1
|
Embaby A, Huijberts SCFA, Wang L, Leite de Oliveira R, Rosing H, Nuijen B, Sanders J, Hofland I, van Steenis C, Kluin RJC, Lieftink C, Smith CG, Blank CU, van Thienen JV, Haanen JBAG, Steeghs N, Opdam FL, Beijnen JH, Huitema ADR, Bernards R, Schellens JHM, Wilgenhof S. A Proof-of-Concept Study of Sequential Treatment with the HDAC Inhibitor Vorinostat following BRAF and MEK Inhibitors in BRAFV600-Mutated Melanoma. Clin Cancer Res 2024; 30:3157-3166. [PMID: 38739109 DOI: 10.1158/1078-0432.ccr-23-3171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/15/2023] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE The development of resistance limits the clinical benefit of BRAF and MEK inhibitors (BRAFi/MEKi) in BRAFV600-mutated melanoma. It has been shown that short-term treatment (14 days) with vorinostat was able to initiate apoptosis of resistant tumor cells. We aimed to assess the antitumor activity of sequential treatment with vorinostat following BRAFi/MEKi in patients with BRAFV600-mutated melanoma who progressed after initial response to BRAFi/MEKi. PATIENTS AND METHODS Patients with BRAFi/MEKi-resistant BRAFV600-mutated melanoma were treated with vorinostat 360 mg once daily for 14 days followed by BRAFi/MEKi. The primary endpoint was an objective response rate of progressive lesions of at least 30% according to Response Evaluation Criteria in Solid Tumors 1.1. Secondary endpoints included progression-free survival, overall survival, safety, pharmacokinetics of vorinostat, and translational molecular analyses using ctDNA and tumor biopsies. RESULTS Of the 26 patients with progressive BRAFi/MEKi-resistant BRAFV600-mutated melanoma receiving treatment with vorinostat, 22 patients were evaluable for response. The objective response rate was 9%, with one complete response for 31.2 months and one partial response for 14.9 months. Median progression-free survival and overall survival were 1.4 and 5.4 months, respectively. Common adverse events were fatigue (23%) and nausea (19%). ctDNA analysis showed emerging secondary mutations in NRAS and MEK in eight patients at the time of BRAFi/MEKi resistance. Elimination of these mutations by vorinostat treatment was observed in three patients. CONCLUSIONS Intermittent treatment with vorinostat in patients with BRAFi/MEKi-resistant BRAFV600-mutated melanoma is well tolerated. Although the primary endpoint of this study was not met, durable antitumor responses were observed in a minority of patients (9%).
Collapse
Affiliation(s)
- Alaa Embaby
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sanne C F A Huijberts
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rodrigo Leite de Oliveira
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- CEMM, Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ingrid Hofland
- Core Facility Molecular Pathology & Biobanking, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Charlaine van Steenis
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Roelof J C Kluin
- Genomics Core Facility, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Christian U Blank
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johannes V van Thienen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Frans L Opdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jos H Beijnen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Rene Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | | | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Meng L, Chen X, Cheng K, Chen N, Zheng Z, Wang F, Sun H, Wong KC. TransPTM: a transformer-based model for non-histone acetylation site prediction. Brief Bioinform 2024; 25:bbae219. [PMID: 38725156 PMCID: PMC11082075 DOI: 10.1093/bib/bbae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Protein acetylation is one of the extensively studied post-translational modifications (PTMs) due to its significant roles across a myriad of biological processes. Although many computational tools for acetylation site identification have been developed, there is a lack of benchmark dataset and bespoke predictors for non-histone acetylation site prediction. To address these problems, we have contributed to both dataset creation and predictor benchmark in this study. First, we construct a non-histone acetylation site benchmark dataset, namely NHAC, which includes 11 subsets according to the sequence length ranging from 11 to 61 amino acids. There are totally 886 positive samples and 4707 negative samples for each sequence length. Secondly, we propose TransPTM, a transformer-based neural network model for non-histone acetylation site predication. During the data representation phase, per-residue contextualized embeddings are extracted using ProtT5 (an existing pre-trained protein language model). This is followed by the implementation of a graph neural network framework, which consists of three TransformerConv layers for feature extraction and a multilayer perceptron module for classification. The benchmark results reflect that TransPTM has the competitive performance for non-histone acetylation site prediction over three state-of-the-art tools. It improves our comprehension on the PTM mechanism and provides a theoretical basis for developing drug targets for diseases. Moreover, the created PTM datasets fills the gap in non-histone acetylation site datasets and is beneficial to the related communities. The related source code and data utilized by TransPTM are accessible at https://www.github.com/TransPTM/TransPTM.
