1
|
Hu Z, Tang M, Huang Y, Cai B, Sun X, Chen G, Huang A, Li X, Shah AR, Jiang L, Li Q, Xu X, Lu W, Mao Z, Wan X. SIRT7 facilitates endometrial cancer progression by regulating PTEN stability in an estrogen-dependent manner. Nat Commun 2025; 16:2989. [PMID: 40148340 PMCID: PMC11950185 DOI: 10.1038/s41467-025-58317-0] [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: 09/26/2023] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
The prognosis of metastatic endometrial carcinoma (EC), one of the most common gynecological malignancies worldwide, remains poor, and the underlying driver of metastases is poorly understood. Dysregulation in estrogen-related signaling and inactivation of tumor suppressor PTEN are two essential risk factors of EC. However, whether and how they are interconnected during EC development remains unclear. Here, we demonstrate that the deacetylase SIRT7 is upregulated in EC patients and mouse models, facilitating EC progression in vitro and in vivo. Mechanistically, in an estrogen-dependent fashion, SIRT7 mediates PTEN deacetylation at K260, promoting PTEN ubiquitination by the E3 ligase NEDD4L, accelerating PTEN degradation and, consequently, expediting EC metastasis. Additionally, SIRT7 expression strongly correlates with poor survival in EC patients with wild-type PTEN, though no significant correlation is observed in PTEN mutation patients. These results lay the foundation for the study of targeting estrogen-SIRT7-PTEN axis, to restore PTEN abundance, offering potential avenues for EC therapy.
Collapse
Affiliation(s)
- Zhiyi Hu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ming Tang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yujia Huang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bailian Cai
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoxiang Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Guofang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ao Huang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmacy, Changsha Medical University, Changsha, China
| | - Xiaoqi Li
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ab Rauf Shah
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lijun Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qian Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xianghong Xu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wen Lu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Xiaoping Wan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
2
|
Zhang Y, Qing J, Li Y, Gao X, Lu D, Wang Y, Gu L, Zhang H, Li Z, Wang X, Zhou Y, Zhang P. PRMT7-Mediated PTEN Activation Enhances Bone Regeneration in Female Mice. Int J Mol Sci 2025; 26:2981. [PMID: 40243588 PMCID: PMC11988880 DOI: 10.3390/ijms26072981] [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: 02/18/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Epigenetic regulation provides new insights into the mechanisms of osteogenic differentiation and identifies potential targets for treating bone-related diseases. However, the specific regulatory networks and mechanisms involved still need further investigation. In this study, we identify PRMT7 as a novel epigenetic regulator of mesenchymal stem cells (MSCs) osteogenic commitment. Conditional knockout of Prmt7 in mice reveals a significant impairment in osteogenesis and bone regeneration, specifically in females, affecting both femurs and mandibles, with no noticeable effect in males. Mechanistically, PRMT7 modulates MSCs osteogenic differentiation by activating PTEN. Specifically, PRMT7 enhances PTEN transcription by increasing H3R2me1 levels at the PTEN promoter. Additionally, PRMT7 interacts with the PTEN protein and stabilizes nuclear PTEN, revealing an unprecedented pathway. Notably, overexpression of PTEN alleviates the osteogenic deficits observed in Prmt7-deficient mice. This research establishes PRMT7 as a potential therapeutic target for promoting bone formation/regeneration and offers novel molecular insights into the PRMT7-PTEN regulatory axis, underscoring its significance in bone biology and regenerative medicine.
Collapse
Affiliation(s)
- Yingfei Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Jia Qing
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yang Li
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Xin Gao
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Dazhuang Lu
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yiyang Wang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Lanxin Gu
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Hui Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Zechuan Li
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Xu Wang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| |
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
|
Sun H, Meng Y, Yao L, Du S, Li Y, Zhou Q, Liu Y, Dian Y, Sun Y, Wang X, Liang X, Deng G, Chen X, Zeng F. Ubiquitin-specific protease 22 controls melanoma metastasis and vulnerability to ferroptosis through targeting SIRT1/PTEN/PI3K signaling. MedComm (Beijing) 2024; 5:e684. [PMID: 39135915 PMCID: PMC11318338 DOI: 10.1002/mco2.684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 08/15/2024] Open
Abstract
Metastasis is a major contributing factor that affects the prognosis of melanoma patients. Nevertheless, the underlying molecular mechanisms involved in melanoma metastasis are not yet entirely understood. Here, we identified ubiquitin-specific protease 22 (USP22) as a pro-oncogenic protein in melanoma through screening the survival profiles of 52 ubiquitin-specific proteases (USPs). USP22 demonstrates a strong association with poor clinical outcomes and is significantly overexpressed in melanoma. Ablation of USP22 expression remarkably attenuates melanoma migration, invasion, and epithelial-mesenchymal transition in vitro and suppresses melanoma metastasis in vivo. Mechanistically, USP22 controls melanoma metastasis through the SIRT1/PTEN/PI3K pathway. In addition, we conducted an United States Food and Drug Administration-approved drug library screening and identified topotecan as a clinically applicable USP22-targeting molecule by promoting proteasomal degradation of USP22. Finally, we found that both pharmacological and genetic silence of USP22 sensitize RSL3-induced ferroptosis through suppressing the PI3K/Akt/mTOR pathway and its downstream SCD, and ferroptosis inhibitor could partly rescued the decreased lung metastasis by topotecan in vivo. Overall, our findings reveal a prometastatic role of USP22 and identify topotecan as a potent USP22-targeting drug to limit melanoma metastasis.
Collapse
Affiliation(s)
- Huiyan Sun
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
- Department of Breast ReconstructionTianjin Medical UniversityCancer Institute and HospitalTianjinChina
| | - Yu Meng
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Lei Yao
- Department of Liver SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Songtao Du
- Department of Colorectal Surgical OncologyThe Tumor Hospital of Harbin Medical UniversityHarbinChina
| | - Yayun Li
- Department of DermatologyThe Third Xiangya Hospital Central South UniversityChangshaChina
| | - Qian Zhou
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yihuang Liu
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yating Dian
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Yuming Sun
- Department of Plastic and Cosmetic SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Xiaomin Wang
- Department of Breast SurgeryXiangya Hospital Central South UniversityChangshaChina
| | - Xiao‐wei Liang
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Guangtong Deng
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Xiang Chen
- Department of DermatologyXiangya Hospital Central South UniversityChangshaChina
- National Engineering Research Center of Personalized Diagnostic and Therapeutic TechnologyChangshaChina
- Furong LaboratoryChangshaChina
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)ChangshaChina
| | - Furong Zeng
- Department of OncologyXiangya Hospital Central South UniversityChangshaChina
| |
Collapse
|
5
|
Li X, Yang P, Hou X, Ji S. Post-Translational Modification of PTEN Protein: Quantity and Activity. Oncol Rev 2024; 18:1430237. [PMID: 39144161 PMCID: PMC11321960 DOI: 10.3389/or.2024.1430237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/04/2024] [Indexed: 08/16/2024] Open
Abstract
Post-translational modifications play crucial roles in regulating protein functions and stabilities. PTEN is a critical tumor suppressor involved in regulating cellular proliferation, survival, and migration processes. However, dysregulation of PTEN is common in various human cancers. PTEN stability and activation/suppression have been extensively studied in the context of tumorigenesis through inhibition of the PI3K/AKT signaling pathway. PTEN undergoes various post-translational modifications, primarily including phosphorylation, acetylation, ubiquitination, SUMOylation, neddylation, and oxidation, which finely tune its activity and stability. Generally, phosphorylation modulates PTEN activity through its lipid phosphatase function, leading to altered power of the signaling pathways. Acetylation influences PTEN protein stability and degradation rate. SUMOylation has been implicated in PTEN localization and interactions with other proteins, affecting its overall function. Neddylation, as a novel modification of PTEN, is a key regulatory mechanism in the loss of tumor suppressor function of PTEN. Although current therapeutic approaches focus primarily on inhibiting PI3 kinase, understanding the post-translational modifications of PTEN could help provide new therapeutic strategies that can restore PTEN's role in PIP3-dependent tumors. The present review summarizes the major recent developments in the regulation of PTEN protein level and activity. We expect that these insights will contribute to better understanding of this critical tumor suppressor and its potential implications for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiao Li
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Pu Yang
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Xiaoli Hou
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
| | - Shaoping Ji
- Department of Basic Medicine, Zhengzhou Shuqing Medical College, Zhengzhou, Henan, China
- Department of Biochemistry and Molecular Biology, Medical School, Henan University, Kaifeng, Henan, China
| |
Collapse
|
6
|
San José-Enériz E, Gimenez-Camino N, Rabal O, Garate L, Miranda E, Gómez-Echarte N, García F, Charalampopoulou S, Sáez E, Vilas-Zornoza A, San Martín-Uriz P, Valcárcel LV, Barrena N, Alignani D, Tamariz-Amador LE, Pérez-Ruiz A, Hilscher S, Schutkowski M, Alfonso-Pierola A, Martinez-Calle N, Larrayoz MJ, Paiva B, Calasanz MJ, Muñoz J, Isasa M, Martin-Subero JI, Pineda-Lucena A, Oyarzabal J, Agirre X, Prósper F. Epigenetic-based differentiation therapy for Acute Myeloid Leukemia. Nat Commun 2024; 15:5570. [PMID: 38956053 PMCID: PMC11219871 DOI: 10.1038/s41467-024-49784-y] [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: 02/02/2023] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
Despite the development of novel therapies for acute myeloid leukemia, outcomes remain poor for most patients, and therapeutic improvements are an urgent unmet need. Although treatment regimens promoting differentiation have succeeded in the treatment of acute promyelocytic leukemia, their role in other acute myeloid leukemia subtypes needs to be explored. Here we identify and characterize two lysine deacetylase inhibitors, CM-444 and CM-1758, exhibiting the capacity to promote myeloid differentiation in all acute myeloid leukemia subtypes at low non-cytotoxic doses, unlike other commercial histone deacetylase inhibitors. Analyzing the acetylome after CM-444 and CM-1758 treatment reveals modulation of non-histone proteins involved in the enhancer-promoter chromatin regulatory complex, including bromodomain proteins. This acetylation is essential for enhancing the expression of key transcription factors directly involved in the differentiation therapy induced by CM-444/CM-1758 in acute myeloid leukemia. In summary, these compounds may represent effective differentiation-based therapeutic agents across acute myeloid leukemia subtypes with a potential mechanism for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Edurne San José-Enériz
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Naroa Gimenez-Camino
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Obdulia Rabal
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Leire Garate
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Estibaliz Miranda
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Nahia Gómez-Echarte
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Fernando García
- ProteoRed-ISCIII, Unidad de Proteómica, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Stella Charalampopoulou
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Casanova 143, 08036, Barcelona, Spain
| | - Elena Sáez
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Amaia Vilas-Zornoza
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Patxi San Martín-Uriz
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Luis V Valcárcel
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- TECNUN, Universidad de Navarra, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Naroa Barrena
- TECNUN, Universidad de Navarra, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Diego Alignani
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Luis Esteban Tamariz-Amador
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - Ana Pérez-Ruiz
- Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Sebastian Hilscher
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
| | - Mike Schutkowski
- Department of Enzymology, Charles Tanford Protein Center, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
- Department of Medicinal Chemistry, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, 06120, Halle, Germany
| | - Ana Alfonso-Pierola
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - Nicolás Martinez-Calle
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - María José Larrayoz
- CIMA LAB Diagnostics, Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Bruno Paiva
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
| | - María José Calasanz
- CIMA LAB Diagnostics, Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Javier Muñoz
- Biocruces Bizkaia Health Research Institute, Cruces Plaza, 48903, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009, Bilbao, Spain
| | - Marta Isasa
- ProteoRed-ISCIII, Unidad de Proteómica, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - José Ignacio Martin-Subero
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Casanova 143, 08036, Barcelona, Spain
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys 23, 08010, Barcelona, Spain
| | - Antonio Pineda-Lucena
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Julen Oyarzabal
- Small-Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Avenida Pío XII 55, 31008, Pamplona, Spain.
| | - Xabier Agirre
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain.
| | - Felipe Prósper
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029, Madrid, Spain.
