discussing how acetylation and deacetylation regulate cancer metabolism, thereby highlighting the central role of these post-translational modifications in metabolic reprogramming, and hoping to provide strong support for the development of novel cancer treatment strategies. KEY POINTS: Protein acetylation and deacetylation are key regulators of metabolic reprogramming in tumour cells. These modifications influence signalling pathways critical for tumour metabolism. They modulate the activity of transcription factors that drive gene expression changes. Metabolic enzymes are also affected, altering cellular metabolism to support tumour growth.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaoxin Ma
- Department of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyang CityLiaoning ProvinceChina
- Key Laboratory of Gynecological Oncology of Liaoning ProvinceDepartment of Obstetrics and GynecologyShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
4
|
Chen H, Qin A, Xu F, Guo S, Zhang G, Zhang A, Li W, Tian F, Zheng Q. HDAC3 inhibitors induce drug resistance by promoting IL-17 A production by T cells. Sci Rep 2024; 14:31937. [PMID: 39738540 PMCID: PMC11685772 DOI: 10.1038/s41598-024-83447-8] [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: 08/09/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
HDAC3 has been demonstrated to play a crucial role in the progression of various tumors and the differentiation and development of T cells. However, its impact on peripheral T cells in the development of murine lung cancer remains unclear. In this experiment, a subcutaneous lung tumor model was established in C57BL/6 mice, and tumor-bearing mice were treated with the specific inhibitor of HDAC3, RGFP966, at different doses to observe changes in tumor size. Additionally, a lung tumor model was established using hdac3fl/flcd4cre+/+ mice to investigate its mechanism. Mice injected with 10 mg/kg RGFP966 had the smallest tumor volume, while those injected with 30 mg/kg RGFP966 had the largest tumors. Flow cytometry analysis revealed that the expression of HDAC3 in splenic T cells was reduced in all groups of mice, while IFN-γ and IL-17 A were increased. Moreover, the expression of granzyme B and perforin in splenic CD8+ T cells was increased in all groups of mice. Compared to the use of 30 mg/kg RGFP966 alone, the combination with anti-IL-17 A mAb reduced the infiltration of Neutrophils and exhausted T cells in mouse tumors, thereby impeding tumor development. These findings demonstrate that the use of RGFP966 or T cell-specific loss of hdac3 promotes the expression of IL-17 A in splenic T cells, leading to tumor resistance and providing insights for clinical treatment.
Collapse
Affiliation(s)
- Hao Chen
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Anqi Qin
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Fan Xu
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Shuai Guo
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China
| | - Ge Zhang
- School of Basic Medical, Xingtai Medical College, Xingtai, 054000, China
| | - Aihong Zhang
- Department of ICU, The Affiliated Hospital of North China University of Science and Technology, Tangshan, 063000, China
| | - WenTing Li
- Department of Laboratory Animal Science, Health Science Center, Peking University, Beijing, 100083, China
| | - Feng Tian
- Department of Laboratory Animal Science, Health Science Center, Peking University, Beijing, 100083, China
| | - Quanhui Zheng
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
5
|
Li Y, Fan A, Zhang Y, Meng W, Pan W, Wu F, Ma Z, Chen W. Circular RNA hsa_circ_0001610 promotes prostate cancer progression by sponging miR-1324 and upregulating PTK6. Gene 2024; 930:148818. [PMID: 39098513 DOI: 10.1016/j.gene.2024.148818] [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: 03/12/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Prostate cancer (PCa) incidence and cancer-related deaths are both high in the male population. Once castration-resistant prostate cancer (CRPC) has developed, PCa can be difficult to manage. Circular RNAs (circRNAs) play essential roles in the regulation of carcinogenesis and cancer progression. In CRPC, however, the potential molecular mechanisms and biological functions of circRNAs are yet to be defined. In this study, we conducted RNA sequencing on four hormone-sensitive prostate cancer (HSPC) tumor tissue samples and three CRPC samples. We recognized hsa_circ_0001610, a novel circRNA that was highly expressed in the cells and tissue of CRPC. We used quantitative real-time PCR (qRT-PCR) to evaluate hsa_circ_0001610 expression. We conducted in vivo and in vitro experiments and found that hsa_circ_0001610 overexpression caused PCa cells to proliferate and migrate and caused enzalutamide resistance. In contrast, the opposite results were found for hsa_circ_0001610 knockdown. We used Western blot, dual-luciferase reporter assays, RNA immunoprecipitation (RIP), qRT-PCR, and rescue experiments to reveal the underlying mechanisms of hsa_circ_0001610. Mechanistically, hsa_circ_0001610 acted as a molecular sponge for miR-1324 and thus reversed its inhibitory effect on its target gene PTK6. As a result, the PTK6 expression was enhanced, which accelerated PCa progression. The findings of this study confirmed that hsa_circ_0001610 drives the progression of PCa through the hsa_circ_0001610/miR-1324/PTK6 axis. Thus, hsa_circ_0001610 is potentially an effective therapeutic target and specific biomarker for advanced PCa.
Collapse
Affiliation(s)
- Yunpeng Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Aoyu Fan
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Yunyan Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Wei Meng
- Lab for Noncoding RNA and Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Wei Pan
- Lab for Noncoding RNA and Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Fan Wu
- Lab for Noncoding RNA and Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Zhongliang Ma
- Lab for Noncoding RNA and Cancer, School of Life Sciences, Shanghai University, Shanghai 200444, China.
| | - Wei Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200030, China.
| |
Collapse
|
6
|
Zou W, Huang C, Chen Y, Tang J, Li Q, Fang Q, Ma Y, Wu W, Feng S. Role of HDAC3 in the epithelial-mesenchymal transition of retinal pigment epithelium cells: Implications for proliferative vitreoretinopathy. Heliyon 2024; 10:e39333. [PMID: 39524785 PMCID: PMC11543911 DOI: 10.1016/j.heliyon.2024.e39333] [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: 03/05/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Proliferative vitreoretinopathy(PVR) is a type of fibrotic eye disease with a poor clinical prognosis. Increasing evidence has shown that the primary pathological mechanism of PVR is the epithelial-mesenchymal transition(EMT) of retinal pigment epithelium(RPE) cells. Histone deacetylase 3(HDAC3) is a crucial enzyme involved in regulating the acetylation level of proteins. Several studies have reported associations between HDAC3 levels and EMT in various tumors; however, the specific effect of HDAC3 on PVR remains largely unknown. The current study found that HDAC3 was highly expressed in both human PVR membranes and experimental PVR. In vivo, silencing HDAC3 in RPE cells reduced their ability to develop experimental PVR through suppression of EMT. In vitro, inhibition of HDAC3 in RPE cells suppressed EGF-mediated cell proliferation, migration, and EMT. Additionally, overexpression of HDAC3 in RPE cells promoted cell proliferation, migration, and EMT. Mechanistically, the results of chromatin immunoprecipitation(ChIP) and luciferase assays revealed a direct binding of the transcription factor MAZ to the promoter region of HDAC3, thereby promoting its transcription. Furthermore, It was demonstrated that HDAC3 facilitated EMT by interacting with AKT and contributing to its deacetylation. In summary, our findings indicated the involvement of HDAC3 in the EMT of RPE cells, as well as its role in PVR through the regulation of the AKT pathway. These results suggested that targeting HDAC3 could be a potential strategy for preventing and treating PVR.
Collapse
Affiliation(s)
- Weikang Zou
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Chunling Huang
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Yuting Chen
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Jing Tang
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Qiqi Li
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Qi Fang
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Yulin Ma
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Wei Wu
- Department of Ophthalmology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Songfu Feng
- Department of Ophthalmology, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| |
Collapse
|
7
|
Kundu M, Das S, Dey A, Mandal M. Dual perspective on autophagy in glioma: Detangling the dichotomous mechanisms of signaling pathways for therapeutic insights. Biochim Biophys Acta Rev Cancer 2024; 1879:189168. [PMID: 39121913 DOI: 10.1016/j.bbcan.2024.189168] [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: 04/16/2024] [Revised: 06/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Autophagy is a normal physiological process that aids the recycling of cellular nutrients, assisting the cells to cope with stressed conditions. However, autophagy's effect on cancer, including glioma, is uncertain and involves complicated molecular mechanisms. Several contradictory reports indicate that autophagy may promote or suppress glioma growth and progression. Autophagy inhibitors potentiate the efficacy of chemotherapy or radiation therapy in glioma. Numerous compounds stimulate autophagy to cause glioma cell death. Autophagy is also involved in the therapeutic resistance of glioma. This review article aims to detangle the complicated molecular mechanism of autophagy to provide a better perception of the two-sided role of autophagy in glioma and its therapeutic implications. The protein and epigenetic modulators of the cytoprotective and cytotoxic role of autophagy are described in this article. Moreover, several signaling pathways are associated with autophagy and its effects on glioma. We have reviewed the molecular pathways and highlighted the signaling axis involved in cytoprotective and cytotoxic autophagy. Additionally, this article discusses the role of autophagy in therapeutic resistance, including glioma stem cell maintenance and tumor microenvironment regulation. It also summarizes several investigations on the anti-glioma effects of autophagy modulators to understand the associated mechanisms and provide insights regarding its therapeutic implications.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; Center for Multidisciplinary Research & Innovations, Brainware University, Barasat, India; Department of Pharmaceutical Technology, Brainware University, Barasat, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; Department of Allied Health Sciences, Brainware University, Barasat, India
| | - Ankita Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
8
|
Li C, Yi Y, Ouyang Y, Chen F, Lu C, Peng S, Wang Y, Chen X, Yan X, Xu H, Li S, Feng L, Xie X. TORSEL, a 4EBP1-based mTORC1 live-cell sensor, reveals nutrient-sensing targeting by histone deacetylase inhibitors. Cell Biosci 2024; 14:68. [PMID: 38824577 PMCID: PMC11143692 DOI: 10.1186/s13578-024-01250-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Mammalian or mechanistic target of rapamycin complex 1 (mTORC1) is an effective therapeutic target for diseases such as cancer, diabetes, aging, and neurodegeneration. However, an efficient tool for monitoring mTORC1 inhibition in living cells or tissues is lacking. RESULTS We developed a genetically encoded mTORC1 sensor called TORSEL. This sensor changes its fluorescence pattern from diffuse to punctate when 4EBP1 dephosphorylation occurs and interacts with eIF4E. TORSEL can specifically sense the physiological, pharmacological, and genetic inhibition of mTORC1 signaling in living cells and tissues. Importantly, TORSEL is a valuable tool for imaging-based visual screening of mTORC1 inhibitors. Using TORSEL, we identified histone deacetylase inhibitors that selectively block nutrient-sensing signaling to inhibit mTORC1. CONCLUSIONS TORSEL is a unique living cell sensor that efficiently detects the inhibition of mTORC1 activity, and histone deacetylase inhibitors such as panobinostat target mTORC1 signaling through amino acid sensing.
