1
|
Ling R, Wang J, Fang Y, Yu Y, Su Y, Sun W, Li X, Tang X. HDAC-an important target for improving tumor radiotherapy resistance. Front Oncol 2023; 13:1193637. [PMID: 37503317 PMCID: PMC10368992 DOI: 10.3389/fonc.2023.1193637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/28/2023] [Indexed: 07/29/2023] Open
Abstract
Radiotherapy is an important means of tumor treatment, but radiotherapy resistance has been a difficult problem in the comprehensive treatment of clinical tumors. The mechanisms of radiotherapy resistance include the repair of sublethal damage and potentially lethal damage of tumor cells, cell repopulation, cell cycle redistribution, and reoxygenation. These processes are closely related to the regulation of epigenetic modifications. Histone deacetylases (HDACs), as important regulators of the epigenetic structure of cancer, are widely involved in the formation of tumor radiotherapy resistance by participating in DNA damage repair, cell cycle regulation, cell apoptosis, and other mechanisms. Although the important role of HDACs and their related inhibitors in tumor therapy has been reviewed, the relationship between HDACs and radiotherapy has not been systematically studied. This article systematically expounds for the first time the specific mechanism by which HDACs promote tumor radiotherapy resistance in vivo and in vitro and the clinical application prospects of HDAC inhibitors, aiming to provide a reference for HDAC-related drug development and guide the future research direction of HDAC inhibitors that improve tumor radiotherapy resistance.
Collapse
Affiliation(s)
- Rui Ling
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jingzhi Wang
- Department of Radiotherapy Oncology, Affiliated Yancheng First Hospital of Nanjing University Medical School, First People’s Hospital of Yancheng, Yancheng, China
| | - Yuan Fang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunpeng Yu
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yuting Su
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Sun
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoqin Li
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Tang
- Department of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
2
|
Liu R, Wu J, Guo H, Yao W, Li S, Lu Y, Jia Y, Liang X, Tang J, Zhang H. Post-translational modifications of histones: Mechanisms, biological functions, and therapeutic targets. MedComm (Beijing) 2023; 4:e292. [PMID: 37220590 PMCID: PMC10200003 DOI: 10.1002/mco2.292] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Histones are DNA-binding basic proteins found in chromosomes. After the histone translation, its amino tail undergoes various modifications, such as methylation, acetylation, phosphorylation, ubiquitination, malonylation, propionylation, butyrylation, crotonylation, and lactylation, which together constitute the "histone code." The relationship between their combination and biological function can be used as an important epigenetic marker. Methylation and demethylation of the same histone residue, acetylation and deacetylation, phosphorylation and dephosphorylation, and even methylation and acetylation between different histone residues cooperate or antagonize with each other, forming a complex network. Histone-modifying enzymes, which cause numerous histone codes, have become a hot topic in the research on cancer therapeutic targets. Therefore, a thorough understanding of the role of histone post-translational modifications (PTMs) in cell life activities is very important for preventing and treating human diseases. In this review, several most thoroughly studied and newly discovered histone PTMs are introduced. Furthermore, we focus on the histone-modifying enzymes with carcinogenic potential, their abnormal modification sites in various tumors, and multiple essential molecular regulation mechanism. Finally, we summarize the missing areas of the current research and point out the direction of future research. We hope to provide a comprehensive understanding and promote further research in this field.