- Departmento de Hematología, Clínica Universidad de Navarra, and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain.
| |
Collapse
|
7
|
Saleh Z, Moccia MC, Ladd Z, Joneja U, Li Y, Spitz F, Hong YK, Gao T. Pancreatic Neuroendocrine Tumors: Signaling Pathways and Epigenetic Regulation. Int J Mol Sci 2024; 25:1331. [PMID: 38279330 PMCID: PMC10816436 DOI: 10.3390/ijms25021331] [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: 12/20/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Pancreatic neuroendocrine tumors (PNETs) are characterized by dysregulated signaling pathways that are crucial for tumor formation and progression. The efficacy of traditional therapies is limited, particularly in the treatment of PNETs at an advanced stage. Epigenetic alterations profoundly impact the activity of signaling pathways in cancer development, offering potential opportunities for drug development. There is currently a lack of extensive research on epigenetic regulation in PNETs. To fill this gap, we first summarize major signaling events that are involved in PNET development. Then, we discuss the epigenetic regulation of these signaling pathways in the context of both PNETs and commonly occurring-and therefore more extensively studied-malignancies. Finally, we will offer a perspective on the future research direction of the PNET epigenome and its potential applications in patient care.
Collapse
Affiliation(s)
- Zena Saleh
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
| | - Matthew C. Moccia
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
| | - Zachary Ladd
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
| | - Upasana Joneja
- Department of Pathology, Cooper University Health Care, Camden, NJ 08103, USA
| | - Yahui Li
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
| | - Francis Spitz
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
| | - Young Ki Hong
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
| | - Tao Gao
- Department of Surgery, Cooper University Health Care, Camden, NJ 08103, USA; (Z.S.); (Z.L.)
- Camden Cancer Research Center, Camden, NJ 08103, USA
| |
Collapse
|
8
|
Wan J, Cheng C, Hu J, Huang H, Han Q, Jie Z, Zou Q, Shi JH, Yu X. De novo NAD + synthesis contributes to CD8 + T cell metabolic fitness and antitumor function. Cell Rep 2023; 42:113518. [PMID: 38041812 DOI: 10.1016/j.celrep.2023.113518] [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: 08/15/2023] [Revised: 09/30/2023] [Accepted: 11/15/2023] [Indexed: 12/04/2023] Open
Abstract
The dysfunction and clonal constriction of tumor-infiltrating CD8+ T cells are accompanied by alterations in cellular metabolism; however, how the cell-intrinsic metabolic pathway specifies intratumoral CD8+ T cell features remains elusive. Here, we show that cell-autonomous generation of nicotinamide adenine dinucleotide (NAD+) via the kynurenine pathway (KP) contributes to the maintenance of intratumoral CD8+ T cell metabolic and functional fitness. De novo NAD+ synthesis is involved in CD8+ T cell metabolism and antitumor function. KP-derived NAD+ promotes PTEN deacetylation, thereby facilitating PTEN degradation and preventing PTEN-dependent metabolic defects. Importantly, impaired cell-autonomous NAD+ synthesis limits CD8+ T cell responses in human colorectal cancer samples. Our results reveal that KP-derived NAD+ regulates the CD8+ T cell metabolic and functional state by restricting PTEN activity and suggest that modulation of de novo NAD+ synthesis could restore CD8+ T cell metabolic fitness and antitumor function.
Collapse
Affiliation(s)
- Jie Wan
- Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Cheng Cheng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 119 South Fourth Ring Western Road, Fengtai District, Beijing, China
| | - Jiajia Hu
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haiyan Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiaoqiao Han
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Zuliang Jie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Qiang Zou
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| | - Jian-Hong Shi
- Central Laboratory, Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding 071000, Hebei Province, China.
| | - Xiaoyan Yu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China.
| |
Collapse
|
9
|
Jang H, Chen J, Iakoucheva LM, Nussinov R. Cancer and Autism: How PTEN Mutations Degrade Function at the Membrane and Isoform Expression in the Human Brain. J Mol Biol 2023; 435:168354. [PMID: 37935253 PMCID: PMC10842829 DOI: 10.1016/j.jmb.2023.168354] [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: 09/08/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Mutations causing loss of PTEN lipid phosphatase activity can promote cancer, benign tumors (PHTS), and neurodevelopmental disorders (NDDs). Exactly how they preferentially trigger distinct phenotypic outcomes has been puzzling. Here, we demonstrate that PTEN mutations differentially allosterically bias P loop dynamics and its connection to the catalytic site, affecting catalytic activity. NDD-related mutations are likely to sample conformations of the functional wild-type state, while sampled conformations for the strong, cancer-related driver mutation hotspots favor catalysis-primed conformations, suggesting that NDD mutations are likely to be weaker, and our large-scale simulations show why. Prenatal PTEN isoform expression data suggest exons 5 and 7, which harbor NDD mutations, as cancer-risk carriers. Since cancer requires more than a single mutation, our conformational and genomic analysis helps discover how same protein mutations can foster different clinical manifestations, articulates a role for co-occurring background latent driver mutations, and uncovers relationships of splicing isoform expression to life expectancy.
Collapse
Affiliation(s)
- Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Jiaye Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA
| | - Lilia M Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
10
|
Luo G, Liu B, Fu T, Liu Y, Li B, Li N, Geng Q. The Role of Histone Deacetylases in Acute Lung Injury-Friend or Foe. Int J Mol Sci 2023; 24:ijms24097876. [PMID: 37175583 PMCID: PMC10178380 DOI: 10.3390/ijms24097876] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
Acute lung injury (ALI), caused by intrapulmonary or extrapulmonary factors such as pneumonia, shock, and sepsis, eventually disrupts the alveolar-capillary barrier, resulting in diffuse pulmonary oedema and microatasis, manifested by refractory hypoxemia, and respiratory distress. Not only is ALI highly lethal, but even if a patient survives, there are also multiple sequelae. Currently, there is no better treatment than supportive care, and we urgently need to find new targets to improve ALI. Histone deacetylases (HDACs) are epigenetically important enzymes that, together with histone acetylases (HATs), regulate the acetylation levels of histones and non-histones. While HDAC inhibitors (HDACis) play a therapeutic role in cancer, inflammatory, and neurodegenerative diseases, there is also a large body of evidence suggesting the potential of HDACs as therapeutic targets in ALI. This review explores the unique mechanisms of HDACs in different cell types of ALI, including macrophages, pulmonary vascular endothelial cells (VECs), alveolar epithelial cells (AECs), and neutrophils.
Collapse
Affiliation(s)
- Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tinglv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Boyang Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
11
|
Ghosh G, Misra S, Ray R, Chowdhury SG, Karmakar P. Phospho PTEN mediated dephosphorylation of mitotic kinase PLK1 and Aurora Kinase A prevents aneuploidy and preserves genomic stability. Med Oncol 2023; 40:119. [PMID: 36930246 DOI: 10.1007/s12032-023-01985-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
PTEN, dual phosphatase tumor suppressor protein, is found to be frequently mutated in various cancers. Post-translational modification of PTEN is important for its sub-cellular localization and catalytic functions. But how these modifications affect cytological damage and aneuploidy is not studied in detail. We focus on the role of phosphatase activity along with C-terminal phosphorylation of PTEN in perspective of cytological damage like micronucleus, nuclear bud, and nuclear bridge formation. Our data suggest that wild-type PTEN, but not phospho-mutant PTEN significantly reduces cytological damage in PTEN null PC3 cells. In case of phosphatase-dead PTEN, cytological damage markers are increased during 24 h recovery after DNA damage. When we use phosphorylation and phosphatase-dead dual mutant PTEN, the extent of different cytological DNA damage parameters are similar to phosphatase-dead PTEN. We also find that both of those activities are essential for maintaining chromosome numbers. PTEN null cells exhibit significantly aberrant γ-tubulin pole formation during metaphase. Interestingly, we observed that p-PTEN localized to spindle poles along with PLK1 and Aurora Kinase A. Further depletion of phosphorylation and phosphatase activity of PTEN increases the expression of p-Aurora Kinase A (T288) and p-PLK1 (T210), compared to cells expressing wild-type PTEN. Again, wild-type PTEN but not phosphorylation-dead mutant is able to physically interact with PLK1 and Aurora Kinase A. Thus, our study suggests that the phosphorylation-dependent interaction of PTEN with PLK1 and Aurora Kinase A causes dephosphorylation of those mitotic kinases and by lowering their hyperphosphorylation status, PTEN prevents aberrant chromosome segregation in metaphase.
Collapse
Affiliation(s)
- Ginia Ghosh
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India
| | - Sandip Misra
- Department of Microbiology, Bidhannagar College, Salt Lake, Kolkata, West Bengal, India
| | - Rachayeeta Ray
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India
| | - Sougata Ghosh Chowdhury
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India
| | - Parimal Karmakar
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, West Bengal, India.
| |
Collapse
|
12
|
Zheng YC, Kang HQ, Wang B, Zhu YZ, Mamun MAA, Zhao LF, Nie HQ, Liu Y, Zhao LJ, Zhang XN, Gao MM, Jiang DD, Liu HM, Gao Y. Curriculum vitae of HDAC6 in solid tumors. Int J Biol Macromol 2023; 230:123219. [PMID: 36642357 DOI: 10.1016/j.ijbiomac.2023.123219] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Histone deacetylase 6 (HDAC6) is the only member of the HDAC family that resides primarily in the cytoplasm with two catalytic domains and a ubiquitin-binding domain. HDAC6 is highly expressed in various solid tumors and participates in a wide range of biological activities, including hormone receptors, the p53 signaling pathway, and the kinase cascade signaling pathway due to its unique structural foundation and abundant substrate types. Additionally, HDAC6 can function as an oncogenic factor in solid tumors, boosting tumor cell proliferation, invasion and metastasis, drug resistance, stemness, and lowering tumor cell immunogenicity, so assisting in carcinogenesis. Pan-HDAC inhibitors for cancer prevention are associated with potential cardiotoxicity in clinical investigations. It's interesting that HDAC6 silencing didn't cause any significant harm to normal cells. Currently, the use of HDAC6 specific inhibitors, individually or in combination, is among the most promising therapies in solid tumors. This review's objective is to give a general overview of the structure, biological functions, and mechanism of HDAC6 in solid tumor cells and in the immunological milieu and discuss the preclinical and clinical trials of selective HDAC6 inhibitors. These endeavors highlight that targeting HDAC6 could effectively kill tumor cells and enhance patients' immunity during solid tumor therapy.