Collapse
Affiliation(s)
- Lingkuan Meng
- Department of Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Xingjian Chen
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, MA 02138, United States
| | - Ke Cheng
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Nanjun Chen
- Department of Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Zetian Zheng
- Department of Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Fuzhou Wang
- Department of Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Hongyan Sun
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
3
|
Pu W, Chu X, Guo H, Huang G, Cui T, Huang B, Dai X, Zhang C. The activated ATM/AMPK/mTOR axis promotes autophagy in response to oxidative stress-mediated DNA damage co-induced by molybdenum and cadmium in duck testes. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120574. [PMID: 36351481 DOI: 10.1016/j.envpol.2022.120574] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 06/16/2023]
Abstract
Cadmium (Cd) and excess molybdenum (Mo) have multiple organ toxicity, and testis is one of their important target organs, but the reproductive toxicity of Mo and Cd combined treatment is still unclear. To explore the effects of Mo and Cd co-exposure on DNA damage and autophagy from the insight of ATM/AMPK/mTOR axis in duck testes, we randomly assigned 40 healthy 8-day-old ducks to control, Mo (100 mg/kg Mo), Cd (4 mg/kg Cd), and Mo + Cd groups for 16 weeks. Results found that Mo and/or Cd exposure caused trace elements imbalance, oxidative stress with a decrease in the activities of GSH-Px, CAT, T-SOD and GSH content, an increase in the concentrations of H2O2 and MDA and pathological damage. Additionally, Mo and/or Cd markedly raised DNA damage-related factors expression levels and 8-OHdG content, caused G1/S arrest followed by decreasing CDK2 and Cyclin E protein levels and increasing CDK1 and Cyclin B protein levels, and activated ATM/AMPK/mTOR axis by enhancing p-ATM/ATM, p-AMPK/AMPK and reducing p-mTOR/mTOR protein levels, eventually triggered autophagy by elevating LC3A, LC3B, Atg5, Beclin-1 mRNA levels and LC3II/LC3I, Beclin-1 protein levels and reducing P62, Dynein, mTOR mRNA levels and P62 protein level. Moreover, these changes were most apparent in the combined group. Altogether, the results reveal that autophagy caused by Mo and/or Cd may be associated with activating the DNA damage-mediated ATM/AMPK/mTOR axis in duck testes, and Mo and Cd co-exposure exacerbates these changes.
Collapse
Affiliation(s)
- Wenjing Pu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Xuesheng Chu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Huiling Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Gang Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Ting Cui
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Bingyan Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Xueyan Dai
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, PR China.
| |
Collapse
|
4
|
Targeting the Interplay between HDACs and DNA Damage Repair for Myeloma Therapy. Int J Mol Sci 2021; 22:ijms221910406. [PMID: 34638744 PMCID: PMC8508842 DOI: 10.3390/ijms221910406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a malignancy of terminally differentiated plasma cells, and accounts for 10% of all hematologic malignancies and 1% of all cancers. MM is characterized by genomic instability which results from DNA damage with certain genomic rearrangements being prognostic factors for the disease and patients’ clinical response. Following genotoxic stress, the evolutionary conserved DNA damage response (DDR) is activated and, in turn, coordinates DNA repair with cell-cycle events. However, the process of carcinogenesis cannot be attributed only to the genetic alterations, but also involves epigenetic processes. Regulation of expression and activity of key players in DNA repair and checkpoint proteins are essential and mediated partly by posttranslational modifications (PTM), such as acetylation. Crosstalk between different PTMs is important for regulation of DNA repair pathways. Acetylation, which is mediated by acetyltransferases (HAT) and histone deacetylases (HDAC), not only affects gene expression through its modulation of histone tails but also has recently been implicated in regulating non-histone proteins. Currently, several HDAC inhibitors (HDACi) have been developed both in pre-clinical and clinical studies, with some of them exhibiting significant anti-MM activities. Due to reversibility of epigenetic changes during the evolutionary process of myeloma genesis, the potency of epigenetic therapies seems to be of great importance. The aim of the present paper is the summary of all data on the role of HDACi in DDR, the interference with each DNA repair mechanism and the therapeutic implications of HDACi in MM.