Collapse
Affiliation(s)
- Canrong Li
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuguo Yi
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yingyi Ouyang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Fengzhi Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Chuxin Lu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shujun Peng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yifan Wang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xinyu Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xiao Yan
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Haolun Xu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shuiming Li
- College of Life Sciences and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen, China
| | - Lin Feng
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoduo Xie
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China.
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
9
|
Patel P, Shrivastava SK, Sharma P, Kurmi BD, Shirbhate E, Rajak H. Hydroxamic acid derivatives as selective HDAC3 inhibitors: computer-aided drug design strategies. J Biomol Struct Dyn 2024; 42:362-383. [PMID: 36995068 DOI: 10.1080/07391102.2023.2192804] [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: 09/01/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023]
Abstract
Histone deacetylases (HDACs) are critical epigenetic drug targets that have gained significant attention in the scientific community for the treatment of cancer. The currently marketed HDAC inhibitors lack selectivity for the various HDAC isoenzymes. Here, we describe our protocol for the discovery of novel potential hydroxamic acid based HDAC3 inhibitors through pharmacophore modeling, virtual screening, docking, molecular dynamics (MD) simulation and toxicity studies. The ten pharmacophore hypotheses were established, and their reliability was validated by different ROC (receiving operator curve) analysis. Among them, the best model (Hypothesis 9 or RRRA) was employed for searching SCHEMBL, ZINC and MolPort database to screen out hit molecules as selective HDAC3 inhibitors, followed by different docking stages. MD simulation (50 ns) and MMGBSA study were performed to study the stability of ligand binding modes and with the help of trajectory analysis, to calculate the ligand-receptor complex RMSD (root-mean-square deviation), RMSF (root-mean-square fluctuation) and H-bond distance, etc. Finally, in-silico toxicity studies were performed on top screened molecules and compared with reference drug SAHA and established structure-activity relationship (SAR). The results indicated that compound 31, with high inhibitory potency and less toxicity (probability value 0.418), is suitable for further experimental analysis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Preeti Patel
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Guru Ghasidas University, Bilaspur, Chhattisgarh, India
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Sushant Kumar Shrivastava
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Piyoosh Sharma
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Ekta Shirbhate
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Guru Ghasidas University, Bilaspur, Chhattisgarh, India
| | - Harish Rajak
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Guru Ghasidas University, Bilaspur, Chhattisgarh, India
| |
Collapse
|
10
|
Raith F, O’Donovan DH, Lemos C, Politz O, Haendler B. Addressing the Reciprocal Crosstalk between the AR and the PI3K/AKT/mTOR Signaling Pathways for Prostate Cancer Treatment. Int J Mol Sci 2023; 24:ijms24032289. [PMID: 36768610 PMCID: PMC9917236 DOI: 10.3390/ijms24032289] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
The reduction in androgen synthesis and the blockade of the androgen receptor (AR) function by chemical castration and AR signaling inhibitors represent the main treatment lines for the initial stages of prostate cancer. Unfortunately, resistance mechanisms ultimately develop due to alterations in the AR pathway, such as gene amplification or mutations, and also the emergence of alternative pathways that render the tumor less or, more rarely, completely independent of androgen activation. An essential oncogenic axis activated in prostate cancer is the phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, as evidenced by the frequent alterations of the negative regulator phosphatase and tensin homolog (PTEN) and by the activating mutations in PI3K subunits. Additionally, crosstalk and reciprocal feedback loops between androgen signaling and the PI3K/AKT/mTOR signaling cascade that activate pro-survival signals and play an essential role in disease recurrence and progression have been evidenced. Inhibitors addressing different players of the PI3K/AKT/mTOR pathway have been evaluated in the clinic. Only a limited benefit has been reported in prostate cancer up to now due to the associated side effects, so novel combination approaches and biomarkers predictive of patient response are urgently needed. Here, we reviewed recent data on the crosstalk between AR signaling and the PI3K/AKT/mTOR pathway, the selective inhibitors identified, and the most advanced clinical studies, with a focus on combination treatments. A deeper understanding of the complex molecular mechanisms involved in disease progression and treatment resistance is essential to further guide therapeutic approaches with improved outcomes.
Collapse
Affiliation(s)
- Fabio Raith
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Daniel H. O’Donovan
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Clara Lemos
- Bayer Research and Innovation Center, Bayer US LLC, 238 Main Street, Cambridge, MA 02142, USA
| | - Oliver Politz
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
| | - Bernard Haendler
- Research & Development, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-2215-41198
| |
Collapse
|
11
|
Valeric acid acts as a novel HDAC3 inhibitor against prostate cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:213. [PMID: 36175803 PMCID: PMC9522682 DOI: 10.1007/s12032-022-01814-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer is the second cause of cancer-related deaths in men worldwide, and new agents for curing the disease are still needed. In this study, we theoretically and experimentally demonstrated that valeric acid (VA) was a HDAC inhibitor, and anti-cancer efficacy of VA in prostate cancer cells was also observed using either 2D or 3D culture systems. VA was cytotoxic for prostate cancer cells but low toxic to normal cells. VA significantly inhibited E2F1/E2F3 expression but increased CASP3 activity. In vivo mouse models further showed its anti-cancer activity and potential property of chemosensitizer with promoting apoptosis. The findings suggest that VA acts as a HDAC3 inhibitor with anti-cancer effect on prostate cancer by regulating E2F1/E2F3/CASP3 axis.
Collapse
|
12
|
Song C, Zhang J, Liu X, Li M, Wang D, Kang Z, Yu J, Chen J, Pan H, Wang H, Li G, Huang H. PTEN loss promotes Warburg effect and prostate cancer cell growth by inducing FBP1 degradation. Front Oncol 2022; 12:911466. [PMID: 36237339 PMCID: PMC9552847 DOI: 10.3389/fonc.2022.911466] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Rationale Fructose-1,6-bisphosphatase (FBP1) is a tumor suppressor and a key enzyme negatively regulating Warburg effect in cancer. However, regulation of FBP1 protein expression and its exact role in prostate cancer (PCa) is largely unclear. Phosphatase and tensin homolog (PTEN) is one of the most frequently deleted tumor suppressor genes in human PCa. However, the role of PTEN loss in aberrant Warburg effect in cancer remains poorly understood. Methods Expression of PTEN and FBP1 was analyzed in several PCa cell lines and prostate tumor tissues in mice. Western blot (WB) and RT-PCR approaches were used to examine how PTEN regulates FBP1 expression. Co-immunoprecipitation (co-IP) and in vivo ubiquitination assays were used to define the regulatory mechanisms. A PCa xenograft model was employed to determine the impact of PTEN regulation of FBP1 on PCa growth in vivo. Result We demonstrated that in a manner dependent of PI3K/AKT signal pathway PTEN regulated FBP1 expression in various PCa cell lines and tumors in mice. We confirmed that this regulation took place at the protein level and was mediated by SKP2 E3 ubiquitin ligase. Mechanistically, we showed that serine 271 phosphorylation of FBP1 by cyclin-dependent kinases (CDKs) was essential for SKP2-mediated degradation of FBP1 protein induced by PTEN loss. Most importantly, we further showed that loss of PTEN expression enhanced Warburg effect and PCa growth in mice in a manner dependent, at least partially on FBP1 protein degradation. Conclusions Our results reveal a novel tumor-suppressive feature of PTEN in restraining FBP1 degradation and the Warburg effect. These results also suggest that prohibiting FBP1 protein degradation could be a viable therapeutic strategy for PTEN-deficient PCa.