Collapse
Affiliation(s)
- Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hui-Qin Kang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Bo Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China; Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Yuan-Zai Zhu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - M A A Mamun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Long-Fei Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hai-Qian Nie
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ying Liu
- Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan 450001, China
| | - Li-Juan Zhao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Xiao-Nan Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mei-Mei Gao
- Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Henan 450001, China
| | - Dan-Dan Jiang
- Department of Pharmacy, People's Hospital of Henan Province, Zhengzhou University, Henan 450001, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, Institute of Drug Discovery and Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
13
|
Zhang H, Song W, Ma X, Yu M, Chen L, Tao Y. Acetylation stabilizes the signaling protein WISP2 by preventing its degradation to suppress the progression of acute myeloid leukemia. J Biol Chem 2023; 299:102971. [PMID: 36736423 PMCID: PMC9996369 DOI: 10.1016/j.jbc.2023.102971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Acute myeloid leukemia (AML) is challenging to treat due to its heterogeneity, prompting a deep understanding of its pathogenesis mechanisms, diagnosis, and treatment. Here, we found reduced expression and acetylation levels of WISP2 in bone marrow mononuclear cells from AML patients and that AML patients with lower WISP2 expression tended to have reduced survival. At the functional level, overexpression of WISP2 in leukemia cells (HL-60 and Kasumi-1) suppressed cell proliferation, induced cell apoptosis, and exerted antileukemic effects in an in vivo model of AML. Our mechanistic investigation demonstrated that WISP2 deacetylation was regulated by the deacetylase histone deacetylase (HDAC)3. In addition, we determined that crosstalk between acetylation and ubiquitination was involved in the modulation of WISP2 expression in AML. Deacetylation of WISP2 decreased the stability of the WISP2 protein by boosting its ubiquitination mediated by NEDD4 and proteasomal degradation. Moreover, pan-HDAC inhibitors (valproic acid and trichostatin A) and an HDAC3-specific inhibitor (RGFP966) induced WISP2 acetylation at lysine K6 and prevented WISP2 degradation. This regulation led to inhibition of proliferation and induction of apoptosis in AML cells. In summary, our study revealed that WISP2 contributes to tumor suppression in AML, which provided an experimental framework for WISP2 as a candidate for gene therapy of AML.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China; Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| | - Wenjun Song
- Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China; Graduate School, Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Xinying Ma
- Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China; Graduate School, Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Mingxiao Yu
- Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China; Graduate School, Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Lulu Chen
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China; Institute of Blood and Marrow Transplantation, Jining Medical University, Jining, Shandong, China
| | - Yanling Tao
- Department of Pediatric Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
14
|
Jang H, Chen J, Iakoucheva LM, Nussinov R. How PTEN mutations degrade function at the membrane and life expectancy of carriers of mutations in the human brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525746. [PMID: 36747841 PMCID: PMC9900933 DOI: 10.1101/2023.01.26.525746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PTEN dysfunction, caused by loss of lipid phosphatase activity or deletion, promotes pathologies, cancer, benign tumors, and neurodevelopmental disorders (NDDs). Despite efforts, exactly how the mutations trigger distinct phenotypic outcomes, cancer or NDD, has been puzzling. It has also been unclear how to distinguish between mutations harbored by isoforms, are they cancer or NDDs-related. Here we address both. We demonstrate that PTEN mutations differentially allosterically bias P-loop dynamics and its connection to the catalytic site, affecting catalytic activity. NDD-related mutations are likely to sample conformations present in the wild-type, while sampled conformations sheltering cancer-related hotspots favor catalysis-prone conformations, suggesting that NDD mutations are weaker. Analysis of isoform expression data indicates that if the transcript has NDD-related mutations, alone or in combination with cancer hotspots, there is high prenatal expression. If no mutations within the measured days, low expression levels. Cancer mutations promote stronger signaling and cell proliferation; NDDs' are weaker, influencing brain cell differentiation. Further, exon 5 is impacted by NDD or non-NDD mutations, while exon 7 is exclusively impacted by NDD mutations. Our comprehensive conformational and genomic analysis helps discover how same allele mutations can foster different clinical manifestations and uncovers correlations of splicing isoform expression to life expectancy.
Collapse
Affiliation(s)
- Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, U.S.A
| | - Jiaye Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, U.S.A
| | - Lilia M Iakoucheva
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, U.S.A
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, U.S.A
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, U.S.A
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
15
|
Targeting PTEN Regulation by Post Translational Modifications. Cancers (Basel) 2022; 14:cancers14225613. [PMID: 36428706 PMCID: PMC9688753 DOI: 10.3390/cancers14225613] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Phosphatidylinositol-3,4,5-triphosphate (PIP3) is a lipidic second messenger present at very low concentrations in resting normal cells. PIP3 levels, though, increase quickly and transiently after growth factor addition, upon activation of phosphatidylinositol 3-kinase (PI3-kinase). PIP3 is required for the activation of intracellular signaling pathways that induce cell proliferation, cell migration, and survival. Given the critical role of this second messenger for cellular responses, PIP3 levels must be tightly regulated. The lipid phosphatase PTEN (phosphatase and tensin-homolog in chromosome 10) is the phosphatase responsible for PIP3 dephosphorylation to PIP2. PTEN tumor suppressor is frequently inactivated in endometrium and prostate carcinomas, and also in glioblastoma, illustrating the contribution of elevated PIP3 levels for cancer development. PTEN biological activity can be modulated by heterozygous gene loss, gene mutation, and epigenetic or transcriptional alterations. In addition, PTEN can also be regulated by post-translational modifications. Acetylation, oxidation, phosphorylation, sumoylation, and ubiquitination can alter PTEN stability, cellular localization, or activity, highlighting the complexity of PTEN regulation. While current strategies to treat tumors exhibiting a deregulated PI3-kinase/PTEN axis have focused on PI3-kinase inhibition, a better understanding of PTEN post-translational modifications could provide new therapeutic strategies to restore PTEN action in PIP3-dependent tumors.
Collapse
|
16
|
Perevalova AM, Kobelev VS, Sisakyan VG, Gulyaeva LF, Pustylnyak VO. Role of Tumor Suppressor PTEN and Its Regulation in Malignant Transformation of Endometrium. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1310-1326. [PMID: 36509719 DOI: 10.1134/s0006297922110104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Tumor-suppressive effects of PTEN are well-known, but modern evidence suggest that they are not limited to its ability to inhibit pro-oncogenic PI3K/AKT signaling pathway. Features of PTEN structure facilitate its interaction with substrates of different nature and display its activity in various ways both in the cytoplasm and in cell nuclei, which makes it possible to take a broader look at its ability to suppress tumor growth. The possible mechanisms of the loss of PTEN effects are also diverse - PTEN can be regulated at many levels, leading to change in the protein activity or its amount in the cell, while their significance for the development of malignant tumors has yet to be studied. Here we summarize the current data on the PTEN structure, its functions and changes in its regulatory mechanisms during malignant transformation of the cells, focusing on one of the most sensitive to the loss of PTEN types of malignant tumors - endometrial cancer.
Collapse
Affiliation(s)
| | - Vyacheslav S Kobelev
- Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, 630117, Russia
| | - Virab G Sisakyan
- Novosibirsk Regional Oncology Center, Novosibirsk, 630108, Russia
| | - Lyudmila F Gulyaeva
- Novosibirsk State University, Novosibirsk, 630090, Russia.,Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, 630117, Russia
| | - Vladimir O Pustylnyak
- Novosibirsk State University, Novosibirsk, 630090, Russia.,Federal Research Center of Fundamental and Translational Medicine, Novosibirsk, 630117, Russia
| |
Collapse
|
17
|
Dang F, Wei W. Targeting the acetylation signaling pathway in cancer therapy. Semin Cancer Biol 2022; 85:209-218. [PMID: 33705871 PMCID: PMC8423867 DOI: 10.1016/j.semcancer.2021.03.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/22/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022]
Abstract
Acetylation represents one of the major post-translational protein modifications, which introduces an acetyl functional group into amino acids such as the lysine residue to yield an acetate ester bond, neutralizing its positive charge. Regulation of protein functions by acetylation occurs in multiple ways, such as affecting protein stability, activity, localization, and interaction with other proteins or DNA. It has been well documented that the recruitment of histone acetyltransferases (HATs) and histone deacetylases (HDACs) to the transcriptional machinery can modulate histone acetylation status, which is directly involved in the dynamic regulation of genes controlling cell proliferation and division. Dysregulation of gene expression is involved in tumorigenesis and aberrant activation of histone deacetylases has been reported in several types of cancer. Moreover, there is growing body of evidence showing that acetylation is widely involved in non-histone proteins to impact their roles in various cellular processes including tumorigenesis. As such, small molecular compounds inhibiting HAT or HDAC enzymatic activities have been developed and investigated for therapeutic purpose. Here we review the recent progress in our understanding of protein acetylation and discuss the therapeutic potential of targeting the acetylation signaling pathway in cancer.
Collapse
Affiliation(s)
- Fabin Dang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
18
|
Kaur S, Rajoria P, Chopra M. HDAC6: A unique HDAC family member as a cancer target. Cell Oncol (Dordr) 2022; 45:779-829. [PMID: 36036883 DOI: 10.1007/s13402-022-00704-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND HDAC6, a structurally and functionally distinct member of the HDAC family, is an integral part of multiple cellular functions such as cell proliferation, apoptosis, senescence, DNA damage and genomic stability, all of which when deregulated contribute to carcinogenesis. Among several HDAC family members known so far, HDAC6 holds a unique position. It differs from the other HDAC family members not only in terms of its subcellular localization, but also in terms of its substrate repertoire and hence cellular functions. Recent findings have considerably expanded the research related to the substrate pool, biological functions and regulation of HDAC6. Studies in HDAC6 knockout mice highlighted the importance of HDAC6 as a cell survival player in stressful situations, making it an important anticancer target. There is ample evidence stressing the importance of HDAC6 as an anti-cancer synergistic partner of many chemotherapeutic drugs. HDAC6 inhibitors have been found to enhance the effectiveness of conventional chemotherapeutic drugs such as DNA damaging agents, proteasome inhibitors and microtubule inhibitors, thereby highlighting the importance of combination therapies involving HDAC6 inhibitors and other anti-cancer agents. CONCLUSIONS Here, we present a review on HDAC6 with emphasis on its role as a critical regulator of specific physiological cellular pathways which when deregulated contribute to tumorigenesis, thereby highlighting the importance of HDAC6 inhibitors as important anticancer agents alone and in combination with other chemotherapeutic drugs. We also discuss the synergistic anticancer effect of combination therapies of HDAC6 inhibitors with conventional chemotherapeutic drugs.
Collapse
Affiliation(s)
- Sumeet Kaur
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Prerna Rajoria
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Madhu Chopra
- Laboratory of Molecular Modeling and Anticancer Drug Development, Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
19
|
Liu Y, Vandekeere A, Xu M, Fendt SM, Altea-Manzano P. Metabolite-derived protein modifications modulating oncogenic signaling. Front Oncol 2022; 12:988626. [PMID: 36226054 PMCID: PMC9549695 DOI: 10.3389/fonc.2022.988626] [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] [Received: 07/07/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Malignant growth is defined by multiple aberrant cellular features, including metabolic rewiring, inactivation of tumor suppressors and the activation of oncogenes. Even though these features have been described as separate hallmarks, many studies have shown an extensive mutual regulatory relationship amongst them. On one hand, the change in expression or activity of tumor suppressors and oncogenes has extensive direct and indirect effects on cellular metabolism, activating metabolic pathways required for malignant growth. On the other hand, the tumor microenvironment and tumor intrinsic metabolic alterations result in changes in intracellular metabolite levels, which directly modulate the protein modification of oncogenes and tumor suppressors at both epigenetic and post-translational levels. In this mini-review, we summarize the crosstalk between tumor suppressors/oncogenes and metabolism-induced protein modifications at both levels and explore the impact of metabolic (micro)environments in shaping these.