Collapse
Affiliation(s)
- Ruiqi Liu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jiajun Wu
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Haiwei Guo
- Otolaryngology & Head and Neck CenterCancer CenterDepartment of Head and Neck SurgeryZhejiang Provincial People's HospitalAffiliated People's Hospital, Hangzhou Medical CollegeHangzhouZhejiangChina
| | - Weiping Yao
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Shuang Li
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentJinzhou Medical UniversityJinzhouLiaoningChina
| | - Yanwei Lu
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Yongshi Jia
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| | - Xiaodong Liang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Graduate DepartmentBengbu Medical College, BengbuAnhuiChina
| | - Jianming Tang
- Department of Radiation OncologyThe First Hospital of Lanzhou UniversityLanzhou UniversityLanzhouGansuChina
| | - Haibo Zhang
- Cancer CenterDepartment of Radiation OncologyZhejiang Provincial People's HospitalAffiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
| |
Collapse
|
3
|
Lian B, Chen X, Shen K. Inhibition of histone deacetylases attenuates tumor progression and improves immunotherapy in breast cancer. Front Immunol 2023; 14:1164514. [PMID: 36969235 PMCID: PMC10034161 DOI: 10.3389/fimmu.2023.1164514] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Breast cancer is one of the common malignancies with poor prognosis worldwide. The treatment of breast cancer patients includes surgery, radiation, hormone therapy, chemotherapy, targeted drug therapy and immunotherapy. In recent years, immunotherapy has potentiated the survival of certain breast cancer patients; however, primary resistance or acquired resistance attenuate the therapeutic outcomes. Histone acetyltransferases induce histone acetylation on lysine residues, which can be reversed by histone deacetylases (HDACs). Dysregulation of HDACs via mutation and abnormal expression contributes to tumorigenesis and tumor progression. Numerous HDAC inhibitors have been developed and exhibited the potent anti-tumor activity in a variety of cancers, including breast cancer. HDAC inhibitors ameliorated immunotherapeutic efficacy in cancer patients. In this review, we discuss the anti-tumor activity of HDAC inhibitors in breast cancer, including dacinostat, belinostat, abexinostat, mocetinotat, panobinostat, romidepsin, entinostat, vorinostat, pracinostat, tubastatin A, trichostatin A, and tucidinostat. Moreover, we uncover the mechanisms of HDAC inhibitors in improving immunotherapy in breast cancer. Furthermore, we highlight that HDAC inhibitors might be potent agents to potentiate immunotherapy in breast cancer.
Collapse
Affiliation(s)
| | | | - Kunwei Shen
- *Correspondence: Xiaosong Chen, ; Kunwei Shen,
| |
Collapse
|
4
|
Kim Y, Park K, Kim YJ, Shin SW, Kim YJ, Choi C, Noh JM. Immunomodulation of HDAC Inhibitor Entinostat Potentiates the Anticancer Effects of Radiation and PD-1 Blockade in the Murine Lewis Lung Carcinoma Model. Int J Mol Sci 2022; 23:ijms232415539. [PMID: 36555180 PMCID: PMC9779092 DOI: 10.3390/ijms232415539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Although the combination of radiotherapy and immunotherapy has proven to be effective in lung cancer treatment, it may not be sufficient to fully activate the antitumor immune response. Here, we investigated whether entinostat, a histone deacetylase inhibitor, could improve the efficacy of radiotherapy and anti-PD-1 in a murine syngeneic LL/2 tumor model. A total of 12 Gy of X-rays administered in two fractions significantly delayed tumor growth in mice, which was further enhanced by oral entinostat administration. Flow cytometry-aided immune cell profiling revealed that entinostat increased radiation-induced infiltration of myeloid-derived suppressor cells and CD8+ T cells with decreased regulatory T-cells (Tregs). Transcriptomics-based immune phenotype prediction showed that entinostat potentiated radiation-activated pathways, such as JAK/STAT3/interferon-gamma (IFN-γ) and PD-1/PD-L1 signaling. Entinostat augmented the antitumor efficacy of radiation and anti-PD-1, which may be related to an increase in IFN-γ-producing CD8+ T-cells with a decrease in Treg cells. Comparative transcriptomic profiling predicted that entinostat increased the number of dendritic cells, B cells, and T cells in tumors treated with radiation and anti-PD-1 by inducing MHC-II genes. In conclusion, our findings provided insights into how entinostat improves the efficacy of ionizing radiation plus anti-PD-1 therapy and offered clues for developing new strategies for clinical trials.