Collapse
Affiliation(s)
- Changze Song
- Department of Urology, The Fourth Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Jianong Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Xiao Liu
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Meilu Li
- Department of Dermatology, The Second Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Dejie Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Zhijian Kang
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Jiaao Yu
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Jiuwei Chen
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Hongxin Pan
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
- Department of Urological Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Honglei Wang
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Guangbin Li
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, China
- National Health Commission (NHC) Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, United States
- Department of Urology, Mayo Clinic College of Medicine, Rochester, MN, United States
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic College of Medicine, Rochester, MN, United States
- *Correspondence: Haojie Huang,
| |
Collapse
|
13
|
Singh T, Kaur P, Singh P, Singh S, Munshi A. Differential molecular mechanistic behavior of HDACs in cancer progression. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:171. [PMID: 35972597 DOI: 10.1007/s12032-022-01770-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022]
Abstract
Genetic aberration including mutation in oncogenes and tumor suppressor genes transforms normal cells into tumor cells. Epigenetic modifications work concertedly with genetic factors in controlling cancer development. Histone acetyltransferases (HATs), histone deacetylases (HDACs), DNA methyltransferases (DNMTs) and chromatin structure modifier are prospective epigenetic regulators. Specifically, HDACs are histone modifiers regulating the expression of genes implicated in cell survival, growth, apoptosis, and metabolism. The majority of HDACs are highly upregulated in cancer, whereas some have a varied function and expression in cancer progression. Distinct HDACs have a positive and negative role in controlling cancer progression. HDACs are also significantly involved in tumor cells acquiring metastatic and angiogenic potential in order to withstand the anti-tumor microenvironment. HDACs' role in modulating metabolic genes has also been associated with tumor development and survival. This review highlights and discusses the molecular mechanisms of HDACs by which they regulate cell survival, apoptosis, metastasis, invasion, stemness potential, angiogenesis, and epithelial to mesenchymal transitions (EMT) in tumor cells. HDACs are the potential target for anti-cancer drug development and various inhibitors have been developed and FDA approved for a variety of cancers. The primary HDAC inhibitors with proven anti-cancer efficacy have also been highlighted in this review.
Collapse
Affiliation(s)
- Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | - Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India
| | | | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
14
|
Rahbari R, Rahimi K, Rasmi Y, Khadem-Ansari MH, Abdi M. miR-589-5p Inhibits Cell Proliferation by Targeting Histone Deacetylase 3 in Triple Negative Breast Cancer. Arch Med Res 2022; 53:483-491. [PMID: 35840467 DOI: 10.1016/j.arcmed.2022.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 06/05/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Histone deacetylase 3 (HDAC3) is a potential oncogene that is significantly up-regulated in patients with breast cancer. MicroRNAs (miRs) are a group of small non-coding and regulatory RNAs which have recently been proposed as promising molecules for breast cancer target therapy. In the current study, we investigated the impact of miR-589-5p/ HDAC3 axis on cancer cell development in triple negative breast cancer (TNBC) cells. METHODS In-silico analysis determined that miR-589-5p potentially targets HDAC3. We evaluated the HDAC3 and mir-589-5p expression levels in clinical samples and breast cancer cell lines including MDA-MB-231, MDA-MB-468, MCF-7 and MCF-10A. HDAC3 was knocked out to investigate its role on cancer cell progression. Anti-cancerous role of the miR-589-5p was assessed using an expression vector. We evaluated possible alteration in the cell cycle progression, cell viability and cell proliferation, after transient transfection. RESULTS HDAC3 was over-expressed in TNBC clinical samples and breast cancer cell lines compared to non-cancerous controls while miR-589-5p was down regulated in cancer cells. Suppression of HDAC3 decreased the cell viability, cell proliferation and colony formation. Similar results were observed after over-expression of the miR-589-5p. Dual-Luciferase reporter assay confirmed the direct targeting of HDAC3 by miR-589-5p. CONCLUSION Our results showed that miR-589-5p mediates its anti-proliferative effects on breast cancer cells via targeting HDAC3. These findings suggest that the miR-589-5p/ HDAC3 axis could be considered as a possible therapeutic strategy in TNBC.
Collapse
Affiliation(s)
- Rezgar Rahbari
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Karim Rahimi
- Department of Molecular Biology and Genetics, Gene Expression and Gene Medicine, Aarhus University, Aarhus, Denmark; Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Mohammad Abdi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
15
|
HDACs and the epigenetic plasticity of cancer cells: Target the complexity. Pharmacol Ther 2022; 238:108190. [PMID: 35430294 DOI: 10.1016/j.pharmthera.2022.108190] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/07/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022]
Abstract
Cancer cells must adapt to the hostile conditions of the microenvironment in terms of nutrition, space, and immune system attack. Mutations of DNA are the drivers of the tumorigenic process, but mutations must be able to hijack cellular functions to sustain the spread of mutant genomes. Transcriptional control is a key function in this context and is controlled by the rearrangement of the epigenome. Unlike genomic mutations, the epigenome of cancer cells can in principle be reversed. The discovery of the first epigenetic drugs triggered a contaminating enthusiasm. Unfortunately, the complexity of the epigenetic machinery has frustrated this enthusiasm. To develop efficient patient-oriented epigenetic therapies, we need to better understand the nature of this complexity. In this review, we will discuss recent advances in understanding the contribution of HDACs to the maintenance of the transformed state and the rational for their selective targeting.
Collapse
|
16
|
Burleson M, Deng JJ, Qin T, Duong TM, Yan Y, Gu X, Das D, Easley A, Liss MA, Yew PR, Bedolla R, Kumar AP, Huang THM, Zou Y, Chen Y, Chen CL, Huang H, Sun LZ, Boyer TG. GLI3 Is Stabilized by SPOP Mutations and Promotes Castration Resistance via Functional Cooperation with Androgen Receptor in Prostate Cancer. Mol Cancer Res 2022; 20:62-76. [PMID: 34610962 PMCID: PMC9258906 DOI: 10.1158/1541-7786.mcr-21-0108] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 08/24/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022]
Abstract
Although the Sonic hedgehog (SHH) signaling pathway has been implicated in promoting malignant phenotypes of prostate cancer, details on how it is activated and exerts its oncogenic role during prostate cancer development and progression is less clear. Here, we show that GLI3, a key SHH pathway effector, is transcriptionally upregulated during androgen deprivation and posttranslationally stabilized in prostate cancer cells by mutation of speckle-type POZ protein (SPOP). GLI3 is a substrate of SPOP-mediated proteasomal degradation in prostate cancer cells and prostate cancer driver mutations in SPOP abrogate GLI3 degradation. Functionally, GLI3 is necessary and sufficient for the growth and migration of androgen receptor (AR)-positive prostate cancer cells, particularly under androgen-depleted conditions. Importantly, we demonstrate that GLI3 physically interacts and functionally cooperates with AR to enrich an AR-dependent gene expression program leading to castration-resistant growth of xenografted prostate tumors. Finally, we identify an AR/GLI3 coregulated gene signature that is highly correlated with castration-resistant metastatic prostate cancer and predictive of disease recurrence. Together, these findings reveal that hyperactivated GLI3 promotes castration-resistant growth of prostate cancer and provide a rationale for therapeutic targeting of GLI3 in patients with castration-resistant prostate cancer (CRPC). IMPLICATIONS: We describe two clinically relevant mechanisms leading to hyperactivated GLI3 signaling and enhanced AR/GLI3 cross-talk, suggesting that GLI3-specific inhibitors might prove effective to block prostate cancer development or delay CRPC.
Collapse
Affiliation(s)
- Marieke Burleson
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas
| | - Janice J Deng
- Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Tai Qin
- Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Thu Minh Duong
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas
| | - Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Xiang Gu
- Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Debodipta Das
- Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Acarizia Easley
- Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, Texas
| | - Michael A Liss
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | - P Renee Yew
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas
| | - Roble Bedolla
- Department of Urology, UT Health San Antonio, San Antonio, Texas
| | | | - Tim Hui-Ming Huang
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas
| | - Yi Zou
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, UT Health San Antonio, San Antonio, Texas
| | - Chun-Liang Chen
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Lu-Zhe Sun
- Department of Cell Systems & Anatomy, UT Health San Antonio, San Antonio, Texas.
| | - Thomas G Boyer
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, Texas.
| |
Collapse
|
17
|
Dong L, Huang J, Zu P, Liu J, Gao X, Du J, Li Y. Transcription factor 3 (TCF3) combined with histone deacetylase 3 (HDAC3) down-regulates microRNA-101 to promote Burkitt lymphoma cell proliferation and inhibit apoptosis. Bioengineered 2021; 12:7995-8005. [PMID: 34658308 PMCID: PMC8806859 DOI: 10.1080/21655979.2021.1977557] [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] [Indexed: 12/20/2022] Open
Abstract
To explore the function of transcription factor 3 (TCF3) on the proliferation and apoptosis of Burkitt lymphoma cells and its mechanism. qRT-PCR was performed to determine the expression of TCF3, histone deacetylase 3 (HDAC3), and microRNA-101 (miR-101) in the Burkitt lymphoma (BL) tumor tissues and lymph node tissues with reactive lymph node hyperplasia (RLNH). We found that the expression of TCF3 and HDAC3 was up-regulated in BL tumor tissues and lymphoma cells, and the miR-101 expression was down-regulated. And TCF3 and HDAC3 were negatively correlated with the expression of miR-101, respectively. In addition, knockdown of TCF3 can inhibit BL cell proliferation, reduce cell viability and promote cell apoptosis, retain the cell cycle in the G0/G1 phase, and inhibit the expression of Akt/mTOR pathway-related proteins (p-Akt and p-mTOR). When miR-101 was overexpressed, the results were the same as when TCF3 was knocked down. Moreover, we used Co-immunoprecipitation (Co-IP) to detect the interaction between TCF3 and HDAC3, and performed the Chromatin immunoprecipitation (ChIP) experiment to detect the enrichment of TCF3 and HDAC3 in the promoter region of miR-101. We found that TCF3 can interact with HDAC3 and is enriched in the miR-101 promoter region. In conclusion, TCF3 combined with HDAC3 down-regulates the expression of miR-101, thereby promoting the proliferation of BL cells and inhibiting their apoptosis.