Collapse
Affiliation(s)
- Yawen Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
- Laboratory of Cellular Metabolism and Metaboli Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Anke Vandekeere
- Laboratory of Cellular Metabolism and Metaboli Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metaboli Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- *Correspondence: Sarah-Maria Fendt, ; Patricia Altea-Manzano,
| | - Patricia Altea-Manzano
- Laboratory of Cellular Metabolism and Metaboli Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- *Correspondence: Sarah-Maria Fendt, ; Patricia Altea-Manzano,
| |
Collapse
|
20
|
Zhang X, Guo N, Jin H, Liu R, Zhang Z, Cheng C, Fan Z, Zhang G, Xiao M, Wu S, Zhao Y, Lu X. Bisphenol A drives di(2-ethylhexyl) phthalate promoting thyroid tumorigenesis via regulating HDAC6/PTEN and c-MYC signaling. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:127911. [PMID: 34910997 DOI: 10.1016/j.jhazmat.2021.127911] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 06/14/2023]
Abstract
Bisphenol A (BPA) and di-(2-ethylhcxyl) phthalate (DEHP) are exist widespread in the environment and produce adverse effect to human as environmental disruptors (EDCs). Epidemiological studies have found that the exposure of DEHP and BPA could increase the susceptibility to thyroid diseases including thyroid cancer and benign thyroid nodules. Due to the existence of multiple pollutants in our daily life, the mixed toxic effects of exposure and their interrelationships may distinguish from the exposure to a single chemical, so it is of great significance to explore the mixed toxic effect of DEHP and BPA co-exposure. Thyroid, as one of the target organs of EDCs, is prone to tumor occurrence, however, whether the mixture of BPA and DEHP will affect the occurrence of thyroid cancer is still obscure. We aim to investigate the effect of single or combined exposure to BPA and DEHP on the occurrence of thyroid cancer. An animal model of exposure to BPA and DEHP was firstly established to evaluate their effect on DMD-induced thyroid cancer. Additionally, human thyroid cancer cells BCPAP and thyroid cells Nthy-ori3-1 were used to further clarify some possible mechanisms of BPA and MEHP, the main metabolite of DEHP. Consequently, we found that BPA alone could increase the incidence of thyroid tumors in female rats compared with DEHP, and DEHP enhanced the effect of BPA on cancer promotion. BPA alone and in combination with DEHP mainly induced the expression of HDAC6, inhibited tumor suppressor gene PTEN upregulated the expression of oncogene c-MYC, and eventually elevate the susceptibility to thyroid tumors. Mechanistically, BPA alone and in combination with MEHP could significantly induce the proliferation of BCPAP cells depending on HDAC6, which could modulate H3K9ac to inhibit PTEN, activate AKT signaling pathway, and simultaneously upregulate the expression of c-MYC. Interestingly, we found that BPA alone and in combination with MEHP could significantly induce the proliferation of Nthy-ori3-1 cells independent on HDAC6 via activating ERK signaling pathway. Taken together, these findings not only provide new evidence of the promoting effect of BPA and DEHP on thyroid cancer but also discusses some possible mechanisms underlying these effects.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China
| | - Nan Guo
- Department of head and Neck Surgery, Cancer hospital of China Medical University/Liaoning Cancer hospital & Institute, Shenyang, PR China
| | - Hao Jin
- Jin Zhou Center for Disease Control and Prevention, Jinzhou, PR China
| | - Renqi Liu
- Jin Zhou Center for Disease Control and Prevention, Jinzhou, PR China
| | - Zhen Zhang
- Jin Zhou Center for Disease Control and Prevention, Jinzhou, PR China
| | - Cheng Cheng
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China
| | - Zhijun Fan
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China
| | - Guopei Zhang
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China
| | - Mingyang Xiao
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China
| | - Shengwen Wu
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China
| | - Yuejiao Zhao
- Department of head and Neck Surgery, Cancer hospital of China Medical University/Liaoning Cancer hospital & Institute, Shenyang, PR China.
| | - Xiaobo Lu
- Department of Toxicology, School of Public health, China Medical University, Shenyang, PR China.
| |
Collapse
|
21
|
Yu B, Liu L, Cai F, Peng Y, Tang X, Zeng D, Li T, Zhang F, Liang Y, Yuan X, Li J, Dai Z, Liao Q, Lv XB. The synergistic anticancer effect of the bromodomain inhibitor OTX015 and histone deacetylase 6 inhibitor WT-161 in osteosarcoma. Cancer Cell Int 2022; 22:64. [PMID: 35135529 PMCID: PMC8822767 DOI: 10.1186/s12935-022-02443-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 01/01/2022] [Indexed: 12/04/2022] Open
Abstract
Background Osteosarcoma (OS) is a tumour with a high malignancy level and a poor prognosis. First-line chemotherapy for OS has not been improved for many decades. Bromodomain and extraterminal domain (BET) and histone deacetylases (HDACs) regulate histone acetylation in tandem, and BET and HDACs have emerged as potential cancer therapeutic targets. Methods Cell proliferation, migration, invasion, colony formation, and sphere-forming assays were performed with the two inhibitors alone or in combination to evaluate their suppressive effect on the malignant properties of OS cells. Apoptosis and the cell cycle profile were measured by flow cytometry. The synergistic inhibitory effect of OTX015/WT-161 on tumours was also examined in a nude mouse xenograft model. Results The combined therapy of OTX015/WT-161 synergistically inhibited growth, migration, and invasion and induced apoptosis, resulting in G1/S arrest of OS cells. Additionally, OTX015/WT-161 inhibited the self-renewal ability of OS stem cells (OSCs) in a synergistic manner. Further mechanistic exploration revealed that the synergistic downregulation of β-catenin by OTX015-mediated suppression of FZD2 and WT-161-mediated upregulation of PTEN may be critical for the synergistic effect. Finally, the results of an in vivo assay showed that tumour xenografts were significantly decreased after treatment with the OTX015/WT-161 combination compared with OTX015 or WT-161 alone. Conclusions Our findings in this study demonstrated that OTX015 and WT-161 had synergistic anticancer efficacy against OS, and their combination might be a promising therapeutic strategy for OS. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02443-y.
Collapse
Affiliation(s)
- Bo Yu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Lang Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Feng Cai
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Yuanxiang Peng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Xiaofeng Tang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Duo Zeng
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Teng Li
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Feifei Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Yiping Liang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Xuhui Yuan
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Jiayu Li
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Zhengzai Dai
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China
| | - Qi Liao
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China. .,Department of Orthopedics, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.
| | - Xiao-Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, Central Laboratory, The First Hospital of Nanchang, The Third Affiliated Hospital of Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, Jiangxi, People's Republic of China.
| |
Collapse
|
22
|
Sun X, Shu Y, Ye G, Wu C, Xu M, Gao R, Huang D, Zhang J. Histone deacetylase inhibitors inhibit cervical cancer growth through Parkin acetylation-mediated mitophagy. Acta Pharm Sin B 2022; 12:838-852. [PMID: 35256949 PMCID: PMC8897022 DOI: 10.1016/j.apsb.2021.07.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/30/2021] [Accepted: 06/16/2021] [Indexed: 02/08/2023] Open
Abstract
Parkin, an E3 ubiquitin ligase, plays a role in maintaining mitochondrial homeostasis through targeting damaged mitochondria for mitophagy. Accumulating evidence suggests that the acetylation modification of the key mitophagy machinery influences mitophagy level, but the underlying mechanism is poorly understood. Here, our study demonstrated that inhibition of histone deacetylase (HDAC) by treatment of HDACis activates mitophagy through mediating Parkin acetylation, leading to inhibition of cervical cancer cell proliferation. Bioinformatics analysis shows that Parkin expression is inversely correlated with HDAC2 expression in human cervical cancer, indicating the low acetylation level of Parkin. Using mass spectrometry, Parkin is identified to interact with two upstream molecules, acetylase acetyl-CoA acetyltransferase 1 (ACAT1) and deacetylase HDAC2. Under treatment of suberoylanilide hydroxamic acid (SAHA), Parkin is acetylated at lysine residues 129, 220 and 349, located in different domains of Parkin protein. In in vitro experiments, combined mutation of Parkin largely attenuate the interaction of Parkin with PTEN induced putative kinase 1 (PINK1) and the function of Parkin in mitophagy induction and tumor suppression. In tumor xenografts, the expression of mutant Parkin impairs the tumor suppressive effect of Parkin and decreases the anticancer activity of SAHA. Our results reveal an acetylation-dependent regulatory mechanism governing Parkin in mitophagy and cervical carcinogenesis, which offers a new mitophagy modulation strategy for cancer therapy.
Collapse
Key Words
- ACAT1
- ACAT1, acetyl-CoA acetyltransferase 1
- Acetylation
- CCK-8, cell counting kit-8
- COXⅣ, cytochrome c oxidase Ⅳ
- Cervical cancer
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- HDAC, histone deacetylase
- HDAC2
- HIF-1α, hypoxia inducible factor-1α
- HSP60, heat shock protein 60 kDa
- LC3, microtubule-associated proteins 1A/1B light chain 3
- MFN2, mitofusion 2
- MS, mass spectrometry
- Mitophagy
- PARK2, Parkin
- PINK1, PTEN induced putative kinase 1
- Parkin
- ROS, reactive oxygen species
- SAHA, suberoylanilide hydroxamic acid
- TIM23, translocase of the inner membrane 23
- TOMM20, translocase of outer mitochondrial membrane 20
- TSA, trichostatin A
- Tumor suppression
- ULK1, unc-51 like autophagy activating kinase 1
- Ubiquitination
- VDAC1, voltage-dependent anion-selective channel protein 1
Collapse
Affiliation(s)
- Xin Sun
- Department of Oncology, Cancer Center of Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Yuhan Shu
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310028, China
| | - Guiqin Ye
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310014, China
| | - Caixia Wu
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Mengting Xu
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou 310028, China
| | - Ruilan Gao
- Department of Hematology, the First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Dongsheng Huang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou Medical College, Hangzhou 310014, China
- Corresponding authors.
| | - Jianbin Zhang
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
- Corresponding authors.
| |
Collapse
|
23
|
Ke DYJ, El-Sahli S, Wang L. The Potential of Natural Products in the Treatment of Triple-Negative Breast Cancer. Curr Cancer Drug Targets 2021; 22:388-403. [PMID: 34970954 DOI: 10.2174/1568009622666211231140623] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/01/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that lacks receptors for targeted therapy. Consequently, chemotherapy is currently the mainstay of systemic treatment options. However, the enrichment of cancer stem cells (CSC, a subpopulation with stem-cell characteristics and tumor-initiating propensity) promotes chemo-resistance and tumorigenesis, resulting in cancer recurrence and relapse. Furthermore, toxic side effects of chemotherapeutics reduce patient wellbeing. Natural products, specifically compounds derived from plants, have the potential to treat TNBC and target CSCs by inhibiting CSC signaling pathways. Literature evidence from six promising compounds were reviewed, including sulforaphane, curcumin, genistein, resveratrol, lycopene, and epigallocatechin-3-gallate. These compounds have been shown to promote cell cycle arrest and apoptosis in TNBC cells. They also could inhibit the epithelial-mesenchymal transition (EMT) that plays an important role in metastasis. In addition, those natural compounds have been found to inhibit pathways important for CSCs, such as NF-κB, PI3K/Akt/mTOR, Notch 1, Wnt/β-catenin, and YAP. Clinicals trials conducted on these compounds have shown varying degrees of effectiveness. Epidemiological case-control studies for the compounds commonly consumed in certain human populations have also been summarized. While in vivo and in vitro data are promising, further basic and clinical investigations are required. Likely, natural products in combination with other drugs may hold great potential to improve TNBC treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
- Danny Yu Jia Ke
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Ottawa, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- The Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Sara El-Sahli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Ottawa, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- The Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Ottawa, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- The Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
24
|
Enhancing Therapeutic Approaches for Melanoma Patients Targeting Epigenetic Modifiers. Cancers (Basel) 2021; 13:cancers13246180. [PMID: 34944799 PMCID: PMC8699560 DOI: 10.3390/cancers13246180] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 12/25/2022] Open
Abstract
Melanoma is the least common but deadliest type of skin cancer. Melanomagenesis is driven by a series of mutations and epigenetic alterations in oncogenes and tumor suppressor genes that allow melanomas to grow, evolve, and metastasize. Epigenetic alterations can also lead to immune evasion and development of resistance to therapies. Although the standard of care for melanoma patients includes surgery, targeted therapies, and immune checkpoint blockade, other therapeutic approaches like radiation therapy, chemotherapy, and immune cell-based therapies are used for patients with advanced disease or unresponsive to the conventional first-line therapies. Targeted therapies such as the use of BRAF and MEK inhibitors and immune checkpoint inhibitors such as anti-PD-1 and anti-CTLA4 only improve the survival of a small subset of patients. Thus, there is an urgent need to identify alternative standalone or combinatorial therapies. Epigenetic modifiers have gained attention as therapeutic targets as they modulate multiple cellular and immune-related processes. Due to melanoma's susceptibility to extrinsic factors and reversible nature, epigenetic drugs are investigated as a therapeutic avenue and as adjuvants for targeted therapies and immune checkpoint inhibitors, as they can sensitize and/or reverse resistance to these therapies, thus enhancing their therapeutic efficacy. This review gives an overview of the role of epigenetic changes in melanoma progression and resistance. In addition, we evaluate the latest advances in preclinical and clinical research studying combinatorial therapies and discuss the use of epigenetic drugs such as HDAC and DNMT inhibitors as potential adjuvants for melanoma patients.