Collapse
Affiliation(s)
- Yeeun Kim
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Kyunghee Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Yeon Jeong Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sung-Won Shin
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Yeon Joo Kim
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Changhoon Choi
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea
- Correspondence: (C.C.); (J.M.N.)
| | - Jae Myoung Noh
- Department of Radiation Oncology, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Radiation Oncology, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Correspondence: (C.C.); (J.M.N.)
| |
Collapse
|
5
|
Song H, Shen R, Liu X, Yang X, Xie K, Guo Z, Wang D. Histone post-translational modification and the DNA damage response. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
6
|
Abstract
In mammalian cells, genomic DNA is packaged with histone proteins and condensed into chromatin. To gain access to the DNA, chromatin remodelling is required that is enhanced through histone post-translational modifications, which subsequently stimulate processes including DNA repair and transcription. Histone acetylation is one of the most well understood modifications and is controlled by histone acetyltransferases (HATs) and histone deacetylases (HDACs). These enzymes play critical roles in normal cellular functioning, and the dysregulation of HDAC expression in particular has been linked with the development of a number of different cancer types. Conversely, tumour cell killing following radiotherapy is triggered through DNA damage and HDACs can help co-ordinate the cellular DNA damage response which promotes radioresistance. Consequently, HDAC inhibitors have been investigated as potential radiosensitizers in vitro and in vivo to improve the efficacy or radiotherapy in specific tumour types. In this review, we provide an up-to-date summary of HDACs and their cellular functions, including in DNA damage repair. We also review evidence demonstrating that HDAC inhibitors can effectively enhance tumour radiosensitisation, and which therefore show potential for translation into the clinic for cancer patient benefit.
Collapse
|
7
|
Wu K, Chen X, Chen X, Zhang S, Xu Y, Xia B, Ma S. Suberoylanilide hydroxamic acid enhances the radiosensitivity of lung cancer cells through acetylated wild-type and mutant p53-dependent modulation of mitochondrial apoptosis. J Int Med Res 2021; 49:300060520981545. [PMID: 33557658 PMCID: PMC7876760 DOI: 10.1177/0300060520981545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Objective Suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, has
shown potential as a candidate radiosensitizer for many types of cancers.
This study aimed to explore the radiosensitization mechanism of SAHA in lung
cancer cells. Methods Mutations in p53 were generated by site-directed mutagenesis using polymerase
chain reaction. Transfection was performed to generate H1299 cells carrying
wild-type or mutant p53. The radiosensitizing enhancement ratio was
determined by clonogenic assays. Mitochondrial apoptosis was detected using
JC-1 staining and flow cytometry analysis. Results Our results showed that SAHA induced radiosensitization in H1299 cells
expressing wild-type p53, p53R175H or p53P223L, but
this enhanced clonogenic cell death was not observed in parental H1299
(p53-null) cells or H1299 cells expressing p53 with K120R, A161T and V274R
mutations. In SAHA-sensitized cells, mitochondrial apoptosis was induced
following exposure to irradiation. Additionally, we observed that a
secondary mutation at K120 (K120R) could eliminate p53-mediated
radiosensitization and mitochondrial apoptosis. Conclusions The results of this study suggest that wild-type and specific mutant forms of
p53 mediate SAHA-induced radiosensitization by regulating mitochondrial
apoptosis, and the stabilization of K120 acetylation by SAHA is the
molecular basis contributing to radiosensitization in lung cancer cells.