Collapse
Affiliation(s)
- Lihua Dong
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingjing Huang
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Peng Zu
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Liu
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xue Gao
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jianwei Du
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yufu Li
- Department of Hematology, Henan Institute of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
18
|
Wawruszak A, Borkiewicz L, Okon E, Kukula-Koch W, Afshan S, Halasa M. Vorinostat (SAHA) and Breast Cancer: An Overview. Cancers (Basel) 2021; 13:4700. [PMID: 34572928 PMCID: PMC8468501 DOI: 10.3390/cancers13184700] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 02/07/2023] Open
Abstract
Vorinostat (SAHA), an inhibitor of class I and II of histone deacetylases, is the first histone deacetylase inhibitor (HDI) approved for the treatment of cutaneous T-cell lymphoma in 2006. HDIs are promising anticancer agents that inhibit the proliferation of many types of cancer cells including breast carcinoma (BC). BC is a heterogeneous disease with variable biological behavior, morphological features, and response to therapy. Although significant progress in the treatment of BC has been made, high toxicity to normal cells, serious side effects, and the occurrence of multi-drug resistance limit the effective therapy of BC patients. Therefore, new active agents which improve the effectiveness of currently used regimens are highly needed. This manuscript analyzes preclinical and clinical trials data of SAHA, applied individually or in combination with other anticancer agents, considering different histological subtypes of BC.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| | - Lidia Borkiewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Syeda Afshan
- Institute of Biomedicine and FICAN West Cancer Centre, University of Turku, 20521 Turku, Finland;
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (L.B.); (E.O.); (M.H.)
| |
Collapse
|
19
|
Nguyen LV, Caldas C. Functional genomics approaches to improve pre-clinical drug screening and biomarker discovery. EMBO Mol Med 2021; 13:e13189. [PMID: 34254730 PMCID: PMC8422077 DOI: 10.15252/emmm.202013189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/23/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Advances in sequencing technology have enabled the genomic and transcriptomic characterization of human malignancies with unprecedented detail. However, this wealth of information has been slow to translate into clinically meaningful outcomes. Different models to study human cancers have been established and extensively characterized. Using these models, functional genomic screens and pre-clinical drug screening platforms have identified genetic dependencies that can be exploited with drug therapy. These genetic dependencies can also be used as biomarkers to predict response to treatment. For many cancers, the identification of such biomarkers remains elusive. In this review, we discuss the development and characterization of models used to study human cancers, RNA interference and CRISPR screens to identify genetic dependencies, large-scale pharmacogenomics studies and drug screening approaches to improve pre-clinical drug screening and biomarker discovery.
Collapse
Affiliation(s)
- Long V Nguyen
- Department of Oncology and Cancer Research UK Cambridge InstituteLi Ka Shing CentreUniversity of CambridgeCambridgeUK
- Cancer Research UK Cambridge Cancer CentreCambridgeUK
| | - Carlos Caldas
- Department of Oncology and Cancer Research UK Cambridge InstituteLi Ka Shing CentreUniversity of CambridgeCambridgeUK
- Cancer Research UK Cambridge Cancer CentreCambridgeUK
| |
Collapse
|
20
|
Jillson LK, Yette GA, Laajala TD, Tilley WD, Costello JC, Cramer SD. Androgen Receptor Signaling in Prostate Cancer Genomic Subtypes. Cancers (Basel) 2021; 13:3272. [PMID: 34208794 PMCID: PMC8269091 DOI: 10.3390/cancers13133272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 12/20/2022] Open
Abstract
While many prostate cancer (PCa) cases remain indolent and treatable, others are aggressive and progress to the metastatic stage where there are limited curative therapies. Androgen receptor (AR) signaling remains an important pathway for proliferative and survival programs in PCa, making disruption of AR signaling a viable therapy option. However, most patients develop resistance to AR-targeted therapies or inherently never respond. The field has turned to PCa genomics to aid in stratifying high risk patients, and to better understand the mechanisms driving aggressive PCa and therapy resistance. While alterations to the AR gene itself occur at later stages, genomic changes at the primary stage can affect the AR axis and impact response to AR-directed therapies. Here, we review common genomic alterations in primary PCa and their influence on AR function and activity. Through a meta-analysis of multiple independent primary PCa databases, we also identified subtypes of significantly co-occurring alterations and examined their combinatorial effects on the AR axis. Further, we discussed the subsequent implications for response to AR-targeted therapies and other treatments. We identified multiple primary PCa genomic subtypes, and given their differing effects on AR activity, patient tumor genetics may be an important stratifying factor for AR therapy resistance.
Collapse
Affiliation(s)
- Lauren K. Jillson
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| | - Gabriel A. Yette
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| | - Teemu D. Laajala
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
- Department of Mathematics and Statistics, University of Turku, 20500 Turku, Finland
| | - Wayne D. Tilley
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia;
- Freemason’s Foundation Centre for Men’s Health, University of Adelaide, Adelaide, SA 5005, Australia
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| | - Scott D. Cramer
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (J.K.L.); (G.A.Y.); (T.D.L.); (J.C.C.)
| |
Collapse
|
21
|
Adhikari N, Jha T, Ghosh B. Dissecting Histone Deacetylase 3 in Multiple Disease Conditions: Selective Inhibition as a Promising Therapeutic Strategy. J Med Chem 2021; 64:8827-8869. [PMID: 34161101 DOI: 10.1021/acs.jmedchem.0c01676] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The acetylation of histone and non-histone proteins has been implicated in several disease states. Modulation of such epigenetic modifications has therefore made histone deacetylases (HDACs) important drug targets. HDAC3, among various class I HDACs, has been signified as a potentially validated target in multiple diseases, namely, cancer, neurodegenerative diseases, diabetes, obesity, cardiovascular disorders, autoimmune diseases, inflammatory diseases, parasitic infections, and HIV. However, only a handful of HDAC3-selective inhibitors have been reported in spite of continuous efforts in design and development of HDAC3-selective inhibitors. In this Perspective, the roles of HDAC3 in various diseases as well as numerous potent and HDAC3-selective inhibitors have been discussed in detail. It will surely open up a new vista in the discovery of newer, more effective, and more selective HDAC3 inhibitors.
Collapse
Affiliation(s)
- Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, P.O. Box 17020, Kolkata, 700032 West Bengal, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, P.O. Box 17020, Kolkata, 700032 West Bengal, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, BITS-Pilani, Hyderabad Campus, Shamirpet, Hyderabad 500078, India
| |
Collapse
|
22
|
Shi J, Cao J, Lu X, Fan L, Guo H, Fu J. Loss of Speckle-Type POZ Protein Promotes Prostate Cancer Cell Migration and Invasion Through Upregulation of MCP-1. Med Sci Monit 2021; 27:e929199. [PMID: 33872295 PMCID: PMC8063634 DOI: 10.12659/msm.929199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background The goal of this study is to verify that the loss of speckle-type POZ protein (SPOP) promotes the migration and invasion of prostate cancer cells, and that this process is brought about by an increase in MCP-1. Material/Methods SPOP knockout C4-2 cells (C4-2 SPOP−/−) were verified by western blotting. Transwell and wound-healing assays were applied to verify different migration and invasion abilities between the C4-2 SPOP−/− and control cells. We used an antibody array to find different soluble chemokine factors in the C4-2 SPOP−/− cells. ELISA and qRT-PCR were applied for confirmation. To test MCP-1 function in conditioned medium, a transwell assay was applied with or without anti-MCP-1 antibody. Results The western blot showed that SPOP was knocked out in sgSPOP-1 and sgSPOP-2 (different clones of C4-2 SPOP−/−). The transwell and wound-healing assays indicated that, compared with control cells, sgSPOP-1 and sgSPOP-2 had stronger migration and invasion abilities. The antibody array found that the expression of MCP-1 was upregulated in sgSPOP-1 and sgSPOP-2 conditioned medium. This result was verified by ELISA and qRT-PCR. In the prostate cancer cells, migration and invasion activity was greatly increased in C4-2 SPOP−/− conditioned medium, while this activity was decreased after anti-MCP-1 antibody neutralization. Conclusions Our findings suggest that the loss of SPOP in C4-2 cells promotes increased cell migration and invasion abilities. This may be realized by upregulating the expression of MCP-1. The inhibition of MCP-1 expression may be an effective treatment for SPOP-mutant prostate cancer.