Collapse
|
5
|
Shi Y, Wang Y, Jiang H, Sun X, Xu H, Wei X, Wei Y, Xiao G, Song Z, Zhou F. Mitochondrial dysfunction induces radioresistance in colorectal cancer by activating [Ca 2+] m-PDP1-PDH-histone acetylation retrograde signaling. Cell Death Dis 2021; 12:837. [PMID: 34489398 PMCID: PMC8421510 DOI: 10.1038/s41419-021-03984-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022]
Abstract
Mitochondrial retrograde signaling (mito-RTG) triggered by mitochondrial dysfunction plays a potential role in regulating tumor metabolic reprogramming and cellular sensitivity to radiation. Our previous studies showed phos-pyruvate dehydrogenase (p-PDH) and PDK1, which involved in aerobic glycolysis, were positively correlated with radioresistance, but how they initiate and work in the mito-RTG pathway is still unknown. Our further genomics analysis revealed that complex I components were widely downregulated in mitochondrial dysfunction model. In the present study, high expression of p-PDH was found in the complex I deficient cells and induced radioresistance. Mechanistically, complex I defects led to a decreased PDH both in cytoplasm and nucleus through [Ca2+]m-PDP1-PDH axis, and decreased PDH in nucleus promote DNA damage repair (DDR) response via reducing histone acetylation. Meanwhile, NDUFS1 (an important component of the complex I) overexpression could enhance the complex I activity, reverse glycolysis and resensitize cancer cells to radiation in vivo and in vitro. Furthermore, low NDUFS1 and PDH expression were validated to be correlated with poor tumor regression grading (TRG) in local advanced colorectal cancer (CRC) patients underwent neoadjuvant radiotherapy. Here, we propose that the [Ca2+]m-PDP1-PDH-histone acetylation retrograde signaling activated by mitochondrial complex I defects contribute to cancer cell radioresistance, which provides new insight in the understanding of the mito-RTG. For the first time, we reveal that NDUFS1 could be served as a promising predictor of radiosensitivity and modification of complex I function may improve clinical benefits of radiotherapy in CRC.
Collapse
Affiliation(s)
- Yingying Shi
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - You Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Huangang Jiang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Xuehua Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Hui Xu
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Xue Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Yan Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Guohui Xiao
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China
| | - Zhiyin Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, Hubei, 430071, China
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.
- Hubei clinical cancer study center, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
6
|
Saccharomyces cerevisiae DNA repair pathways involved in repair of lesions induced by mixed ternary mononuclear Cu(II) complexes based on valproic acid with 1,10-phenanthroline or 2,2'- bipyridine ligands. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2021; 868-869:503390. [PMID: 34454693 DOI: 10.1016/j.mrgentox.2021.503390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/25/2021] [Accepted: 08/07/2021] [Indexed: 11/21/2022]
Abstract
The sodium valproate has been largely used as an anti-epilepsy drug and, recently, as a putative drug in cancer therapy. However, the treatment with sodium valproate has some adverse effects. In this sense, more effective and secure complexes than sodium valproate should be explored in searching for new active drugs. This study aims to evaluate the cytotoxicity of sodium valproate, mixed ternary mononuclear Cu(II) complexes based on valproic acid (VA) with 1,10-phenanthroline (Phen) or 2,2'- bipyridine (Bipy) ligands - [Cu2(Valp)4], [Cu(Valp)2Phen] and [Cu(Valp)2Bipy] - in yeast Saccharomyces cerevisiae, proficient or deficient in different repair pathways, such as base excision repair (BER), nucleotide excision repair (NER), translesion synthesis (TLS), DNA postreplication repair (PRR), homologous recombination (HR) and non-homologous end-joining (NHEJ). The results indicated that the Cu(II) complexes have higher cytotoxicity than sodium valproate in the following order: [Cu(Valp)2Phen] > [Cu(Valp)2Bipy] > [Cu2(Valp)4] > sodium valproate. The treatment with Cu(II) complexes and sodium valproate induced mutations in S. cerevisiae. The data indicated that yeast strains deficient in BER (Ogg1p), NER (complex Rad1p-Rad10p) or TLS (Rev1p, Rev3p and Rad30p) proteins are associated with increased sensitivity to sodium valproate. The BER mutants (ogg1Δ, apn1Δ, rad27Δ, ntg1Δ and ntg2Δ) showed increased sensitivity to Cu(II) complexes. DNA damage induced by the complexes requires proteins from NER (Rad1p and Rad10p), TLS (Rev1p, Rev3p and Rad30p), PRR (Rad6 and Rad18p) and HR (Rad52p and Rad50p) for efficient repair. Therefore, Cu(II) complexes display enhanced cytotoxicity when compared to the sodium valproate and induce distinct DNA lesions, indicating a potential application as cytotoxic agents.
Collapse
|
7
|
Li Z, Yang L, Zhang S, Song J, Sun H, Shan C, Wang D, Liu S. Valproic acid Suppresses Breast Cancer Cell Growth Through Triggering Pyruvate Kinase M2 Isoform Mediated Warburg Effect. Cell Transplant 2021; 30:9636897211027524. [PMID: 34263655 PMCID: PMC8287349 DOI: 10.1177/09636897211027524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Energy metabolism programming is a hallmark of cancer, and serves as a potent
target of cancer therapy. Valproic acid (VPA), a broad Class I histone
deacetylases (HDACs) inhibitor, has been used as a therapeutic agent for cancer.
However, the detail mechanism about the potential role of VPA on the Warburg
effect in breast cancer remains unclear. In this study, we highlight that VPA
significantly attenuates the Warburg effect by decreasing the expression of
pyruvate kinase M2 isoform (PKM2), leading to inhibited cell proliferation and
reduced colony formation in breast cancer MCF-7 and MDA-MB-231 cells.