Collapse
|
25
|
Lanzi C, Favini E, Dal Bo L, Tortoreto M, Arrighetti N, Zaffaroni N, Cassinelli G. Upregulation of ERK-EGR1-heparanase axis by HDAC inhibitors provides targets for rational therapeutic intervention in synovial sarcoma. J Exp Clin Cancer Res 2021; 40:381. [PMID: 34857011 PMCID: PMC8638516 DOI: 10.1186/s13046-021-02150-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Synovial sarcoma (SS) is an aggressive soft tissue tumor with limited therapeutic options in advanced stage. SS18-SSX fusion oncogenes, which are the hallmarks of SS, cause epigenetic rewiring involving histone deacetylases (HDACs). Promising preclinical studies supporting HDAC targeting for SS treatment were not reflected in clinical trials with HDAC inhibitor (HDACi) monotherapies. We investigated pathways implicated in SS cell response to HDACi to identify vulnerabilities exploitable in combination treatments and improve the therapeutic efficacy of HDACi-based regimens. METHODS Antiproliferative and proapoptotic effects of the HDACi SAHA and FK228 were examined in SS cell lines in parallel with biochemical and molecular analyses to bring out cytoprotective pathways. Treatments combining HDACi with drugs targeting HDACi-activated prosurvival pathways were tested in functional assays in vitro and in a SS orthotopic xenograft model. Molecular mechanisms underlying synergisms were investigated in SS cells through pharmacological and gene silencing approaches and validated by qRT-PCR and Western blotting. RESULTS SS cell response to HDACi was consistently characterized by activation of a cytoprotective and auto-sustaining axis involving ERKs, EGR1, and the β-endoglycosidase heparanase, a well recognized pleiotropic player in tumorigenesis and disease progression. HDAC inhibition was shown to upregulate heparanase by inducing expression of the positive regulator EGR1 and by hampering negative regulation by p53 through its acetylation. Interception of HDACi-induced ERK-EGR1-heparanase pathway by cell co-treatment with a MEK inhibitor (trametinib) or a heparanase inhibitor (SST0001/roneparstat) enhanced antiproliferative and pro-apoptotic effects. HDAC and heparanase inhibitors had opposite effects on histone acetylation and nuclear heparanase levels. The combination of SAHA with SST0001 prevented the upregulation of ERK-EGR1-heparanase induced by the HDACi and promoted caspase-dependent cell death. In vivo, the combined treatment with SAHA and SST0001 potentiated the antitumor efficacy against the CME-1 orthotopic SS model as compared to single agent administration. CONCLUSIONS The present study provides preclinical rationale and mechanistic insights into drug combinatory strategies based on the use of ERK pathway and heparanase inhibitors to improve the efficacy of HDACi-based antitumor therapies in SS. The involvement of classes of agents already clinically available, or under clinical evaluation, indicates the transferability potential of the proposed approaches.
Collapse
Affiliation(s)
- Cinzia Lanzi
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Enrica Favini
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Laura Dal Bo
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Monica Tortoreto
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Noemi Arrighetti
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Nadia Zaffaroni
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy
| | - Giuliana Cassinelli
- Department of Applied Research and Technological Development, Molecular Pharmacology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133, Milan, Italy.
| |
Collapse
|
26
|
A global map of associations between types of protein posttranslational modifications and human genetic diseases. iScience 2021; 24:102917. [PMID: 34430807 PMCID: PMC8365368 DOI: 10.1016/j.isci.2021.102917] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/27/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
There are >200 types of protein posttranslational modifications (PTMs) described in eukaryotes, each with unique proteome coverage and functions. We hypothesized that some genetic diseases may be caused by the removal of a specific type of PTMs by genomic variants and the consequent deregulation of particular functions. We collected >320,000 human PTMs representing 59 types and crossed them with >4M nonsynonymous DNA variants annotated with predicted pathogenicity and disease associations. We report >1.74M PTM-variant co-occurrences that an enrichment analysis distributed into 215 pairwise associations between 18 PTM types and 148 genetic diseases. Of them, 42% were not previously described. Removal of lysine acetylation exerts the most pronounced effect, and less studied PTM types such as S-glutathionylation or S-nitrosylation show relevance. Using pathogenicity predictions, we identified PTM sites that may produce particular diseases if prevented. Our results provide evidence of a substantial impact of PTM-specific removal on the pathogenesis of genetic diseases and phenotypes. There is an enrichment of disease-associated nsSNVs preventing certain types of PTMs We report 215 pairwise associations between 18 PTM types and 148 genetic diseases The removal of lysine acetylation exerts the most pronounced effect We report a set of PTM sites that may produce particular diseases if prevented
Collapse
|
27
|
Wu HJ, Chu PY. Epigenetic Regulation of Breast Cancer Stem Cells Contributing to Carcinogenesis and Therapeutic Implications. Int J Mol Sci 2021; 22:ijms22158113. [PMID: 34360879 PMCID: PMC8348144 DOI: 10.3390/ijms22158113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Globally, breast cancer has remained the most commonly diagnosed cancer and the leading cause of cancer death among women. Breast cancer is a highly heterogeneous and phenotypically diverse group of diseases, which require different selection of treatments. Breast cancer stem cells (BCSCs), a small subset of cancer cells with stem cell-like properties, play essential roles in breast cancer progression, recurrence, metastasis, chemoresistance and treatments. Epigenetics is defined as inheritable changes in gene expression without alteration in DNA sequence. Epigenetic regulation includes DNA methylation and demethylation, as well as histone modifications. Aberrant epigenetic regulation results in carcinogenesis. In this review, the mechanism of epigenetic regulation involved in carcinogenesis, therapeutic resistance and metastasis of BCSCs will be discussed, and finally, the therapies targeting these biomarkers will be presented.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975611855; Fax: +886-47227116
| |
Collapse
|
28
|
He T, Zhang X, Hao J, Ding S. Phosphatase and Tensin Homolog in Non-neoplastic Digestive Disease: More Than Just Tumor Suppressor. Front Physiol 2021; 12:684529. [PMID: 34140896 PMCID: PMC8204087 DOI: 10.3389/fphys.2021.684529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
The Phosphatase and tensin homolog (PTEN) gene is one of the most important tumor suppressor genes, which acts through its unique protein phosphatase and lipid phosphatase activity. PTEN protein is widely distributed and exhibits complex biological functions and regulatory modes. It is involved in the regulation of cell morphology, proliferation, differentiation, adhesion, and migration through a variety of signaling pathways. The role of PTEN in malignant tumors of the digestive system is well documented. Recent studies have indicated that PTEN may be closely related to many other benign processes in digestive organs. Emerging evidence suggests that PTEN is a potential therapeutic target in the context of several non-neoplastic diseases of the digestive tract. The recent discovery of PTEN isoforms is expected to help unravel more biological effects of PTEN in non-neoplastic digestive diseases.
Collapse
Affiliation(s)
- Tianyu He
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xiaoyun Zhang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jianyu Hao
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
29
|
Ge Q, Huang X, Fang S, Guo S, Liu Y, Lin W, Xiong M. Conditional Generative Adversarial Networks for Individualized Treatment Effect Estimation and Treatment Selection. Front Genet 2020; 11:585804. [PMID: 33362849 PMCID: PMC7759680 DOI: 10.3389/fgene.2020.585804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/18/2020] [Indexed: 11/13/2022] Open
Abstract
Treatment response is heterogeneous. However, the classical methods treat the treatment response as homogeneous and estimate the average treatment effects. The traditional methods are difficult to apply to precision oncology. Artificial intelligence (AI) is a powerful tool for precision oncology. It can accurately estimate the individualized treatment effects and learn optimal treatment choices. Therefore, the AI approach can substantially improve progress and treatment outcomes of patients. One AI approach, conditional generative adversarial nets for inference of individualized treatment effects (GANITE) has been developed. However, GANITE can only deal with binary treatment and does not provide a tool for optimal treatment selection. To overcome these limitations, we modify conditional generative adversarial networks (MCGANs) to allow estimation of individualized effects of any types of treatments including binary, categorical and continuous treatments. We propose to use sparse techniques for selection of biomarkers that predict the best treatment for each patient. Simulations show that MCGANs outperform seven other state-of-the-art methods: linear regression (LR), Bayesian linear ridge regression (BLR), k-Nearest Neighbor (KNN), random forest classification [RF (C)], random forest regression [RF (R)], logistic regression (LogR), and support vector machine (SVM). To illustrate their applications, the proposed MCGANs were applied to 256 patients with newly diagnosed acute myeloid leukemia (AML) who were treated with high dose ara-C (HDAC), Idarubicin (IDA) and both of these two treatments (HDAC+IDA) at M. D. Anderson Cancer Center. Our results showed that MCGAN can more accurately and robustly estimate the individualized treatment effects than other state-of-the art methods. Several biomarkers such as GSK3, BILIRUBIN, SMAC are identified and a total of 30 biomarkers can explain 36.8% of treatment effect variation.
Collapse
Affiliation(s)
- Qiyang Ge
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
- School of Mathematical Sciences, Fudan University, Shanghai, China
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shicheng Guo
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, United States
| | - Yuanyuan Liu
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Wei Lin
- School of Mathematical Sciences, Fudan University, Shanghai, China
| | - Momiao Xiong
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
30
|
Rodrigues DA, Pinheiro PSM, Fraga CAM. Multitarget Inhibition of Histone Deacetylase (HDAC) and Phosphatidylinositol-3-kinase (PI3K): Current and Future Prospects. ChemMedChem 2020; 16:448-457. [PMID: 33049098 DOI: 10.1002/cmdc.202000643] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/06/2020] [Indexed: 12/11/2022]
Abstract
The discovery of histone deacetylase (HDAC) inhibitors is a hot topic in the medicinal chemistry community regarding cancer research. This is related primarily to two factors: success in the clinic, e. g., the four FDA-approved HDAC inhibitors, and strong versatility to combine their pharmacophoric features to design new hybrid compounds with multitarget profiles. Thus, the selection of adequate pharmacophores to combine, i. e., combining targets that can result in a synergistic effect, is desirable, as it increases the probability of discovering a new useful therapeutic strategy. In this work, we highlight the design of multitarget HDAC/PI3K inhibitors. Although this approach is still in its early stages, many significant works have described the design and pharmacological evaluation of this new promising class of multitarget inhibitors, where compound CUDC-907, which is already in clinical trials, stands out. Therefore, the question emerges of whether there still space for the design and evaluation of new multitarget HDAC/PI3K inhibitors. When considering the selectivity profile of the described multitarget compounds, the answer appears to be in the affirmative, especially since the first examples of compounds with a certain selectivity profile only recently appeared in 2020.