Collapse
Affiliation(s)
- Junlin Shi
- Key Laboratory of Longevity and Aging-Related Disease of the Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Center for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ji Cao
- Department of Experimental Pathology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Xiaomei Lu
- Key Laboratory of Longevity and Aging-Related Disease of the Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Center for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Langlin Fan
- Key Laboratory of Longevity and Aging-Related Disease of the Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Center for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-Related Disease of the Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Center for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jiejun Fu
- Key Laboratory of Longevity and Aging-Related Disease of the Chinese Ministry of Education, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Center for Translational Medicine and School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|
23
|
Shi L, Yan Y, He Y, Yan B, Pan Y, Orme JJ, Zhang J, Xu W, Pang J, Huang H. Mutated SPOP E3 Ligase Promotes 17βHSD4 Protein Degradation to Drive Androgenesis and Prostate Cancer Progression. Cancer Res 2021; 81:3593-3606. [PMID: 33762355 DOI: 10.1158/0008-5472.can-20-3258] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/01/2021] [Accepted: 03/22/2021] [Indexed: 11/16/2022]
Abstract
Molecular mechanisms underlying intratumoral androgenesis and aberrant androgen receptor (AR) activation in prostate cancer remain poorly understood. Here we demonstrate that ectopic expression of the E3 ubiquitin ligase adaptor speckle-type poxvirus and zinc finger domain protein (SPOP) stabilizes 17βHSD4. SPOP bound a functional substrate-binding consensus (SBC) motif 315RATST319 in 17βHSD4 and promoted nondegradable K27- and K29-linked polyubiquitination of 17βHSD4. The effect of SPOP was antagonized by serum- and glucocorticoid kinase-3 (SGK3)-mediated phosphorylation of serine 318 (S318) in the SBC and S318 phosphorylation-dependent binding of SKP2 E3 ligase and subsequent K48-linked polyubiquitination and proteasomal degradation of 17βHSD4. Prostate cancer-associated SPOP mutations impaired the SPOP-17βHSD4 interaction, caused 17βHSD4 protein destruction in prostate cancer cells in culture and patient specimens, and increased testosterone production and prostate cancer cell growth in vitro and in mouse models. Thus, we have identified SPOP and SKP2 as two essential E3 ubiquitin ligases that exert opposite effects on 17βHSD4 protein degradation and intratumoral androgenesis in prostate cancer cells. We further demonstrate that SPOP mutations or SKP2 overexpression contribute to prostate cancer progression by decreasing 17βHSD4 expression and increasing intratumoral androgen synthesis. SIGNIFICANCE: This study reveals a novel mechanism of aberrant AR activation in SPOP-mutated prostate cancer and uncovers putative biomarkers for effective treatment by AR-targeted therapies.
Collapse
Affiliation(s)
- Lei Shi
- Department of Radiation Oncology, the Fourth Hospital of Harbin Medical University, Harbin, China.,Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Yundong He
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Binyuan Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota.,Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jacob J Orme
- Division of Medical Oncology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Scottsdale, Arizona
| | - Wanhai Xu
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jun Pang
- Department of Urology, Kidney and Urology Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota. .,Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota.,Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| |
Collapse
|
24
|
Su W, Zeng L, Chen W. Moscatilin Suppresses the Breast Cancer Both In Vitro and In Vivo by Inhibiting HDAC3. Dose Response 2021; 19:15593258211001251. [PMID: 33795998 PMCID: PMC7968028 DOI: 10.1177/15593258211001251] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Moscatilin, a natural compound isolated from the orchid Dendrobium moscatum, has multiple pharmacological actions. The present study investigated the anti-tumor role of moscatilin in breast cancer and elucidated the underlying mechanisms. Cell proliferation, viability, and apoptosis of moscatilin treated MDA-MB-231 cells were determined by CCK-8 assay and flow cytometry. Histone deacetylases (HDACs) expression levels and global acetylated status of breast cancer cells were detected by Western blot and qPCR. Mouse xenograft model was established to evaluate the anti-cancer effects of moscatilin. Moscatilin treatment dose dependently suppressed proliferation and increased apoptosis of breast cancer cells. Moreover, moscatilin administration dramatically repressed tumor growth and extended survival time of mouse model. Mechanistically, moscatilin down-regulated HDAC3 expression, and then enhanced the global acetylated status of histone H3 (H3K9Ac) and H4 (H4K16Ac). Our findings indicate that moscatilin can inhibit the proliferation and promote apoptosis of breast cancer in vitro and in vivo, which suggests that moscatilin can be used as a potential therapeutic agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Wenjie Su
- Department of Tumor Surgery, Quanzhou Guangqian Hospital, Meishan Town, Nan'an City, Quanzhou, Fujian, China
| | - Lianfu Zeng
- Department of Tumor Surgery, Quanzhou Guangqian Hospital, Meishan Town, Nan'an City, Quanzhou, Fujian, China
| | - Weida Chen
- Department of Tumor Surgery, Quanzhou Guangqian Hospital, Meishan Town, Nan'an City, Quanzhou, Fujian, China
| |
Collapse
|
25
|
Carneiro BA, Lotan TL, de Souza A, Aggarwal R. Emerging Subtypes and New Treatments for Castration-Resistant Prostate Cancer. Am Soc Clin Oncol Educ Book 2021; 40:e319-e332. [PMID: 32479115 DOI: 10.1200/edbk_100025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Genomic characterization of metastatic castration-resistant prostate cancer (mCRPC) has been remodeling the treatment landscape of this disease in the past decade. The emergence of molecularly defined subsets of mCRPC is altering the treatment paradigm from therapeutics with nonspecific activity across the spectrum, including androgen receptor (AR)-directed treatments, docetaxel, and cabazitaxel, to targeted approaches directed at molecular subsets of disease. The meaningful benefit of PARP inhibitors in mCRPC carrying mutations in DNA repair genes demonstrated in a phase III trial epitomizes this transition in the treatment paradigm of mCRPC and brings new challenges related to how to sequence and integrate the targeted therapies on top of the treatments with broad activity in all mCRPC. To enable and sustain the advance of precision oncology in the management of mCRPC, genomic characterization is required, including somatic and germline testing, for all patients with the ultimate goal of longitudinal molecular profiling guiding treatment decisions and sequential treatments of this lethal disease. This article reviews the emerging molecular subtypes of mCRPC that are driving the evolution of mCRPC treatment.
Collapse
Affiliation(s)
- Benedito A Carneiro
- Warren Alpert Medical School, Brown University, Providence, RI.,Lifespan Cancer Institute, Providence, RI
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD
| | - Andre de Souza
- Warren Alpert Medical School, Brown University, Providence, RI.,Lifespan Cancer Institute, Providence, RI
| | | |
Collapse
|
26
|
Maitland NJ. Resistance to Antiandrogens in Prostate Cancer: Is It Inevitable, Intrinsic or Induced? Cancers (Basel) 2021; 13:327. [PMID: 33477370 PMCID: PMC7829888 DOI: 10.3390/cancers13020327] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Increasingly sophisticated therapies for chemical castration dominate first-line treatments for locally advanced prostate cancer. However, androgen deprivation therapy (ADT) offers little prospect of a cure, as resistant tumors emerge rather rapidly, normally within 30 months. Cells have multiple mechanisms of resistance to even the most sophisticated drug regimes, and both tumor cell heterogeneity in prostate cancer and the multiple salvage pathways result in castration-resistant disease related genetically to the original hormone-naive cancer. The timing and mechanisms of cell death after ADT for prostate cancer are not well understood, and off-target effects after long-term ADT due to functional extra-prostatic expression of the androgen receptor protein are now increasingly being recorded. Our knowledge of how these widely used treatments fail at a biological level in patients is deficient. In this review, I will discuss whether there are pre-existing drug-resistant cells in a tumor mass, or whether resistance is induced/selected by the ADT. Equally, what is the cell of origin of this resistance, and does it differ from the treatment-naïve tumor cells by differentiation or dedifferentiation? Conflicting evidence also emerges from studies in the range of biological systems and species employed to answer this key question. It is only by improving our understanding of this aspect of treatment and not simply devising another new means of androgen inhibition that we can improve patient outcomes.
Collapse
Affiliation(s)
- Norman J Maitland
- Department of Biology, University of York, Heslington, York YO10 5DD, UK
| |
Collapse
|
27
|
Kase AM, Copland III JA, Tan W. Novel Therapeutic Strategies for CDK4/6 Inhibitors in Metastatic Castrate-Resistant Prostate Cancer. Onco Targets Ther 2020; 13:10499-10513. [PMID: 33116629 PMCID: PMC7576355 DOI: 10.2147/ott.s266085] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022] Open
Abstract
The majority of patients with castrate-resistant prostate cancer will have metastatic disease at the time of diagnosis. Investigative efforts on new therapeutics for this patient population have improved with the development of androgen signaling inhibitors, such as abiraterone and enzalutamide, and PARP inhibitors, such as rucaparib and olaparib, to accompany the previously FDA-approved docetaxel, cabazitaxel, sipuleucel-T, and Radium 223. However, new therapeutic strategies are necessary to prolong survival as progression after these agents is inevitable. CDK4/6 inhibitors have advanced the field of estrogen receptor positive breast cancer treatment and are being investigated in prostate cancer given the role of androgen receptor signaling effects on the cell cycle. Response to CDK4/6 inhibitors may be predicted by the tumors' genomic profile and may provide insight into combinatory therapy with CDK4/6 inhibitors in order to delay resistance or provide synergistic effects. Here, we review the use of CDK4/6 inhibitors in prostate cancer and potential combinations based on known resistance mechanisms to CDK4/6 inhibitors, prostate cancer regulatory pathways, and prostate-cancer-specific genomic alterations.