Collapse
Affiliation(s)
- Kan Wu
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Xueqin Chen
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Xufeng Chen
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
| | - Shirong Zhang
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Yasi Xu
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Bing Xia
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China.,Department of Oncology, Jiande Second People's Hospital, Jiande, P.R. China
| | - Shenglin Ma
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| |
Collapse
|
8
|
Mabeta P. Paradigms of vascularization in melanoma: Clinical significance and potential for therapeutic targeting. Biomed Pharmacother 2020; 127:110135. [PMID: 32334374 DOI: 10.1016/j.biopha.2020.110135] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is the most aggressive form of skin cancer. Malignant melanoma in particular has a poor prognosis and although treatment has improved, drug resistance continues to be a challenge. Angiogenesis, the formation of blood vessels from existing microvessels, precedes the progression of melanoma from a radial growth phase to a malignant phenotype. In addition, melanoma cells can form networks of vessel-like fluid conducting channels through vasculogenic mimicry (VM). Both angiogenesis and VM have been postulated to contribute to the development of resistance to treatment and to enable metastasis. Also, the metastatic spread of melanoma is highly dependent on lymphangiogenesis, the formation of lymphatic vessels from pre-existing vessels. Interestingly, the design and clinical testing of drugs that target VM and lymphangiogenesis lag behind that of angiogenesis inhibitors. Despite this, antiangiogenic drugs have not significantly improved the overall survival of melanoma patients, thus necessitating the targeting of alternative mechanisms. In this article, I review the roles of the three paradigms of tissue perfusion, namely, angiogenesis, VM and lymphangiogenesis, in promoting melanoma progression and metastasis. This article also explores the latest development and potential opportunities in the therapeutic targeting of these processes.
Collapse
Affiliation(s)
- Peace Mabeta
- Angiogenesis Laboratory, Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
9
|
Ward C, Meehan J, Gray ME, Murray AF, Argyle DJ, Kunkler IH, Langdon SP. The impact of tumour pH on cancer progression: strategies for clinical intervention. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:71-100. [PMID: 36046070 PMCID: PMC9400736 DOI: 10.37349/etat.2020.00005] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of cellular pH is frequent in solid tumours and provides potential opportunities for therapeutic intervention. The acidic microenvironment within a tumour can promote migration, invasion and metastasis of cancer cells through a variety of mechanisms. Pathways associated with the control of intracellular pH that are under consideration for intervention include carbonic anhydrase IX, the monocarboxylate transporters (MCT, MCT1 and MCT4), the vacuolar-type H+-ATPase proton pump, and the sodium-hydrogen exchanger 1. This review will describe progress in the development of inhibitors to these targets.
Collapse
Affiliation(s)
- Carol Ward
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Mark E Gray
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Alan F Murray
- School of Engineering, Institute for Integrated Micro and Nano Systems, EH9 3JL Edinburgh, UK
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, EH25 9RG Midlothian, UK
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Edinburgh Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, EH4 2XU Edinburgh, UK
| |
Collapse
|
10
|
Su T, Liu P, Ti X, Wu S, Xue X, Wang Z, Dioum E, Zhang Q. ΗΙF1α, EGR1 and SP1 co-regulate the erythropoietin receptor expression under hypoxia: an essential role in the growth of non-small cell lung cancer cells. Cell Commun Signal 2019; 17:152. [PMID: 31752873 PMCID: PMC6869211 DOI: 10.1186/s12964-019-0458-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/10/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Overexpression of erythropoietin (EPO) and EPO receptor (EPO-R) is associated with poor prognosis in non-small-cell lung carcinoma (NSCLC). Hypoxia, a potent EPO inducer, is a major stimulating factor in the growth of solid tumors. However, how EPO-R expression is regulated under hypoxia is largely unknown. METHODS The role of EPO-R in NSCLC cell proliferation was assessed by RNA interference in vitro. Luciferase reporter assays were performed to map the promoter elements involved in the EPO-R mRNA transcription. Nuclear co-immunoprecipitation and chromatin immunoprecipitation were performed to assess the interaction among transcription factors HIF1α, SP1, and EGR1 in the regulation of EPO-R under hypoxia. The expression of key EPO-R transcription factors in clinical specimens were determined by immunohistochemistry. RESULTS Hypoxia induced a dosage and time dependent EPO-R mRNA expression in NSCLC cells. Knockdown of EPO-R reduced NSCLC cell growth under hypoxia (P < 0.05). Mechanistically, a SP1-EGR1 overlapped DNA binding sequence was essential to the hypoxia induced EPO-R transcription. In the early phase of hypoxia, HIF1α interacted with EGR1 that negatively regulated EPO-R. With the exit of EGR1 in late phase, HIF1α positively regulated EPO-R expression through additive interaction with SP1. In clinical NSCLC specimen, SP1 was positively while EGR1 was negatively associated with active EPO-R expression (P < 0.05). CONCLUSIONS HIF1α, SP1 and EGR1 mediated EPO-R expression played an essential role in hypoxia-induced NSCLC cell proliferation. Our study presents a novel mechanism of EPO-R regulation in the tumor cells, which may provide information support for NSCLC diagnosis and treatment.