Collapse
Affiliation(s)
- Daniel A Rodrigues
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Pedro S M Pinheiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| | - Carlos A M Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Fan X, Kraynak J, Knisely JPS, Formenti SC, Shen WH. PTEN as a Guardian of the Genome: Pathways and Targets. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036194. [PMID: 31932469 DOI: 10.1101/cshperspect.a036194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Faithful transmission of genetic information is only possible with the structural and functional integrity of the genome. PTEN has been recognized as a guardian of the genome since the identification of its noncanonical localization and function in the nucleus. Yet, the role of PTEN in guarding the genome relies on integration of diverse mechanisms elicited by its canonical activity in antagonizing PI3K as well as emerging noncanonical functions. In the nucleus, PTEN maintains the structural integrity of chromosomes and the architecture of heterochromatin by physically interacting with chromosomal and nucleosomal components. PTEN also controls the functional integrity of key genetic transmission machineries by promoting proper assembly of the replisome and mitotic spindles. Deregulation of PTEN signaling impairs genome integrity, leading to spontaneous replication/mitotic stress and subsequent stress tolerance. Identification of novel targets of PTEN signaling and illumination of the interplay of diverse PTEN pathways in genome maintenance will help us better understand mechanisms underlying tumor evolution and therapeutic resistance.
Collapse
Affiliation(s)
- Xinyi Fan
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Jonathan P S Knisely
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| | - Wen H Shen
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA.,Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, New York 10065, USA
| |
Collapse
|
32
|
Posttranslational Regulation and Conformational Plasticity of PTEN. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036095. [PMID: 31932468 DOI: 10.1101/cshperspect.a036095] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor that is frequently down-modulated in human cancer. PTEN inhibits the phosphatidylinositol 3-phosphate kinase (PI3K)/AKT pathway through its lipid phosphatase activity. Multiple PI3K/AKT-independent actions of PTEN, protein-phosphatase activities and functions within the nucleus have also been described. PTEN, therefore, regulates many cellular processes including cell proliferation, survival, genomic integrity, polarity, migration, and invasion. Even a modest decrease in the functional dose of PTEN may promote cancer development. Understanding the molecular and cellular mechanisms that regulate PTEN protein levels and function, and how these may go awry in cancer contexts, is, therefore, key to fully understanding the role of PTEN in tumorigenesis. Here, we discuss current knowledge on posttranslational control and conformational plasticity of PTEN, as well as therapeutic possibilities toward reestablishment of PTEN tumor-suppressor activity in cancer.
Collapse
|
33
|
Marques AEM, do Nascimento Filho CHV, Marinho Bezerra TM, Guerra ENS, Castilho RM, Squarize CH. Entinostat is a novel therapeutic agent to treat oral squamous cell carcinoma. J Oral Pathol Med 2020; 49:771-779. [PMID: 32450006 DOI: 10.1111/jop.13039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/18/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Alterations of the epigenome may influence cancer initiation and progression. At the cellular level, histones are key regulators of chromatin accessibility and gene transcription; thus, the inhibition of histone deacetylase enzymes (HDACs) constitutes an attractive target for therapy. In this study, we investigated the effects of the HDAC inhibitor entinostat on oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS We tested the effects of entinostat on OSCC cell lines. Cell viability and growth were analyzed using MTT assay. Cell cycle analysis, cell apoptosis, cancer stem cell (CSC) content, and the concentration of reactive oxygen species (ROS) in OSCC tumor cells were assessed using flow cytometry. The expression of histones and cell cycle regulatory proteins was examined by Western blot. RESULTS The administration of entinostat resulted in reduced proliferation of OSCC cells, followed by cell cycle arrest at the G0/G1 phase, as well as substantial tumor apoptosis. We found an increase in ROS production and significant reductions in CSCs. We also found that entinostat caused increased acetylation histone H3 and histone H4, and changes in the expression of cell cycle-associated proteins such as p21. CONCLUSION This study indicates that entinostat is a potential novel therapeutic agent for OSCC by halting tumor proliferation, inducing cytotoxicity and intracellular ROS, and attacking the CSCs.
Collapse
Affiliation(s)
- Ana Elizia M Marques
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasilia, Brasilia, Brazil
| | - Carlos Henrique V do Nascimento Filho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Thamara M Marinho Bezerra
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,School of Pharmacy, Dentistry and Nursing, Federal University of Ceara, Fortaleza, Brazil
| | - Eliete N S Guerra
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasilia, Brasilia, Brazil
| | - Rogerio M Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,The University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Cristiane H Squarize
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, Division of Oral Pathology Oral Radiology and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,The University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| |
Collapse
|
34
|
Zhao L, Li R, Qiu JZ, Yu JB, Cao Y, Yuan RT. YY1-mediated PTEN dephosphorylation antagonizes IR-induced DNA repair contributing to tongue squamous cell carcinoma radiosensitization. Mol Cell Probes 2020; 53:101577. [PMID: 32334006 DOI: 10.1016/j.mcp.2020.101577] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022]
Abstract
Ionizing radiation (IR) confers a survival advantage in tongue squamous cell carcinoma (TSCC), however, IR resistance limits its efficacy. Although Yin Yang 1 (YY1) has been reported to play a role in genotoxic drug resistance by accelerating DNA repair, its role in TSCC radioresistance remains unclear. In this study, we examined YY1 mRNA and protein expression in human tongue cancer samples using qRT-PCR and western blotting, respectively. DNA array data identified YY1 mRNA expression in IR sensitivity or resistance cell lines and tissues. Tongue carcinoma primary cells and CAL27 cells with YY1 stably overexpressed or knocked-down were exposed to IR and evaluated for cell proliferation and apoptosis by CCK8-assay and caspase-3 assay, respectively. We also examined DNA damage- or repair-related indicators, such as YY1, p-H2AX, nuclear PTEN, p-PTEN, and Rad51 through Western blot analysis. Additionally, we explored the mechanism of IR-induced PTEN nuclear translocation by introducing a series of PTEN phosphorylation site mutations and co-IP assay. We observed that YY1 mRNA and protein are highly expressed in TSCC tissues, which was correlated with worse overall survival. Moreover, higher expression of YY1 and Rad51 was observed in radioresistant cells and tissues, overexpression of YY1 led to IR resistance in TSCC cells, whereas YY1 knockdown sensitized TSCC cells to IR. The underlying mechanism showed that the overexpression of YY1 upregulated nuclear PTEN and Rad51 expression, which is essential for DNA repair. IR upregulated YY1, nuclear PTEN, and Rad51; thus, knockdown of YY1 completely blocked IR-induced upregulation of nuclear PTEN/Rad51. IR upregulated PTEN phosphorylation, and mutation of the phosphorylation site of Ser380 nearly completely blocked IR-induced PTEN nuclear translocation. Furthermore, the phosphatase PP2A negatively regulated pS380-PTEN, and knockdown of YY1 completely blocked IR-induced pS380-PTEN through PP2A. In conclusion, knockdown of YY1 enhanced TSCC radiosensitivity through PP2A-mediated dephosphorylation of PTEN Ser380; thus, antagonizing the IR-induced nuclear PTEN/Rad51 axis and targeting YY1 may reverse IR resistance in TSCC.
Collapse
Affiliation(s)
- Lu Zhao
- Center of Oral Medicine, Qingdao Municipal Hospital, #5 Donghai Middle Road, Qingdao, 266000, PR China; School of Stomatology, Qingdao University, #19 JiangSu Road, Qingdao, 266000, PR China
| | - Ran Li
- Department of Oral and Maxillo-facial Surgery, Weifang Medical University Affiliated Qingdao Stomatological Hospital, #17 Dexian Road, Qingdao, 266000, PR China
| | - Jian-Zhong Qiu
- Center of Oral Medicine, Qingdao Municipal Hospital, #5 Donghai Middle Road, Qingdao, 266000, PR China; School of Stomatology, Qingdao University, #19 JiangSu Road, Qingdao, 266000, PR China
| | - Jiang-Bo Yu
- Center of Oral Medicine, Qingdao Municipal Hospital, #5 Donghai Middle Road, Qingdao, 266000, PR China; School of Stomatology, Qingdao University, #19 JiangSu Road, Qingdao, 266000, PR China
| | - Yang Cao
- Center of Oral Medicine, Qingdao Municipal Hospital, #5 Donghai Middle Road, Qingdao, 266000, PR China; School of Stomatology, Qingdao University, #19 JiangSu Road, Qingdao, 266000, PR China
| | - Rong-Tao Yuan
- Center of Oral Medicine, Qingdao Municipal Hospital, #5 Donghai Middle Road, Qingdao, 266000, PR China; School of Stomatology, Qingdao University, #19 JiangSu Road, Qingdao, 266000, PR China.
| |
Collapse
|
35
|
Pulido R, Mingo J, Gaafar A, Nunes-Xavier CE, Luna S, Torices L, Angulo JC, López JI. Precise Immunodetection of PTEN Protein in Human Neoplasia. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a036293. [PMID: 31501265 DOI: 10.1101/cshperspect.a036293] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PTEN is a major tumor-suppressor protein whose expression and biological activity are frequently diminished in sporadic or inherited cancers. PTEN gene deletion or loss-of-function mutations favor tumor cell growth and are commonly found in clinical practice. In addition, diminished PTEN protein expression is also frequently observed in tumor samples from cancer patients in the absence of PTEN gene alterations. This makes PTEN protein levels a potential biomarker parameter in clinical oncology, which can guide therapeutic decisions. The specific detection of PTEN protein can be achieved by using highly defined anti-PTEN monoclonal antibodies (mAbs), characterized with precision in terms of sensitivity for the detection technique, specificity for PTEN binding, and constraints of epitope recognition. This is especially relevant taking into consideration that PTEN is highly targeted by mutations and posttranslational modifications, and different PTEN protein isoforms exist. The precise characterization of anti-PTEN mAb reactivity is an important step in the validation of these reagents as diagnostic and prognostic tools in clinical oncology, including their routine use in analytical immunohistochemistry (IHC). Here, we review the current status on the use of well-defined anti-PTEN mAbs for PTEN immunodetection in the clinical context and discuss their potential usefulness and limitations for a more precise cancer diagnosis and patient benefit.