Mechanistically, Warburg effect suppression triggered by VPA was mediated by
inactivation of ERK1/2 phosphorylation through reduced HDAC1 expression,
resulting in suppressing breast cancer growth. In summary, we uncover a novel
mechanism of VPA in regulating the Warburg effect which is essential for
developing the effective approach in breast cancer therapy.
Collapse
Affiliation(s)
- Zhen Li
- Biomedical Translational Research Institute, 47855Jinan University, Guangzhou, Guangdong, China
| | - Lina Yang
- Department of Pathology, Medical School, 74547Dalian University, Dalian, Liaoning, China
| | - Shuai Zhang
- School of Integrative Medicine, 58301Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiaqi Song
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, 12538Nankai University, Tianjin, China
| | - Huanran Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, 12538Nankai University, Tianjin, China
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, 12538Nankai University, Tianjin, China
| | - Dan Wang
- Department of Pharmacology, 12396Yanbian University, Yanji, Jilin, China
| | - Shuangping Liu
- Department of Pathology, Medical School, 74547Dalian University, Dalian, Liaoning, China
| |
Collapse
|
8
|
Linciano P, Benedetti R, Pinzi L, Russo F, Chianese U, Sorbi C, Altucci L, Rastelli G, Brasili L, Franchini S. Investigation of the effect of different linker chemotypes on the inhibition of histone deacetylases (HDACs). Bioorg Chem 2020; 106:104462. [PMID: 33213894 DOI: 10.1016/j.bioorg.2020.104462] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
Abstract
Histone Deacetylases (HDACs) are among the most attractive and interesting targets in anticancer drug discovery. The clinical relevance of HDAC inhibitors (HDACIs) is testified by four FDA-approved drugs for cancer treatment. However, one of the main drawbacks of these drugs resides in the lack of selectivity against the different HDAC isoforms, resulting in severe side effects. Thus, the identification of selective HDACIs represents an exciting challenge for medicinal chemists. HDACIs are composed of a cap group, a linker region, and a metal-binding group interacting with the catalytic zinc ion. While the cap group has been extensively investigated, less information is available about the effect of the linker on isoform selectivity. To this aim, in this work, we explored novel linker chemotypes to direct isoform selectivity. A small library of 25 hydroxamic acids with hitherto unexplored linker chemotypes was prepared. In vitro tests demonstrated that, depending on the linker type, some candidates selectively inhibit HDAC1 over HDAC6 isoform or vice versa. Docking calculations were performed to rationalize the effect of the novel linker chemotypes on biologic activity. Moreover, four compounds were able to increase the levels of acetylation of histone H3 or tubulin. These compounds were also assayed in breast cancer MCF7 cells to test their antiproliferative effect. Three compounds showed a significant reduction of cancer proliferation, representing valuable starting points for further optimization.
Collapse
Affiliation(s)
- Pasquale Linciano
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Rosaria Benedetti
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", via L. De Crecchio 7, 80138 Napoli, Italy
| | - Luca Pinzi
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Fabiana Russo
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Ugo Chianese
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", via L. De Crecchio 7, 80138 Napoli, Italy
| | - Claudia Sorbi
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy.
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania "Luigi Vanvitelli", via L. De Crecchio 7, 80138 Napoli, Italy
| | - Giulio Rastelli
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Livio Brasili
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Silvia Franchini
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, via G. Campi 103, 41125 Modena, Italy.
| |
Collapse
|
9
|
Liu YC, Su CW, Ko PS, Lee RC, Liu CJ, Huang YH, Gau JP, Liu JH. A clinical trial with valproic acid and hydralazine in combination with gemcitabine and cisplatin followed by doxorubicin and dacarbazine for advanced hepatocellular carcinoma. Asia Pac J Clin Oncol 2020; 18:19-27. [PMID: 32964588 DOI: 10.1111/ajco.13443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Survival benefit from chemotherapy in advanced hepatocellular carcinoma (HCC) was limited till now. New chemoregimens with cytotoxicity modulators were explored to improve efficacy. Chemotherapy modulated with valproic acid (VA) as a deacetylation inhibitor of histone and DNA damage response proteins, and hydralazine (HZ) as a DNA hypomethylating agent, hypothetically suppressing DNA repair, were used in phase II trial here for advanced HCC. METHODS Between July 2008 and March 2016, patients with chemo-naive advanced HCC, regardless of previous sorafenib treatment, not amenable to local therapy and with Child Pugh score ≤7, were treated with VA (200 mg thrice per day) and HZ (12.5 mg twice per day) in conjunction with gemcitabine and cisplatin (GCGG): gemcitabine (1000 mg/m2 , D1; 800 mg/m2 D8, 15) and cisplatin (70 mg/m2 , D1) every 28 days till disease progression and then with Dox-DTIC: doxorubicin (45 mg/m2 ) and dacarbazine (450 mg/m2 ) every 28 days. The primary endpoint was overall survival (OS); the secondary endpoints were safety, progression-free survival (PFS) and response rate (RR). RESULTS Thirty-seven patients with 16 sorafenib-experienced, underwent GCGG treatment, and 30 of them underwent the following Dox-DTIC treatment. The median OS was 14.6 months (95% confidence interval: 6.0-23.1). The median PFSs for patients treated with VA- and HZ-combined GCGG and Dox-DTIC were 3.7 and 4.2 months, respectively; the RRs were 10/37 (27.0%) and 7/30 (23.3%); and grade 3/4 neutropenia were 54% and 51%. However, there were no chemotherapy-related deaths. CONCLUSION VA- and HZ-combined sequential chemotherapy was effective in advanced HCC with manageable toxicities.