Collapse
Affiliation(s)
- Tianhong Su
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, 2 Hoblitzelle, Suite 252, 3500 Gaston Avenue, Dallas, TX, 75246, USA
- Department of Liver Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Pi Liu
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, 2 Hoblitzelle, Suite 252, 3500 Gaston Avenue, Dallas, TX, 75246, USA
- Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xinyu Ti
- Department of Respiratory Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Shouzhen Wu
- Shaanxi Institute of Pediatric Diseases, Xi'an Children's Hospital, Xi'an, 710003, China
| | - Xiaochang Xue
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
- Present Address: Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwestern China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Zenglu Wang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, China
| | - Elhardji Dioum
- Department of Pharmacology, Department of Internal Medicine, the University of Texas Southwestern Medical School, Dallas, TX, 75390, USA
- Present Address: Diabetes Department, Nestle Institute of Health Science, EPFL Campus, 1015, Lausanne, Switzerland
| | - Qiuyang Zhang
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, 2 Hoblitzelle, Suite 252, 3500 Gaston Avenue, Dallas, TX, 75246, USA.
| |
Collapse
|
11
|
Ventero MP, Fuentes-Baile M, Quereda C, Perez-Valeciano E, Alenda C, Garcia-Morales P, Esposito D, Dorado P, Manuel Barbera V, Saceda M. Radiotherapy resistance acquisition in Glioblastoma. Role of SOCS1 and SOCS3. PLoS One 2019; 14:e0212581. [PMID: 30811476 PMCID: PMC6392282 DOI: 10.1371/journal.pone.0212581] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/05/2019] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a poor prognosis type of tumour due to its resistance to chemo and radiotherapy. SOCS1 and SOCS3 have been associated with tumour progression and response to treatments in different kinds of cancers, including GBM. In this study, cell lines of IDH-wildtype GBM from primary cultures were obtained, and the role of SOCS1 and SOCS3 in the radiotherapy response was analysed. Fifty-two brain aspirates from GBM patients were processed, and six new cell lines of IDH-wildtype GBM were established. These new cell lines were characterized according to the WHO classification of CNS tumours. SOCS1 and SOCS3 expression levels were determined, at mRNA level by Q-PCR, at protein level by immunocytochemistry, and Western blot analysis. The results showed that SOCS1 and SOCS3 are overexpressed in GBM, as compared to a non-tumoral brain RNA pool. SOCS1 and SOCS3 expression were reduced by siRNA, and it was found that SOCS3 inhibition increases radioresistance in GBM cell lines, suggesting a key role of SOCS3 in radioresistant acquisition. In addition, radioresistant clonal populations obtained by selective pressure on these cell cultures also showed a significant decrease in SOCS3 expression, while SOCS1 remained unchanged. Furthermore, the induction of SOCS3 expression, under a heterologous promoter, in a radiotherapy resistant GBM cell line increased its radiosensitivity, supporting an important implication of SOCS3 in radiotherapy resistance acquisition. Finally, the treatment with TSA in the most radioresistant established cell line produced an increase in the effect of radiotherapy, that correlated with an increase in the expression of SOCS3. These effects of TSA disappeared if the increase in the expression of SOCS3 prevented with an siRNA against SOCS3. Thus, SOCS3 signal transduction pathway (JAK/STAT) could be useful to unmask new putative targets to improve radiotherapy response in GBM.