Collapse
Affiliation(s)
- Adam M Kase
- Mayo Clinic Florida Division of Hematology Oncology, Jacksonville, FL32224, USA
| | - John A Copland III
- Mayo Clinic Florida Department of Cancer Biology, Jacksonville, FL32224, USA
| | - Winston Tan
- Mayo Clinic Florida Division of Hematology Oncology, Jacksonville, FL32224, USA
| |
Collapse
|
28
|
Li G, Tian Y, Zhu WG. The Roles of Histone Deacetylases and Their Inhibitors in Cancer Therapy. Front Cell Dev Biol 2020; 8:576946. [PMID: 33117804 PMCID: PMC7552186 DOI: 10.3389/fcell.2020.576946] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
Genetic mutations and abnormal gene regulation are key mechanisms underlying tumorigenesis. Nucleosomes, which consist of DNA wrapped around histone cores, represent the basic units of chromatin. The fifth amino group (Nε) of histone lysine residues is a common site for post-translational modifications (PTMs), and of these, acetylation is the second most common. Histone acetylation is modulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), and is involved in the regulation of gene expression. Over the past two decades, numerous studies characterizing HDACs and HDAC inhibitors (HDACi) have provided novel and exciting insights concerning their underlying biological mechanisms and potential anti-cancer treatments. In this review, we detail the diverse structures of HDACs and their underlying biological functions, including transcriptional regulation, metabolism, angiogenesis, DNA damage response, cell cycle, apoptosis, protein degradation, immunity and other several physiological processes. We also highlight potential avenues to use HDACi as novel, precision cancer treatments.
Collapse
Affiliation(s)
- Guo Li
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Yuan Tian
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Wei-Guo Zhu
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shenzhen University Health Science Center, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
29
|
Aquila S, Santoro M, Caputo A, Panno ML, Pezzi V, De Amicis F. The Tumor Suppressor PTEN as Molecular Switch Node Regulating Cell Metabolism and Autophagy: Implications in Immune System and Tumor Microenvironment. Cells 2020; 9:cells9071725. [PMID: 32708484 PMCID: PMC7408239 DOI: 10.3390/cells9071725] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Recent studies conducted over the past 10 years evidence the intriguing role of the tumor suppressor gene Phosphatase and Tensin Homolog deleted on Chromosome 10 PTEN in the regulation of cellular energy expenditure, together with its capability to modulate proliferation and survival, thus expanding our knowledge of its physiological functions. Transgenic PTEN mice models are resistant to oncogenic transformation, present decreased adiposity and reduced cellular glucose and glutamine uptake, together with increased mitochondrial oxidative phosphorylation. These acquisitions led to a novel understanding regarding the role of PTEN to counteract cancer cell metabolic reprogramming. Particularly, PTEN drives an “anti-Warburg state” in which less glucose is taken up, but it is more efficiently directed to the mitochondrial Krebs cycle. The maintenance of cellular homeostasis together with reduction of metabolic stress are controlled by specific pathways among which autophagy, a catabolic process strictly governed by mTOR and PTEN. Besides, a role of PTEN in metabolic reprogramming and tumor/stroma interactions in cancer models, has recently been established. The genetic inactivation of PTEN in stromal fibroblasts of mouse mammary glands, accelerates breast cancer initiation and progression. This review will discuss our novel understanding in the molecular connection between cell metabolism and autophagy by PTEN, highlighting novel implications regarding tumor/stroma/immune system interplay. The newly discovered action of PTEN opens innovative avenues for investigations relevant to counteract cancer development and progression.
Collapse
Affiliation(s)
- Saveria Aquila
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Marta Santoro
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Annalisa Caputo
- Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences; University of Calabria, 87036 Rende, Italy; (S.A.); (M.S.); (M.L.P.); (V.P.)
- Health Center, University of Calabria, 87036 Rende, Italy
- Correspondence:
| |
Collapse
|
30
|
Yu X, Yang F, Jiang H, Fan L. RGFP966 Suppresses Tumor Growth and Migration Through Inhibition of EGFR Expression in Hepatocellular Carcinoma Cells in vitro. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:121-128. [PMID: 32021097 PMCID: PMC6959505 DOI: 10.2147/dddt.s234871] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/21/2019] [Indexed: 12/14/2022]
Abstract
Purpose Histone deacetylase 3 (HDAC3) has been suggested to play a role in hepatocellular carcinoma (HCC). In the present report, we aimed to identify the effects of RGFP966, a specific HDAC3 inhibitor, on the cell proliferation and migration of HCC cell lines. Methods Human HCC cell lines, which were identified using short tandem repeat (STR) DNA profiling analysis, were used in this report. Cell proliferation assay was used to identify the growth viability of cells. Wound healing and transwell assay were used to identify the migration ability of cells. Further, a human phospho-receptor tyrosine kinases array kit was used to screen out RGFP966 effects on key receptor tyrosine kinases. Then, the mRNA expression was quantified by real-time PCR, and protein expression was identified by Western blot immunoassay. Results We found that RGFP966 inhibited both proliferation and migration of HCC cells. Further, RGFP966 represses the expression and phosphorylation levels of epidermal growth factor receptor (EGFR) in HCC cells. Moreover, HDAC3 is involved in the inhibition of EGFR by RGFP966. Overall, we elucidated an inhibitive function of RGFP966 in HCC progression. Conclusion RGFP966 inhibits EGFR signaling pathway and suppresses proliferation and migration of HCC cells.
Collapse
Affiliation(s)
- Xinying Yu
- Second Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Fan Yang
- Third Neonatal Ward, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Hong Jiang
- Second Neonatal Ward, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Ling Fan
- Second Pediatric Intensive Care Unit, Shengjing Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| |
Collapse
|
31
|
Wen S, Niu Y, Huang H. Posttranslational regulation of androgen dependent and independent androgen receptor activities in prostate cancer. Asian J Urol 2019; 7:203-218. [PMID: 33024699 PMCID: PMC7525085 DOI: 10.1016/j.ajur.2019.11.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/21/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer among men in western countries. Androgen receptor (AR) signaling plays key roles in the development of PCa. Androgen deprivation therapy (ADT) remains the standard therapy for advanced PCa. In addition to its ligand androgen, accumulating evidence indicates that posttranscriptional modification is another important mechanism to regulate AR activities during the progression of PCa, especially in castration resistant prostate cancer (CRPC). To date, a number of posttranscriptional modifications of AR have been identified, including phosphorylation (e.g. by CDK1), acetylation (e.g. by p300 and recognized by BRD4), methylation (e.g. by EZH2), ubiquitination (e.g. by SPOP), and SUMOylation (e.g. by PIAS1). These modifications are essential for the maintenance of protein stability, nuclear localization and transcriptional activity of AR. This review summarizes posttranslational modifications that influence androgen-dependent and -independent activities of AR, PCa progression and therapy resistance. We further emphasize that in addition to androgen, posttranslational modification is another important way to regulate AR activity, suggesting that targeting AR posttranslational modifications, such as proteolysis targeting chimeras (PROTACs) of AR, represents a potential and promising alternate for effective treatment of CRPC. Potential areas to be investigated in the future in the field of AR posttranslational modifications are also discussed.
Collapse
Affiliation(s)
- Simeng Wen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China.,Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, USA
| | - Yuanjie Niu
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, USA.,Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, USA.,Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, USA
| |
Collapse
|
32
|
Fan L, Ye H, Wan Y, Qin L, Zhu L, Su J, Zhu X, Zhang L, Miao Q, Zhang Q, Zhang Z, Xu A, Li Y, Li X, Wang Y. Adaptor protein APPL1 coordinates HDAC3 to modulate brown adipose tissue thermogenesis in mice. Metabolism 2019; 100:153955. [PMID: 31390528 DOI: 10.1016/j.metabol.2019.153955] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/12/2019] [Accepted: 07/31/2019] [Indexed: 01/17/2023]
Abstract
OBJECTIVES The activation of brown adipose tissue (BAT) is considered as a promising therapeutic target for obesity. APPL1 (Adaptor protein containing the Pleckstrin homology domain, Phosphotyrosine binding domain and Leucine zipper motif) is an intracellular adaptor protein and its genetic variation is correlated with BMI and body fat distribution in diabetic patients. However, little is known about the roles of APPL1 in BAT thermogenesis. MATERIALS/METHODS In this study, adipose tissue specific knockout (ASKO) mice were generated to evaluate APPL1's role in BAT thermogenesis in vivo, and possible signaling pathways were further explored in cultured brown adipocytes. RESULTS After high fat diet challenge, APPL1 ASKO mice developed more severe obesity, glucose intolerance and insulin resistance compared with control mice. Metabolic cage study showed that APPL1 deficiency impaired energy expenditure and adaptive thermogenesis in ASKO mice. PET-CT analysis showed decreased standardized uptake value (SUV) in the inter-scapular region which indicated impaired BAT activity in ASKO mice. Further study showed deletion of APPL1 attenuated brown fat specific gene expression, such as UCP1 and PGC1α in both BAT and brown adipocytes. In cultured brown adipocytes, upon cAMP stimulation, APPL1 shuttled from cytosol to nuclei. Co-IP and ChIP study showed that APPL1 could directly interact with histone deacetylase 3 (HDAC3) to mediate chromatin remodeling and UCP1 gene expression. CONCLUSIONS Our data demonstrated the essential role of APPL1 in regulating brown adipocytes thermogenesis via interaction with HDAC3, which may have potential therapeutic implications for treatment of obesity.