Collapse
Affiliation(s)
- Yao-Chung Liu
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chien-Wei Su
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Shen Ko
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Rheun-Chuan Lee
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Jen Liu
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Hsiang Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jyh-Pyng Gau
- Division of Hematology and Oncology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jin-Hwang Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Hematology and Oncology, Cheng Hsin General Hospital, Taipei, Taiwan.,Chong Hin Loon Cancer and Biotherapy Research Center and Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
10
|
Sertic S, Quadri R, Lazzaro F, Muzi-Falconi M. EXO1: A tightly regulated nuclease. DNA Repair (Amst) 2020; 93:102929. [DOI: 10.1016/j.dnarep.2020.102929] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
11
|
Yang F, Zhu B, Liu J, Liu Y, Jiang C, Sheng Q, Qiu J, Nie Z. The effect of acetylation on the protein stability of BmApoLp-III in the silkworm, Bombyx mori. INSECT MOLECULAR BIOLOGY 2020; 29:104-111. [PMID: 31390480 DOI: 10.1111/imb.12613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/26/2019] [Accepted: 07/30/2019] [Indexed: 06/10/2023]
Abstract
Acetylation is an important, reversible posttranslational modification to a protein. In a previous study, we found that there were a large number of acetylated sites in various nutrient storage proteins of the silkworm haemolymph. In this study, we confirmed that acetylation can affect the stability of nutrient storage protein Bombyx mori apolipophorin-III (BmApoLp-III). First, the expression of BmApoLp-III could be upregulated when BmN cells were treated with the deacetylase inhibitor panobinostat (LBH589); similarly, the expression was downregulated when the cells were treated with the acetylase inhibitor C646. Furthermore, the increase in acetylation by LBH589 could inhibit the degradation and improve the accumulation of BmApoLp-III in BmN cells treated with cycloheximide and MG132 respectively. Moreover, we found that an increase in acetylation could decrease the ubiquitination of BmApoLp-III and vice versa; therefore, we predicted that acetylation could improve the stability of BmApoLp-III by competing for ubiquitination and inhibiting the protein degradation pathway mediated by ubiquitin. Additionally, BmApoLp-III had an antiapoptosis function that increased after LBH589 treatment, which might have been due to the improved protein stability after acetylation. These results have laid the foundation for further study on the mechanism of acetylation in regulating the storage and utilization of silkworm nutrition.
Collapse
Affiliation(s)
- F Yang
- College of Life Sciences and medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - B Zhu
- College of Life Sciences and medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - J Liu
- School of Forestry and Biotechnology, Zhejiang A&F University, Linan, China
| | - Y Liu
- Zhejiang Economic & Trade Polytechnic, Hangzhou, China
| | - C Jiang
- College of Life Sciences and medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Q Sheng
- College of Life Sciences and medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - J Qiu
- College of Life Sciences and medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Z Nie
- College of Life Sciences and medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
12
|
Lin T, Ren Q, Zuo W, Jia R, Xie L, Lin R, Zhao H, Chen J, Lei Y, Wang P, Dong H, Huang L, Cai J, Peng Y, Yu Z, Tan J, Wang S. Valproic acid exhibits anti-tumor activity selectively against EGFR/ErbB2/ErbB3-coexpressing pancreatic cancer via induction of ErbB family members-targeting microRNAs. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:150. [PMID: 30961642 PMCID: PMC6454766 DOI: 10.1186/s13046-019-1160-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/31/2019] [Indexed: 12/15/2022]
Abstract
Background Deregulated ErbB signaling plays an important role in tumorigenesis of pancreatic cancer. However, patients with pancreatic cancer benefit little from current existed therapies targeting the ErbB signaling. Here, we explore the potential anti-tumor activity of Valproic acid against pancreatic cancer via targeting ErbB family members. Methods Cell viability assay and apoptosis evaluation were carried out to determine the efficacy of VPA on pancreatic cancer cells. Western blot analyses were performed to determine the expression and activation of proteins. Apoptosis enzyme-linked immunosorbent assay was used to quantify cytoplasmic histone associated DNA fragments. Lentiviral expression system was used to introduce overexpression of exogeneous genes or gene-targeting short hairpin RNAs (shRNAs). qRT-PCR was carried out to analyze the mRNAs and miRNAs expression levels. Tumor xenograft model was established to evaluate the in vivo anti-pancreatic cancer activity of VPA. Results VPA preferentially inhibited cell proliferation/survival of, and induced apoptosis