Collapse
Affiliation(s)
- Maria Paz Ventero
- Hospital General Universitario de Elche, FISABIO, Camí de l'Almazara, Elx (Alicante), Spain
| | - Maria Fuentes-Baile
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Hospital General Universitario de Alicante, C/ Maestro Alonso, Alicante (Alicante), Spain
| | - Cristina Quereda
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante (Alicante), Spain
| | - Elizabeth Perez-Valeciano
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, Elche (Alicante), Spain
| | - Cristina Alenda
- Instituto de Investigación Biomédica y Sanitaria de Alicante (ISABIAL), Hospital General Universitario de Alicante, C/ Maestro Alonso, Alicante (Alicante), Spain
| | - Pilar Garcia-Morales
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, Elche (Alicante), Spain
| | - Danilo Esposito
- Unidad de Oncología Radioterápica, ERESA, Hospital General Universitario de Elche, Camí de l'Almazara, Elx (Alicante), Spain
| | - Pilar Dorado
- Unidad de Oncología Radioterápica, ERESA, Hospital General Universitario de Elche, Camí de l'Almazara, Elx (Alicante), Spain
| | - Victor Manuel Barbera
- Hospital General Universitario de Elche, FISABIO, Camí de l'Almazara, Elx (Alicante), Spain
- Departamento de Fisiología, Genética y Microbiología, Facultad de Ciencias, Universidad de Alicante (Alicante), Spain
- * E-mail: (VMB); (MS)
| | - Miguel Saceda
- Hospital General Universitario de Elche, FISABIO, Camí de l'Almazara, Elx (Alicante), Spain
- Instituto de Biología Molecular y Celular, Ed. Torregaitan, Universidad Miguel Hernández, Elche (Alicante), Spain
- * E-mail: (VMB); (MS)
| |
Collapse
|
12
|
Cacabelos R, Carril JC, Sanmartín A, Cacabelos P. Pharmacoepigenetic Processors: Epigenetic Drugs, Drug Resistance, Toxicoepigenetics, and Nutriepigenetics. PHARMACOEPIGENETICS 2019:191-424. [DOI: 10.1016/b978-0-12-813939-4.00006-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Kobayashi M, Mikami D, Uwada J, Yazawa T, Kamiyama K, Kimura H, Taniguchi T, Iwano M. A short-chain fatty acid, propionate, enhances the cytotoxic effect of cisplatin by modulating GPR41 signaling pathways in HepG2 cells. Oncotarget 2018; 9:31342-31354. [PMID: 30140374 PMCID: PMC6101142 DOI: 10.18632/oncotarget.25809] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate are generated by microbial fermentation of indigestible fiber by gut flora. SCFAs are ligands of two orphan G protein-coupled receptors, GPR41 and GPR43, that modulate cell proliferation and induce apoptosis. However, it is unclear if SCFAs enhance the effects of chemotherapy in a GPR41- or GPR43-dependent manner. The aim of this study was to investigate whether SCFAs, and particularly propionate, activate GPR41 or GPR43, and thereby enhance the antitumor effects of cisplatin in HepG2 human hepatocellular carcinoma (HCC) cells. The inhibitory effects of propionate and cisplatin on proliferation of HCC cells were determined by MTS assay. Changes in apoptosis rate were analyzed by flow cytometry. The effects of combined propionate and cisplatin on these properties in HCC cells were significantly higher than those of cisplatin alone. With combined treatment, the levels of cleaved caspase-3, active caspase-3 forms, and acetylated histone H3 were enhanced in a GPR41-dependent manner; expression of histone deacetylases (HDAC) 3, 4, 5, 6, 8 proteins was significantly reduced; and induction of TNF-α expression was significantly enhanced. These results suggest that propionate and cisplatin synergistically and significantly induce apoptosis of HepG2 cells by increasing expression of autocrine TNF-α via reduction of HDACs through GPR41 signaling. From clinical and translational perspectives, our data suggest that a combination of propionate with cisplatin may have better therapeutic effects on HCC compared with conventional treatment, and that a selective GPR41 agonist may be a candidate as an adjuvant therapeutic agent for HCC.