Collapse
Affiliation(s)
- Linling Fan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lang Qin
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lu Zhu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Su
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoming Zhu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Lv Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Miao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiongyue Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Aimin Xu
- Department of Medicine, the University of Hong Kong, Hong Kong
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, China.
| | - Yi Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
33
|
Lou T, Zhuang H, Liu C, Zhang Z. HDAC3 positively regulates HE4 expression to promote ovarian carcinoma progression. Arch Biochem Biophys 2019; 675:108044. [DOI: 10.1016/j.abb.2019.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/12/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022]
|
34
|
Yan Y, Ma J, Wang D, Lin D, Pang X, Wang S, Zhao Y, Shi L, Xue H, Pan Y, Zhang J, Wahlestedt C, Giles FJ, Chen Y, Gleave ME, Collins CC, Ye D, Wang Y, Huang H. The novel BET-CBP/p300 dual inhibitor NEO2734 is active in SPOP mutant and wild-type prostate cancer. EMBO Mol Med 2019; 11:e10659. [PMID: 31559706 PMCID: PMC6835201 DOI: 10.15252/emmm.201910659] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 01/26/2023] Open
Abstract
CULLIN3‐based E3 ubiquitin ligase substrate‐binding adaptor gene SPOP is frequently mutated in prostate cancer (PCa). PCa harboring SPOP hotspot mutants (e.g., F133V) are resistant to BET inhibitors because of aberrant elevation of BET proteins. Here, we identified a previously unrecognized mutation Q165P at the edge of SPOP MATH domain in primary and metastatic PCa of a patient. The Q165P mutation causes structural changes in the MATH domain and impairs SPOP dimerization and substrate degradation. Different from F133V hotspot mutant tumors, Q165P mutant patient‐derived xenografts (PDXs) and organoids were modestly sensitive to the BET inhibitor JQ1. Accordingly, protein levels of AR, BRD4 and downstream effectors such as RAC1 and phosphorylated AKT were not robustly elevated in Q165P mutant cells as in F133V mutant cells. However, NEO2734, a novel dual inhibitor of BET and CBP/p300, is active in both hotspot mutant (F133V) and non‐hotspot mutant (Q165P) PCa cells in vitro and in vivo. These data provide a strong rationale to clinically investigate the anti‐cancer efficacy of NEO2734 in SPOP‐mutated PCa patients.
Collapse
Affiliation(s)
- Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jian Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dejie Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Dong Lin
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Xiaodong Pang
- Department of Physics, State Key Laboratory of Surface Physics, Fudan University, Shanghai, China
| | - Shangqian Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu Zhao
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jun Zhang
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Phoenix, AZ, USA
| | - Claes Wahlestedt
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin E Gleave
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Collin C Collins
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Centre, Vancouver, BC, Canada.,The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
35
|
Ma L, Yan Y, Bai Y, Yang Y, Pan Y, Gang X, Karnes RJ, Zhang J, Lv Q, Wu Q, Huang H. Overcoming EZH2 Inhibitor Resistance by Taxane in PTEN-Mutated Cancer. Am J Cancer Res 2019; 9:5020-5034. [PMID: 31410199 PMCID: PMC6691386 DOI: 10.7150/thno.34700] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/07/2019] [Indexed: 12/24/2022] Open
Abstract
Rationale: The Polycomb group (PcG) protein EZH2 is implicated in cancer progression due to its frequent overexpression in many cancer types and therefore is a promising therapeutic target. Forkhead box transcription factor-1 (FOXO1) is a tumor suppressor that is often transcriptionally downregulated in human cancers such as prostate cancer although the underlying regulatory mechanisms remain elusive. Methods: Analysis of EZH2 ChIP-seq and ChIP-on-chip data in various cell types was performed. ChIP-qPCR, RT-qPCR, and western blot analyses were conducted to determine the mechanism by which EZH2 represses FOXO1 expression. Immunohistochemistry was employed to assess the correlation between EZH2 and FOXO1 protein expression in prostate cancer patient specimens. In vitro MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) and animal experiments were performed to determine the anti-cancer efficacy of EZH2 inhibitor alone or in combination of docetaxel, a chemotherapy agent of the taxane family, and dependency of the efficacy on FOXO1 expression. Results: We demonstrated that EZH2 binds to the FOXO1 gene promoter. EZH2 represses FOXO1 gene expression at the transcriptional level. EZH2 protein level inversely correlated with FOXO1 protein expression in prostate cancer patient specimens. This repression requires the methyltransferase activity and the functional PRC2 complex. While effectively inducing loss of viability of PTEN-positive 22Rv1 prostate cancer cells, EZH2 inhibitor failed to inhibit growth of PTEN-negative C4-2 prostate cancer cells. Co-treatment with docetaxel overcame EZH2 inhibitor resistance in PTEN-negative cancer cells in vitro and in mice. This effect was largely mediated by docetaxel-induced nuclear localization and activation of FOXO1. Conclusions: This study identifies FOXO1 as a bona fide repression target of EZH2 and an essential mediator of EZH2 inhibition-induced cell death. Our findings suggest that EZH2 repression of FOXO1 can be targeted by EZH2 inhibitor as a monotherapy for PTEN-proficient cancers or in combination with taxane for treatment of cancers with PTEN mutation or deletion.
Collapse
|
36
|
Bai Y, Yang Y, Yan Y, Zhong J, Blee AM, Pan Y, Ma T, Karnes RJ, Jimenez R, Xu W, Huang H. RUNX2 overexpression and PTEN haploinsufficiency cooperate to promote CXCR7 expression and cellular trafficking, AKT hyperactivation and prostate tumorigenesis. Theranostics 2019; 9:3459-3475. [PMID: 31281490 PMCID: PMC6587168 DOI: 10.7150/thno.33292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Rationale: The overall success rate of prostate cancer (PCa) diagnosis and therapy has been improved over the years. However, genomic and phenotypic heterogeneity remains a major challenge for effective detection and treatment of PCa. Efforts to better classify PCa into functional subtypes and elucidate the molecular mechanisms underlying prostate tumorigenesis and therapy resistance are warranted for further improvement of PCa outcomes. Methods: We generated Cre+;Runx2-cTg;Ptenp/+ (Runx2-Pten double mutant) mice by crossbreeding Cre+;Runx2-cTg males with Pten conditional (Ptenp/p) females. By using Hematoxylin and Eosin (H&E) staining, SMA and Masson's Trichrome staining, we investigated the effect of PTEN haploinsufficiency in combination with Runx2 overexpression on prostate tumorigenesis. Moreover, we employed immunohistochemistry (IHC) to stain Ki67 for cell proliferation, cleaved caspase 3 for apoptosis and AKT phosphorylation for signaling pathway in prostate tissues. Chromatin immunoprecipitation coupled quantitative PCR (ChIP-qPCR), reverse transcription coupled quantitative PCR (RT-qPCR), western blot (WB) analyses and immunofluorescence (IF) were conducted to determine the underlying mechanism by which RUNX2 regulates CXCR7 and AKT phosphorylation in PCa cells. Results: We demonstrated that mice with prostate-specific Pten heterozygous deletion and Runx2 overexpression developed high-grade prostatic intraepithelial neoplasia (HGPIN) and cancerous lesions at age younger than one year, with concomitant high level expression of Akt phosphorylation and the chemokine receptor Cxcr7 in malignant glands. RUNX2 overexpression induced CXCR7 transcription and membrane location and AKT phosphorylation in PTEN-deficient human PCa cell lines. Increased expression of RUNX2 also promoted growth of PCa cells and this effect was largely mediated by CXCR7. CXCR7 expression also positively correlated with AKT phosphorylation in PCa patient specimens. Conclusions: Our results reveal a previously unidentified cooperative role of RUNX2 overexpression and PTEN haploinsufficiency in prostate tumorigenesis, suggesting that the defined RUNX2-CXCR7-AKT axis can be a viable target for effective treatment of PCa.
Collapse
Affiliation(s)
- Yang Bai
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yinhui Yang
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Jian Zhong
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Alexandra M. Blee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Tao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - R. Jeffrey Karnes
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Rafael Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Wanhai Xu
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
37
|
Zhao J, Meng Z, Xie C, Yang C, Liu Z, Wu S, Wang B, Fan P, Jin X, Wu H. B7-H3 is regulated by BRD4 and promotes TLR4 expression in pancreatic ductal adenocarcinoma. Int J Biochem Cell Biol 2019; 108:84-91. [PMID: 30664982 DOI: 10.1016/j.biocel.2019.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 12/24/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies worldwide. PDAC is resistant to chemotherapy and radiotherapy which leads to the poor prognosis of PDAC patients and a 5-year survival rate of less than 5%. Exploring the mechanism of the pancreatic cancer tumorigenesis is the key to finding a novel therapeutic strategy for cancer treatment. B7-H3 belongs to the B7 family of immunoregulatory proteins, and the overexpression of B7-H3 is found in various types of cancer. The regulation of B7-H3 expression in pancreatic cancer is still unclear. Here, we showed that B7-H3 acted as a negative prognostic biomarker in PDAC and promoted cell proliferation, invasion and metastasis in pancreatic cancer. Next, we applied the drug screening method to identify bromodomain and extra-terminal motif (BET) inhibitors that decreased the protein and mRNA levels of B7-H3 in pancreatic cancer cells. Moreover, we verified that BRD4 was responsible for regulating the expression of B7-H3 at the transcriptional level. Finally, our data indicated that the BRD4/B7-H3 axis modulated the expression of TLR4 in pancreatic cancer cells. Taken together, our results elucidated the regulation of B7-H3 expression in pancreatic cancer and uncovered the importance of BRD4/B7-H3/TLR4 pathway. The targeting of B7-H3 by the BET inhibitors may be a novel therapeutic strategy to overcome the immunotherapy and chemotherapy resistance in pancreatic cancer.