in EGFR/ErbB2/ErbB3-coexpressing pancreatic cancer cells within its clinically achievable range [40~100 mg/L (0.24~0.6 mmol/L)]. Mechanistic investigations revealed that VPA treatment resulted in simultaneous significant down-regulation of EGFR, ErbB2, and ErbB3 in pancreatic cancer cells likely via induction of ErbB family members-targeting microRNAs. Moreover, the anti-pancreatic cancer activity of VPA was further validated in tumor xenograft model. Conclusions Our data strongly suggest that VPA may be added to the treatment regimens for pancreatic cancer patients with co-overexpression of the ErbB family members. Electronic supplementary material The online version of this article (10.1186/s13046-019-1160-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tingting Lin
- Department of Urology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China. 156 Xi'er Huan Bei Road, Fuzhou, 350025, Fujian Province, China.,Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China.,Department of Medical Oncology, First Hospital of Sanming, Sanming, 365000, Fujian Province, China
| | - Qun Ren
- Department of Urology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China. 156 Xi'er Huan Bei Road, Fuzhou, 350025, Fujian Province, China.,Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Weimin Zuo
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China.,Department of Medical Ultrasound, Guangzhou First Peoples's Hospital, Guangzhou Medical University, Guangdong Province, Guangzhou, 510180, China
| | - Ruxue Jia
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Linhui Xie
- Department of Clinical Medicine, Fujian Health Vocational and Technical College, Fuzhou, 350101, Fujian Province, China
| | - Rong Lin
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Hu Zhao
- Department of Urology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China. 156 Xi'er Huan Bei Road, Fuzhou, 350025, Fujian Province, China.,Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Jin Chen
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Yan Lei
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Ping Wang
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Huiyue Dong
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Lianghu Huang
- Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Jinquan Cai
- Department of Urology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China. 156 Xi'er Huan Bei Road, Fuzhou, 350025, Fujian Province, China
| | - Yonghai Peng
- Department of Medical Oncology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou, 350025, Fujian Province, China
| | - Zongyang Yu
- Department of Medical Oncology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou, 350025, Fujian Province, China
| | - Jianming Tan
- Department of Urology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China. 156 Xi'er Huan Bei Road, Fuzhou, 350025, Fujian Province, China.,Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China
| | - Shuiliang Wang
- Department of Urology, The 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital), Fujian Medical University, Fuzhou 350025, China. 156 Xi'er Huan Bei Road, Fuzhou, 350025, Fujian Province, China. .,Fujian Key Laboratory of Transplant Biology, Affiliated Dongfang Hospital, Xiamen University School of Medicine, Fuzhou, 350025, Fujian Province, China.
| |
Collapse
|
13
|
Wang X, Shen C, Liu Z, Peng F, Chen X, Yang G, Zhang D, Yin Z, Ma J, Zheng Z, Zhao B, Liu H, Wang L, Wu J, Han D, Wang K, Zhong C, Hou X, Zhao W, Shu M, Wang X, Zhao S. Nitazoxanide, an antiprotozoal drug, inhibits late-stage autophagy and promotes ING1-induced cell cycle arrest in glioblastoma. Cell Death Dis 2018; 9:1032. [PMID: 30302016 PMCID: PMC6177448 DOI: 10.1038/s41419-018-1058-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/07/2018] [Accepted: 09/03/2018] [Indexed: 11/09/2022]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor in adults. New drug design and development is still a major challenge for glioma treatment. Increasing evidence has shown that nitazoxanide, an antiprotozoal drug, has a novel antitumor role in various tumors and exhibits multiple molecular functions, especially autophagic regulation. However, whether nitazoxanide-associated autophagy has an antineoplastic effect in glioma remains unclear. Here, we aimed to explore the underlying molecular mechanism of nitazoxanide in glioblastoma. Our results showed that nitazoxanide suppressed cell growth and induced cell cycle arrest in glioblastoma by upregulating ING1 expression with a favorable toxicity profile. Nitazoxanide inhibited autophagy through blockage of late-stage lysosome acidification, resulting in decreased cleavage of ING1. A combination with chloroquine or Torin1 enhanced or impaired the chemotherapeutic effect of nitazoxanide in glioblastoma cells. Taken together, these findings indicate that nitazoxanide as an autophagy inhibitor induces cell cycle arrest in glioblastoma via upregulated ING1 due to increased transcription and decreased post-translational degradation by late-stage autophagic inhibition.