Collapse
Affiliation(s)
- Mamiko Kobayashi
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Daisuke Mikami
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Junsuke Uwada
- Division of Cellular Signal Transduction, Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Takashi Yazawa
- Division of Cellular Signal Transduction, Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Kazuko Kamiyama
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hideki Kimura
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui, Japan
| | - Takanobu Taniguchi
- Division of Cellular Signal Transduction, Department of Biochemistry, Asahikawa Medical University, Asahikawa, Japan
| | - Masayuki Iwano
- Department of Nephrology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
14
|
Abstract
Lung cancer is the leading cause of cancer-related deaths in the world. Despite significant advances in the early detection and treatment of the disease, the prognosis remains poor, with an overall 5-year survival rate ranging from 15% to 20%. This poor prognosis results largely from early micrometastatic spread of cancer cells to nearby lymph nodes or tissues and partially from early recurrence after curative surgical resection. Recently, precision medicines that target potential oncogenic driver mutations have been approved to treat lung cancer. However, some lung cancer patients do not have targetable mutations, and many patients develop resistance to targeted therapy. Tumor heterogeneity and mutational density are also challenges in treating lung cancer, which underscores the need for developing alternative therapeutic strategies for treating lung cancer. Epigenetic therapy may circumvent the problems of tumor heterogeneity and drug resistance by affecting the expression of several hundred target genes. This review highlights precision medicine using an innovative approach of epigenetic priming prior to conventional standard therapy or targeted cancer therapy in lung cancer.
Collapse
Affiliation(s)
- Dongho Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Duk-Hwan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea. .,Samsung Medical Center, Research Institute for Future Medicine, Seoul, South Korea.
| |
Collapse
|
15
|
Schernberg A, Blanchard P, Chargari C, Deutsch E. Neutrophils, a candidate biomarker and target for radiation therapy? Acta Oncol 2017; 56:1522-1530. [PMID: 28835188 DOI: 10.1080/0284186x.2017.1348623] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neutrophils are the most abundant blood-circulating white blood cells, continuously generated in the bone marrow. Growing evidence suggests they regulate the innate and adaptive immune system during tumor evolution. This review will first summarize the recent findings on neutrophils as a key player in cancer evolution, then as a potential biomarker, and finally as therapeutic targets, with respective focuses on the interplay with radiation therapy. A complex interplay: Neutrophils have been associated with tumor progression through multiple pathways. Ionizing radiation has cytotoxic effects on cancer cells, but the sensitivity to radiation therapy in vivo differ from isolated cancer cells in vitro, partially due to the tumor microenvironment. Different microenvironmental states, whether baseline or induced, can modulate or even attenuate the effects of radiation, with consequences for therapeutic efficacy. Inflammatory biomarkers: Inflammation-based scores have been widely studied as prognostic biomarkers in cancer patients. We have performed a large retrospective cohort of patients undergoing radiation therapy (1233 patients), with robust relationship between baseline blood neutrophil count and 3-year's patient's overall survival in patients with different cancer histologies. (Pearson's correlation test: p = .001, r = -.93). Therapeutic approaches: Neutrophil-targeting agents are being developed for the treatment of inflammatory and autoimmune diseases. Neutrophils either can exert antitumoral (N1 phenotype) or protumoral (N2 phenotype) activity, depending on the Tumor Micro Environment. Tumor associated N2 neutrophils are characterized by high expression of CXCR4, VEGF, and gelatinase B/MMP9. TGF-β within the tumor microenvironment induces a population of TAN with a protumor N2 phenotype. TGF-β blockade slows tumor growth through activation of CD8 + T cells, macrophages, and tumor associated neutrophils with an antitumor N1 phenotype. CONCLUSIONS This supports the need for prospective neutrophils evaluation in clinical trials, making neutrophils a predictive biomarker with potential specific therapies.
Collapse
Affiliation(s)
- Antoine Schernberg
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Pierre Blanchard
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Cyrus Chargari
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM 1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine du Kremlin-Bicetre, Université Paris Sud, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Eric Deutsch
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM 1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine du Kremlin-Bicetre, Université Paris Sud, Université Paris Saclay, Le Kremlin-Bicetre, France
| |
Collapse
|