Collapse
Affiliation(s)
- Jingyuan Zhao
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zibo Meng
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chao Xie
- Department of Hepatobiliary pancreatic surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chong Yang
- Organ Transplantation Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - Zhiqiang Liu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shihong Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Fan
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Jin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
38
|
Yan Y, Huang H. Interplay Among PI3K/AKT, PTEN/FOXO and AR Signaling in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1210:319-331. [DOI: 10.1007/978-3-030-32656-2_14] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
39
|
Nutsford AN, Galvin HD, Ahmed F, Husain M. The Class IV human deacetylase, HDAC11, exhibits anti-influenza A virus properties via its involvement in host innate antiviral response. Cell Microbiol 2018; 21:e12989. [PMID: 30511515 DOI: 10.1111/cmi.12989] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/06/2018] [Accepted: 11/22/2018] [Indexed: 01/01/2023]
Abstract
Histone deacetylase 11 (HDAC11) is most recently discovered deacetylase. Here, we demonstrate that human HDAC11 exhibits anti-influenza A virus (IAV) properties. We found that knockdown of HDAC11 expression augments IAV growth kinetics in human lung epithelial cells A549 by up to 1 log. One of the ways HDAC11 exerts its anti-IAV function is by being a part of IAV-induced host antiviral response. We found that the kinetics of both IAV- and interferon-induced innate antiviral response is significantly delayed in HDAC11-depleted cells. Further, in the absence of HDAC11 expression, there was a significant decrease in the expression of interferon-stimulated genes-IFITM3, ISG15, and viperin-previously implicated in anti-IAV function. One of the ways IAV antagonises HDAC11 is by downregulating its expression in host cells. We found that there was up to 93% reduction in HDAC11 transcript levels in A549 cells in response to IAV infection. HDAC11 is the smallest HDAC with majority of its polypeptide assigned to catalytic domain. Evolutionarily, it seems to be the least evolved and most closely related to common ancestral HDAC gene(s). Furthermore, HDAC11 has also been described as a deacylase. Therefore, our findings present exciting prospects for further investigations into significance of HDAC11 in virus infections.
Collapse
Affiliation(s)
- Ashley N Nutsford
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Henry D Galvin
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Farjana Ahmed
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Matloob Husain
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
40
|
Wang B, Fan P, Zhao J, Wu H, Jin X, Wu H. FBP1 loss contributes to BET inhibitors resistance by undermining c-Myc expression in pancreatic ductal adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:224. [PMID: 30201002 PMCID: PMC6131902 DOI: 10.1186/s13046-018-0888-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/21/2018] [Indexed: 01/05/2023]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal tumor types worldwide. BET inhibitors display anti-tumor activity in pancreatic cancer, however the cells often develop resistance after a long-term treatment and the underlying molecular basis is not fully understood. Methods Drug screening assay in Fructose-1, 6-biphosphatase (FBP1) knockdown or overexpressing pancreatic cancer cells was performed. Tumor cell motility, FBP1 protein and mRNA changes were investigated after BET inhibitors treatment. The interaction between TRIM28 and FBP1 after BET inhibitors treatment was examined by Co-immunoprecipitation (IP) and GST pull-down. The relationship between FBP1 and c-Myc was examined by western blot, RT-qPCR and immunohistochemistry (IHC). Results The expression of FBP1 protein increased the sensitivity of pancreatic cancer cells to JQ1. Furthermore, we showed that JQ1 stabilized FBP1 protein level by disrupting the interaction between FBP1 and TRIM28 in pancreatic cancer cells. Moreover, we demonstrated that FBP1 promoted c-Myc degradation through disrupting the ERK-c-Myc axis. Conclusions FBP1 modulates the sensitivity of pancreatic cancer cells to BET inhibitors by decreasing the expression of c-Myc. These findings highlight FBP1 could be used as a therapeutic niche for patient-tailored therapies. Electronic supplementary material The online version of this article (10.1186/s13046-018-0888-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Fan
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingyuan Zhao
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Heyu Wu
- Operating Room, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xin Jin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
41
|
Wei X, Fried J, Li Y, Hu L, Gao M, Zhang S, Xu B. Functional roles of Speckle-Type Poz (SPOP) Protein in Genomic stability. J Cancer 2018; 9:3257-3262. [PMID: 30271484 PMCID: PMC6160670 DOI: 10.7150/jca.25930] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/27/2018] [Indexed: 12/27/2022] Open
Abstract
Understanding the functional significance of the essential elements in maintaining genomic stability provides insights into the process of tumor initiation and progression, and predicts therapeutic responses. One such element that has recently attracted significant attention is the Speckle-Type Poz Protein (SPOP), an E3 ubiquitin ligase adaptor protein. SPOP is frequently mutated or has altered expression in various cancers, including prostate, renal and endometrial. SPOP is involved in the regulation of proteasome-mediated degradation of several oncoproteins. Moreover, recent data also indicate SPOP's direct involvement in the DNA damage response. SPOP mutants induce alternations in the DNA damage repair pathway by promoting the error-prone Non-homologous end joining (NHEJ) pathway. SPOP has been linked with significant functions in cellular signaling pathways and cancer suppression. This mini-review will discuss recent findings regarding SPOP's role in genomic stability in the pathological setting.
Collapse
Affiliation(s)
- Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Joshua Fried
- Department of Oncology, Southern Research Institute and Cancer Cell Biology Program, University of Alabama at Birmingham Graduate School, Birmingham, AL, 35205.,Cancer Cell Biology Program, University of Alabama at Birmingham Comprehensive Cancer Center Birmingham, AL 35205, USA
| | - Ying Li
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Linfei Hu
- Department of Thyroid and Cervical Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ming Gao
- Department of Thyroid and Cervical Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Sheng Zhang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Bo Xu
- Department of Oncology, Southern Research Institute and Cancer Cell Biology Program, University of Alabama at Birmingham Graduate School, Birmingham, AL, 35205.,Cancer Cell Biology Program, University of Alabama at Birmingham Comprehensive Cancer Center Birmingham, AL 35205, USA.,Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
42
|
HDAC Inhibition Counteracts Metastatic Re-Activation of Prostate Cancer Cells Induced by Chronic mTOR Suppression. Cells 2018; 7:cells7090129. [PMID: 30200497 PMCID: PMC6162415 DOI: 10.3390/cells7090129] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
This study was designed to investigate whether epigenetic modulation by histone deacetylase (HDAC) inhibition might circumvent resistance towards the mechanistic target of rapamycin (mTOR) inhibitor temsirolimus in a prostate cancer cell model. Parental (par) and temsirolimus-resistant (res) PC3 prostate cancer cells were exposed to the HDAC inhibitor valproic acid (VPA), and tumor cell adhesion, chemotaxis, migration, and invasion were evaluated. Temsirolimus resistance was characterized by reduced binding of PC3res cells to endothelium, immobilized collagen, and fibronectin, but increased adhesion to laminin, as compared to the parental cells. Chemotaxis, migration, and invasion of PC3res cells were enhanced following temsirolimus re-treatment. Integrin α and β receptors were significantly altered in PC3res compared to PC3par cells. VPA significantly counteracted temsirolimus resistance by down-regulating tumor cell–matrix interaction, chemotaxis, and migration. Evaluation of integrin expression in the presence of VPA revealed a significant down-regulation of integrin α5 in PC3res cells. Blocking studies demonstrated a close association between α5 expression on PC3res and chemotaxis. In this in vitro model, temsirolimus resistance drove prostate cancer cells to become highly motile, while HDAC inhibition reversed the metastatic activity. The VPA-induced inhibition of metastatic activity was accompanied by a lowered integrin α5 surface level on the tumor cells.
Collapse
|
43
|
Abstract
Co‐targeting strategies strive to improve cancer outcomes by combining therapies under contextualized genetic and environmental conditions that selectively target exploitable alterations in tumor cells. Adaptive survival pathways triggered by inhibition of driver genes in the androgen receptor (AR) or PI3K/AKT pathways are of great interest, since they are among the most frequently altered in castrate‐resistant prostate cancer (CRPC). Unfortunately, negative feedback loops exist between the AR and PI3K/AKT pathways such that targeting AR leads to activation of PI3K/AKT signaling, while PI3K/AKT pathway inhibition leads to increased AR transcriptional activity. Hence, targeting both pathways provides an opportunity for conditional lethality and a high therapeutic index. In this issue of EMBO Molecular Medicine , Yan et al (2018 ) present an elegant study showing that histone deacetylase 3 (HDAC3) acts as a common upstream activator of both AR and AKT signaling pathways, and use HDAC3 inhibitors as a monotherapy to co‐target two major pathways driving CRPC growth.
Collapse
Affiliation(s)
- Amina Zoubeidi
- The Vancouver Prostate CentreDepartment of Urologic SciencesUniversity of British ColumbiaVancouverBCCanada
| | - Martin E Gleave
- The Vancouver Prostate CentreDepartment of Urologic SciencesUniversity of British ColumbiaVancouverBCCanada
| |
Collapse
|