Collapse
Affiliation(s)
- Xiaoxiong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Chen Shen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhendong Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Fei Peng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Xin Chen
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhiqin Yin
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Jichao Ma
- Department of Pharmacology, The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, College of Pharmacy of Harbin Medical University, No. 157 Baojian Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Zhixing Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Boxian Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Huailei Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Ligang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Jianing Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Dayong Han
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Kaikai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Chen Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Xu Hou
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Wenyang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Mengting Shu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Xinzhuang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China
| | - Shiguang Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China. .,Institute of Brain Science, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China. .,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, No. 23 Youzheng Street, Nangang District, 150001, Harbin, Heilongjiang Province, People's Republic of China.
| |
Collapse
|
14
|
Bressy C, Majhen D, Raddi N, Jdey W, Cornilleau G, Zig L, Guirouilh-Barbat J, Lopez BS, Bawa O, Opolon P, Grellier E, Benihoud K. Combined therapy of colon carcinomas with an oncolytic adenovirus and valproic acid. Oncotarget 2017; 8:97344-97360. [PMID: 29228615 PMCID: PMC5722567 DOI: 10.18632/oncotarget.22107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 08/04/2017] [Indexed: 12/27/2022] Open
Abstract
The anti-tumor potential of oncolytic adenoviruses (CRAds) has been demonstrated in preclinical and clinical studies. While these agents failed to eradicate tumors when used as a monotherapy, they may be more effective if combined with conventional treatments such as radiotherapy or chemotherapy. This study seeks to evaluate the combination of a CRAd bearing a ∆24 deletion in E1A with valproic acid (VPA), a histone deacetylase inhibitor, for the treatment of human colon carcinomas. This combination led to a strong inhibition of cell growth both in vitro and in vivo compared to treatment with CRAd or VPA alone. This effect did not stem from a better CRAd replication and production in the presence of VPA. Inhibition of cell proliferation and cell death were induced by the combined treatment. Moreover, whereas cells treated only with CRAd displayed a polyploidy (> 4N population), this phenotype was increased in cells treated with both CRAd and VPA. In addition, the increase in polyploidy triggered by combined treatment with CRAd and VPA was associated with the enhancement of H2AX phosphorylation (γH2AX), a hallmark of DNA damage, but also with a decrease of several DNA repair proteins. Finally, viral replication (or E1A expression) was shown to play a key role in the observed effects since no enhancement of polyploidy nor increase in γH2AX were found following cell treatment with a replication-deficient Ad and VPA. Taken together, our results suggest that CRAd and VPA could be used in combination for the treatment of colon carcinomas.
Collapse
Affiliation(s)
- Christian Bressy
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Dragomira Majhen
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Najat Raddi
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Wael Jdey
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Gaétan Cornilleau
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Léna Zig
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Josée Guirouilh-Barbat
- Laboratoire Recombinaison-Réparation et Cancer, UMR 8200 CNRS Stabilité Génétique et Oncogenèse, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Bernard S Lopez
- Laboratoire Recombinaison-Réparation et Cancer, UMR 8200 CNRS Stabilité Génétique et Oncogenèse, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Olivia Bawa
- Unité de pathologie expérimentale de l'IRCIV, Gustave Roussy, Villejuif 94805, France
| | - Paule Opolon
- Unité de pathologie expérimentale de l'IRCIV, Gustave Roussy, Villejuif 94805, France
| | - Elodie Grellier
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| | - Karim Benihoud
- Vectorologie et Thérapeutiques Anticancéreuses, UMR 8203 CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, Villejuif 94805, France
| |
Collapse
|
15
|