Collapse
Affiliation(s)
- Rafael Pulido
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao 48011, Spain
| | - Janire Mingo
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Ayman Gaafar
- Department of Pathology, Cruces University Hospital, Barakaldo 48903, Spain
| | - Caroline E Nunes-Xavier
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain.,Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo N-0310, Norway
| | - Sandra Luna
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Leire Torices
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Javier C Angulo
- Department of Urology, University Hospital of Getafe, Getafe, Madrid 28904, Spain.,Clinical Department, European University of Madrid, Laureate Universities, Madrid 28904, Spain
| | - José I López
- Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain.,Department of Pathology, Cruces University Hospital, Barakaldo 48903, Spain.,University of the Basque Country, Leioa 48940, Spain
| |
Collapse
|
36
|
Rahhal R, Seto E. Emerging roles of histone modifications and HDACs in RNA splicing. Nucleic Acids Res 2019; 47:4911-4926. [PMID: 31162605 PMCID: PMC6547430 DOI: 10.1093/nar/gkz292] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022] Open
Abstract
Histone modifications and RNA splicing, two seemingly unrelated gene regulatory processes, greatly increase proteome diversity and profoundly influence normal as well as pathological eukaryotic cellular functions. Like many histone modifying enzymes, histone deacetylases (HDACs) play critical roles in governing cellular behaviors and are indispensable in numerous biological processes. While the association between RNA splicing and histone modifications is beginning to be recognized, a lack of knowledge exists regarding the role of HDACs in splicing. Recent studies however, reveal that HDACs interact with spliceosomal and ribonucleoprotein complexes, actively control the acetylation states of splicing-associated histone marks and splicing factors, and thereby unexpectedly could modulate splicing. Here, we review the role of histone/protein modifications and HDACs in RNA splicing and discuss the convergence of two parallel fields, which supports the argument that HDACs, and perhaps most histone modifying enzymes, are much more versatile and far more complicated than their initially proposed functions. Analogously, an HDAC-RNA splicing connection suggests that splicing is regulated by additional upstream factors and pathways yet to be defined or not fully characterized. Some human diseases share common underlying causes of aberrant HDACs and dysregulated RNA splicing and, thus, further support the potential link between HDACs and RNA splicing.
Collapse
Affiliation(s)
- Raneen Rahhal
- George Washington Cancer Center, Department of Biochemistry & Molecular Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC 20037, USA
| | - Edward Seto
- George Washington Cancer Center, Department of Biochemistry & Molecular Medicine, George Washington University School of Medicine & Health Sciences, Washington, DC 20037, USA
| |
Collapse
|
37
|
Chang H, Cai Z, Roberts TM. The Mechanisms Underlying PTEN Loss in Human Tumors Suggest Potential Therapeutic Opportunities. Biomolecules 2019; 9:biom9110713. [PMID: 31703360 PMCID: PMC6921025 DOI: 10.3390/biom9110713] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/01/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
In this review, we will first briefly describe the diverse molecular mechanisms associated with PTEN loss of function in cancer. We will then proceed to discuss the molecular mechanisms linking PTEN loss to PI3K activation and demonstrate how these mechanisms suggest possible therapeutic approaches for patients with PTEN-null tumors.
Collapse
Affiliation(s)
- Hyeyoun Chang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (H.C.); (Z.C.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
- KIST-DFCI On-Site Lab, Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Zhenying Cai
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (H.C.); (Z.C.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Thomas M. Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (H.C.); (Z.C.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: ; Tel.: +1-617-632-3049
| |
Collapse
|
38
|
Silencing of HDAC6 as a therapeutic target in chronic lymphocytic leukemia. Blood Adv 2019; 2:3012-3024. [PMID: 30425065 DOI: 10.1182/bloodadvances.2018020065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 10/06/2018] [Indexed: 12/22/2022] Open
Abstract
Although the treatment paradigm for chronic lymphocytic leukemia (CLL) is rapidly changing, the disease remains incurable, except with allogeneic bone marrow transplantation, and resistance, relapsed disease, and partial responses persist as significant challenges. Recent studies have uncovered roles for epigenetic modification in the regulation of mechanisms contributing to malignant progression of CLL B cells. However, the extent to which epigenetic modifiers can be targeted for therapeutic benefit in CLL patients remains poorly explored. We report for the first time that expression of epigenetic modifier histone deacetylase 6 (HDAC6) is upregulated in CLL patient samples, cell lines, and euTCL1 transgenic mouse models compared with HDAC6 in normal controls. Genetic silencing of HDAC6 conferred survival benefit in euTCL1 mice. Administration of isoform-specific HDAC6 inhibitor ACY738 in the euTCL1 aging and adoptive transfer models deterred proliferation of CLL B cells, delayed disease onset via disruption of B-cell receptor signaling, and sensitized CLL B cells to apoptosis. Furthermore, coadministration of ACY738 and ibrutinib displayed synergistic cell kill against CLL cell lines and improved overall survival compared with either single agent in vivo. These results demonstrate for the first time the therapeutic efficacy of selective HDAC6 inhibition in preclinical CLL models and suggest a rationale for the clinical development of HDAC6 inhibitors for CLL treatment, either alone or in combination with Bruton tyrosine kinase inhibition.
Collapse
|
39
|
Chi Z, Byeon HE, Seo E, Nguyen QAT, Lee W, Jeong Y, Choi J, Pandey D, Berkowitz DE, Kim JH, Lee SY. Histone deacetylase 6 inhibitor tubastatin A attenuates angiotensin II-induced hypertension by preventing cystathionine γ-lyase protein degradation. Pharmacol Res 2019; 146:104281. [PMID: 31125601 DOI: 10.1016/j.phrs.2019.104281] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/20/2019] [Accepted: 05/20/2019] [Indexed: 12/11/2022]
Abstract
Cystathionine γ-lyase (CSEγ) is a hydrogen sulfide (H2S)-producing enzyme. Endothelial H2S production can mediate vasodilatory effects, contributing to the alleviation of hypertension (high blood pressure). Recent studies have suggested a role of histone deacetylase 6 (HDAC6) in hypertension, although its underlying mechanisms are poorly understood. Here, we addressed the potential regulation of CSEγ by HDAC6 in angiotensin II (AngII)-induced hypertension and its molecular details focusing on CSEγ posttranslational modification. Treatment of mice with a selective HDAC6 inhibitor tubastatin A (TubA) alleviated high blood pressure and vasoconstriction induced by AngII. Cotreatment of the aorta and human aortic endothelial cells with TubA recovered AngII-mediated decreased H2S levels. AngII treatment upregulated HDAC6 mRNA and protein expression, but conversely downregulated CSEγ protein. Notably, potent HDAC6 inhibitors and HDAC6 siRNA as well as a proteasomal inhibitor increased CSEγ protein levels and blocked the downregulatory effect of AngII on CSEγ. In contrast, other HDAC isoforms-specific inhibitors and siRNAs did not show such blocking effects. Transfected CSEγ protein levels were also reciprocally regulated by AngII and TubA, and were reduced by wild-type, but not by deacetylase-deficient, HDAC6. Moreover, TubA significantly increased both protein stability and K73 acetylation level of CSEγ. Consistent with these results, AngII induced CSEγ ubiquitination and degradation, which was inhibited by TubA. Our results indicate that AngII promoted HDAC6-dependent deacetylation of CSEγ at K73 residue, leading to its ubiquitin-mediated proteolysis, which underlies AngII-induced hypertension. Overall, this study suggests that upregulation of CSEγ and H2S through HDAC6 inhibition may be considered as a valid strategy for preventing the progression of hypertension.
Collapse
Affiliation(s)
- Zhexi Chi
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hye-Eun Byeon
- Institute of Medical Science, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Eunjeong Seo
- Department of Biomedical Sciences, Chronic Inflammatory Disease Research Center, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Quynh-Anh T Nguyen
- Department of Biomedical Sciences, Chronic Inflammatory Disease Research Center, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Wonbeom Lee
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Republic of Korea
| | - Yunyong Jeong
- Department of Anesthesiology and Pain Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Juyong Choi
- Department of Biomedical Sciences, Chronic Inflammatory Disease Research Center, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| | - Deepesh Pandey
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan E Berkowitz
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jae Hyung Kim
- Department of Anesthesiology and Pain Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Republic of Korea.
| | - Sang Yoon Lee
- Department of Biomedical Sciences, Chronic Inflammatory Disease Research Center, Ajou University Graduate School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
40
|
Immunoepigenetics Combination Therapies: An Overview of the Role of HDACs in Cancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20092241. [PMID: 31067680 PMCID: PMC6539010 DOI: 10.3390/ijms20092241] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/23/2019] [Accepted: 04/28/2019] [Indexed: 12/26/2022] Open
Abstract
Long-standing efforts to identify the multifaceted roles of histone deacetylase inhibitors (HDACis) have positioned these agents as promising drug candidates in combatting cancer, autoimmune, neurodegenerative, and infectious diseases. The same has also encouraged the evaluation of multiple HDACi candidates in preclinical studies in cancer and other diseases as well as the FDA-approval towards clinical use for specific agents. In this review, we have discussed how the efficacy of immunotherapy can be leveraged by combining it with HDACis. We have also included a brief overview of the classification of HDACis as well as their various roles in physiological and pathophysiological scenarios to target key cellular processes promoting the initiation, establishment, and progression of cancer. Given the critical role of the tumor microenvironment (TME) towards the outcome of anticancer therapies, we have also discussed the effect of HDACis on different components of the TME. We then have gradually progressed into examples of specific pan-HDACis, class I HDACi, and selective HDACis that either have been incorporated into clinical trials or show promising preclinical effects for future consideration. Finally, we have included examples of ongoing trials for each of the above categories of HDACis as standalone agents or in combination with immunotherapeutic approaches.
Collapse
|
41
|
Selective HDAC6 inhibitors improve anti-PD-1 immune checkpoint blockade therapy by decreasing the anti-inflammatory phenotype of macrophages and down-regulation of immunosuppressive proteins in tumor cells. Sci Rep 2019; 9:6136. [PMID: 30992475 PMCID: PMC6467894 DOI: 10.1038/s41598-019-42237-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/19/2019] [Indexed: 01/29/2023] Open
Abstract
Histone deacetylases (HDACs) are involved in diverse cellular regulatory mechanisms including non-canonical functions outside the chromatin environment. Several publications have demonstrated that selective HDAC inhibitors (HDACi) can influence tumor immunogenicity and the functional activity of specific immune cells. In particular, the selective inhibition of HDAC6 has been reported to decrease tumor growth in several malignancies. However, there is still no clarity about the cellular components mediating this effect. In this study, we evaluated the HDAC6i Nexturastat A as a priming agent to facilitate the transition of the tumor microenvironment from “cold” to “hot”, and potentially augment immune check-point blockade therapies. This combination modality demonstrated to significantly reduce tumor growth in syngeneic melanoma tumor models. Additionally, we observed a complete neutralization of the up-regulation of PD-L1 and other immunosuppressive pathways induced by the treatment with anti-PD-1 blockade. This combination also showed profound changes in the tumor microenvironment such as enhanced infiltration of immune cells, increased central and effector T cell memory, and a significant reduction of pro-tumorigenic M2 macrophages. The evaluation of individual components of the tumor microenvironment suggested that the in vivo anti-tumor activity of HDAC6i is mediated by its effect on tumor cells and tumor-associated macrophages, and not directly over T cells. Overall, our results indicate that selective HDAC6i could be used as immunological priming agents to sensitize immunologically “cold” tumors and subsequently improve ongoing immune check-point blockade therapies.
Collapse
|
42
|
PTEN arginine methylation by PRMT6 suppresses PI3K-AKT signaling and modulates pre-mRNA splicing. Proc Natl Acad Sci U S A 2019; 116:6868-6877. [PMID: 30886105 DOI: 10.1073/pnas.1811028116] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Arginine methylation is a ubiquitous posttranslational modification that regulates critical cellular processes including signal transduction and pre-mRNA splicing. Here, we report that the tumor-suppressor PTEN is methylated by protein arginine methyltransferase 6 (PRMT6). Mass-spectrometry analysis reveals that PTEN is dimethylated at arginine 159 (R159). We found that PTEN is mutated at R159 in cancers, and the PTEN mutant R159K loses its capability to inhibit the PI3K-AKT cascade. Furthermore, PRMT6 is physically associated with PTEN, promotes asymmetrical dimethylation of PTEN, and regulates the PI3K-AKT cascade through PTEN R159 methylation. In addition, using transcriptome analyses, we found that PTEN R159 methylation is involved in modulation of pre-mRNA alternative splicing. Our results demonstrate that PTEN is functionally regulated by arginine methylation. We propose that PTEN arginine methylation modulates pre-mRNA alternative splicing and influences diverse physiologic processes.
Collapse
|
43
|
Depetter Y, Geurs S, De Vreese R, Goethals S, Vandoorn E, Laevens A, Steenbrugge J, Meyer E, de Tullio P, Bracke M, D'hooghe M, De Wever O. Selective pharmacological inhibitors of HDAC6 reveal biochemical activity but functional tolerance in cancer models. Int J Cancer 2019; 145:735-747. [DOI: 10.1002/ijc.32169] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/14/2018] [Accepted: 01/22/2019] [Indexed: 02/03/2023]
Affiliation(s)
- Yves Depetter
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering; Ghent University; Ghent Belgium
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences; Ghent University; Ghent Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Silke Geurs
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering; Ghent University; Ghent Belgium
| | - Rob De Vreese
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering; Ghent University; Ghent Belgium
| | - Sophie Goethals
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences; Ghent University; Ghent Belgium
| | - Elien Vandoorn
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences; Ghent University; Ghent Belgium
| | - Alien Laevens
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences; Ghent University; Ghent Belgium
| | - Jonas Steenbrugge
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Evelyne Meyer
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
- Laboratory of Biochemistry, Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Pascal de Tullio
- Center for Interdisciplinary Research on Medicines (CIRM), Metabolomics Group; Université de Liège; Liège Belgium
| | - Marc Bracke
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences; Ghent University; Ghent Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering; Ghent University; Ghent Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences; Ghent University; Ghent Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| |
Collapse
|
44
|
Yang L, Zhao L, Cui L, Huang Y, Ye J, Zhang Q, Jiang X, Zhang D, Huang Y. Decreased α-tubulin acetylation induced by an acidic environment impairs autophagosome formation and leads to rat cardiomyocyte injury. J Mol Cell Cardiol 2019; 127:143-153. [DOI: 10.1016/j.yjmcc.2018.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 12/02/2018] [Accepted: 12/20/2018] [Indexed: 11/26/2022]
|
45
|
Yang F, Wang F, Liu Y, Wang S, Li X, Huang Y, Xia Y, Cao C. Sulforaphane induces autophagy by inhibition of HDAC6-mediated PTEN activation in triple negative breast cancer cells. Life Sci 2018; 213:149-157. [DOI: 10.1016/j.lfs.2018.10.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/17/2018] [Accepted: 10/18/2018] [Indexed: 02/09/2023]
|
46
|
Yang J, Li A, Li Y, Guo X, Wang M. A novel approach for drug response prediction in cancer cell lines via network representation learning. Bioinformatics 2018; 35:1527-1535. [DOI: 10.1093/bioinformatics/bty848] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/09/2018] [Accepted: 10/09/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jianghong Yang
- School of Information Science and Technology, University of Science and Technology of China, Hefei AH230037, China
| | - Ao Li
- School of Information Science and Technology, University of Science and Technology of China, Hefei AH230037, China
- Centers for Biomedical Engineering, University of Science and Technology of China, Hefei AH230037, China
| | - Yongqiang Li
- Department of Preventive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Xiangqian Guo
- Department of Preventive Medicine, Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Minghui Wang
- School of Information Science and Technology, University of Science and Technology of China, Hefei AH230037, China
- Centers for Biomedical Engineering, University of Science and Technology of China, Hefei AH230037, China
| |
Collapse
|
47
|
Sun S, Zhang Y, Zheng J, Duan B, Cui J, Chen Y, Deng W, Ye B, Liu L, Chen Y, Du J, Gu L. HDAC6 inhibitor TST strengthens the antiproliferative effects of PI3K/mTOR inhibitor BEZ235 in breast cancer cells via suppressing RTK activation. Cell Death Dis 2018; 9:929. [PMID: 30206202 PMCID: PMC6134008 DOI: 10.1038/s41419-018-0931-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/30/2018] [Accepted: 07/31/2018] [Indexed: 11/23/2022]
Abstract
NVP-BEZ235 (BEZ235), an available dual PI3K/mTOR inhibitor, showed antitumor effect and provided a therapy strategy in carcinomas. However, the acquired upregulation of multiple receptor tyrosine kinases (RTKs) by NVP-BEZ235 in tumors limits its clinical efficacy. HDAC6, a class II histone deacetylase, is associated with expressions of multiple RTKs. The aim of this study was to detect whether co-treatment with HDAC6 inhibitor Tubastatin A (TST) would enhance the anticancer effects of BEZ235 in breast cancer cells. In this study, we described that treatment of breast cancer cell lines (T47D, BT474, and MDA-MB-468) with BEZ235 significantly triggered PI3K/mTOR signaling inactivation and increased multiple RTK expression, including EGFR, HER2, HER3, IGF-1 receptor, insulin receptor, and their phosphorylation levels. The adding of TST destabilized these RTKs in those breast cancer cells. Co-treatment with BEZ235 and TST reduced cell proliferative rate by strengthening Akt inactivation. In addition, the combination of these two drugs also cooperatively arrested cell cycle and DNA synthesis. In conclusion, the co-treatment with PI3K/mTOR inhibitor BEZ235 and HDAC6 inhibitor TST displayed additive antiproliferative effects on breast cancer cells through inactivating RTKs and established a rationable combination therapy to treat breast cancer.
Collapse
Affiliation(s)
- Shixiu Sun
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jianchao Zheng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, Guangdong, 518083, China
| | - Biao Duan
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jie Cui
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yan Chen
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wenjie Deng
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Bixing Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Lei Liu
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Luo Gu
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China. .,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
48
|
M JR, S V. BMI1 and PTEN are key determinants of breast cancer therapy: A plausible therapeutic target in breast cancer. Gene 2018; 678:302-311. [PMID: 30096458 DOI: 10.1016/j.gene.2018.08.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/11/2018] [Accepted: 08/04/2018] [Indexed: 12/12/2022]
Abstract
BMI-1 (B-lymphoma Mo-MLV insertion region 1) is a key protein partner in polycomb repressive complex 1 (PRC1) that helps in maintaining the integrity of the complex. It is also a key player in ubiquitination of histone H2A which affects gene expression pattern involved in various cellular processes such as cell proliferation, growth, DNA repair, apoptosis and senescence. In many cancers, Overexpression of BMI1correlates with advanced stages of disease, aggressive clinicopathological behavior, poor prognosis resistance to radiation and chemotherapy. BMI1 is emerging as a key player in EMT, chemo-resistance and cancer stemness. Overexpression is observed in various cancer types such as breast, primary hepatocellular carcinoma (HCC), gastric, ovarian, head and neck, pancreatic and lung cancer. Studies have shown that experimental reduction of BMI protein level in tumor cells results in inhibition of cell proliferation, induction of apoptosis and/or senescence, and increases susceptibility to cytotoxic agents and radiation therapy. Thus, inhibition of BMI1 expression particularly in breast cancer stem cells can be used as a potential strategy for the complete elimination of tumor and to prevent disease relapse. On other hand PTEN is known to be an important tumor suppressor next to p53. In many cancers particularly in breast cancer, p53 and PTEN undergo mutations. Studies have indicated the functional and mechanistic link between the BMI-1oncoprotein and tumor suppressor PTEN in the development and progression of cancer. The current review focuses on recent findings of how oncogenicity and chemo-resistance are caused by BMI1. It also highlights the transcriptional regulation between BMI1 and PTEN that dictates the therapeutic outcome in cancers where the functional p53 is absent. Herein, we have clearly demonstrated the regulation of transcription at genomic loci of BMI1 and PTEN in cancerous tissue or cells and the possible epigenetic regulation by histone deacetylase inhibitors (HDACi) at BMI1 and PTEN loci that may provide some clue for the possible therapy against TNBC in near future.
Collapse
Affiliation(s)
- Janaki Ramaiah M
- School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, India.
| | - Vaishnave S
- School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, India
| |
Collapse
|
49
|
Dempsey DR, Cole PA. Protein Chemical Approaches to Understanding PTEN Lipid Phosphatase Regulation. Methods Enzymol 2018; 607:405-422. [PMID: 30149868 DOI: 10.1016/bs.mie.2018.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Since the discovery of C-tail phosphorylation of PTEN almost 20 years ago, much progress has been made in understanding its regulatory influences on the cellular function of PTEN. Phosphorylation of Ser380, Thr382, Thr383, and Ser385 drives a PTEN conformational change from an open to closed state where catalytic function is impaired, plasma membrane binding is reduced, and cellular stability is enhanced. Despite these advances, a detailed structural and mechanistic model of how these phosphorylations impact PTEN function is lacking. We discuss here several recent approaches to analyzing PTEN phosphorylation and highlight several insights that have come from this work. We also discuss remaining challenges for the PTEN regulation field and potential directions for future research.
Collapse
Affiliation(s)
- Daniel R Dempsey
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| | - Philip A Cole
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
50
|
Chen Y, Huang Q, Liu W, Zhu Q, Cui CP, Xu L, Guo X, Wang P, Liu J, Dong G, Wei W, Liu CH, Feng Z, He F, Zhang L. Mutually exclusive acetylation and ubiquitylation of the splicing factor SRSF5 control tumor growth. Nat Commun 2018; 9:2464. [PMID: 29942010 PMCID: PMC6018636 DOI: 10.1038/s41467-018-04815-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/18/2018] [Indexed: 12/30/2022] Open
Abstract
Most tumor cells take up more glucose than normal cells. Splicing dysregulation is one of the molecular hallmarks of cancer. However, the role of splicing factor in glucose metabolism and tumor development remains poorly defined. Here, we show that upon glucose intake, the splicing factor SRSF5 is specifically induced through Tip60-mediated acetylation on K125, which antagonizes Smurf1-mediated ubiquitylation. SRSF5 promotes the alternative splicing of CCAR1 to produce CCAR1S proteins, which promote tumor growth by enhancing glucose consumption and acetyl-CoA production. Conversely, upon glucose starvation, SRSF5 is deacetylated by HDAC1, and ubiquitylated by Smurf1 on the same lysine, resulting in proteasomal degradation of SRSF5. The CCAR1L proteins accumulate to promote apoptosis. Importantly, SRSF5 is hyperacetylated and upregulated in human lung cancers, which correlates with increased CCAR1S expression and tumor progression. Thus, SRSF5 responds to high glucose to promote cancer development, and SRSF5-CCAR1 axis may be valuable targets for cancer therapeutics.
Collapse
Affiliation(s)
- Yuhan Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Affiliated BaYi Children's Hospital, PLA Army General Hospital, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China
| | - Qingyang Huang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Qiong Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chun-Ping Cui
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China.,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Liang Xu
- Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xing Guo
- Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Department of Neurobiology, Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ping Wang
- Department of Central Laboratory, Shanghai Tenth People's Hospital, School of Life Science and Technology, Tongji University, Shanghai, 200072, China
| | - Jingwen Liu
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Guanglong Dong
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhichun Feng
- Affiliated BaYi Children's Hospital, PLA Army General Hospital, National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China. .,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China. .,Department of Genomics and Proteomics, Beijing Institute of Radiation Medicine, Beijing, 100850, China. .,School of Life Science, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, China.
| |
Collapse
|