Zhang X, Xu R, Zhang C, Xu Y, Han M, Huang B, Chen A, Qiu C, Thorsen F, Prestegarden L, Bjerkvig R, Wang J, Li X. Trifluoperazine, a novel autophagy inhibitor, increases radiosensitivity in glioblastoma by impairing homologous recombination. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:118. [PMID: 28870216 PMCID: PMC5584019 DOI: 10.1186/s13046-017-0588-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/28/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND Resistance to adjuvant radiotherapy is a major cause of treatment failure in patients with glioblastoma (GBM). Autophagy inhibitors have been shown to enhance the efficacy of radiotherapy for certain solid tumors. However, current inhibitors do not penetrate the blood-brain-barrier (BBB). Here, we assessed the radiosensitivity effects of the antipsychotic drug trifluoperazine (TFP) on GBM in vitro and in vivo. METHODS U251 and U87 GBM cell lines as well as GBM cells from a primary human biopsy (P3), were used in vitro and in vivo to evaluate the efficacy of TFP treatment. Viability and cytotoxicity was evaluated by CCK-8 and clonogenic formation assays. Molecular studies using immunohistochemistry, western blots, immunofluorescence and qPCR were used to gain mechanistic insight into the biological activity of TFP. Preclinical therapeutic efficacy was evaluated in orthotopic xenograft mouse models. RESULTS IC50 values of U251, U87 and P3 cells treated with TFP were 16, 15 and 15.5 μM, respectively. TFP increased the expression of LC3B-II and p62, indicating a potential disruption of autophagy flux. These results were further substantiated by a decreased Lysotracker Red uptake, indicating impaired acidification of the lysosomes. We show that TFP and radiation had an additive effect when combined. This effect was in part due to impaired TFP-induced homologous recombination. Mechanistically we show that down-regulation of cathepsin L might explain the radiosensitivity effect of TFP. Finally, combining TFP and radiation resulted in a significant antitumor effect in orthotopic GBM xenograft models. CONCLUSIONS This study provides a strong rationale for further clinical studies exploring the combination therapy of TFP and radiation to treat GBM patients.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Ran Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Chao Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Yangyang Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China
| | - Chen Qiu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, People's Republic of China
| | - Frits Thorsen
- Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.,The Molecular Imaging Center, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway
| | - Lars Prestegarden
- Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.,Department of Dermatology, Haukeland University Hospital, 5009, Bergen, Norway
| | - Rolf Bjerkvig
- Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.,Department of Oncology, Luxembourg Institute of Health, L-1526, Strassen, Luxembourg
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China. .,Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Brain Science Research Institute, Shandong University, Jinan, 250012, People's Republic of China.
| |
Collapse
|
16
|
Yu Q, Liu L, Wang P, Yao Y, Xue Y, Liu Y. EMAP-II sensitize U87MG and glioma stem-like cells to temozolomide via induction of autophagy-mediated cell death and G2/M arrest. Cell Cycle 2017; 16:1085-1092. [PMID: 28436750 DOI: 10.1080/15384101.2017.1315492] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite the fact that temozolomide (TMZ) has been widely accepted as the key chemotherapeutic agent to prolong the survival of patients with glioblastoma, failure and recurrence cases can still be observed in clinics. Glioma stem-like cells (GSCs) are thought to be responsible for the drug resistance. In this study, we investigate whether endothelial monocyte-activating polypeptide-II (EMAP-II), a pro-inflammatory cytokine, can enhance TMZ cytotoxicity on U87MG and GSCs or not. As described in prior research, GSCs have been isolated from U87MG and maintained in the serum-free DMEM/F12 medium containing EGF, b-FGF, and B27. TMZ and/or EMAP-II administration were performed for 72 h, respectively. The results showed that TMZ combined with EMAP-II inhibit the proliferation of U87MG and GSCs by a larger measure than TMZ single treatment by decreasing the IC50. EMAP-II also enhanced TMZ-induced autophagy-mediated cell death and G2/M arrest. Moreover, we found that EMAP-II functioned a targeted suppression on mTOR, which may involve in the anti-neoplasm mechanism. The results suggest that EMAP-II could be considered as a combined chemotherapeutic agent against glioblastoma by sensitizing U87MG and GSCs to TMZ.
Collapse
Affiliation(s)
- Qi Yu
- a Department of Neurosurgery , Shengjing Hospital of China Medical University , Shenyang , China.,b Liaoning Research Center for Translational Medicine in Nervous System Disease , Shenyang China
| | - Libo Liu
- c Department of Neurobiology , College of Basic Medicine, China Medical University , Shenyang China.,d Key Laboratory of Cell Biology , Ministry of Public Health of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang China
| | - Ping Wang
- c Department of Neurobiology , College of Basic Medicine, China Medical University , Shenyang China.,d Key Laboratory of Cell Biology , Ministry of Public Health of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang China
| | - Yilong Yao
- a Department of Neurosurgery , Shengjing Hospital of China Medical University , Shenyang , China.,b Liaoning Research Center for Translational Medicine in Nervous System Disease , Shenyang China
| | - Yixue Xue
- c Department of Neurobiology , College of Basic Medicine, China Medical University , Shenyang China.,d Key Laboratory of Cell Biology , Ministry of Public Health of China, Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University , Shenyang China
| | - Yunhui Liu
- a Department of Neurosurgery , Shengjing Hospital of China Medical University , Shenyang , China.,b Liaoning Research Center for Translational Medicine in Nervous System Disease , Shenyang China
| |
Collapse
|
17
|
Ji MM, Wang L, Zhan Q, Xue W, Zhao Y, Zhao X, Xu PP, Shen Y, Liu H, Janin A, Cheng S, Zhao WL. Induction of autophagy by valproic acid enhanced lymphoma cell chemosensitivity through HDAC-independent and IP3-mediated PRKAA activation. Autophagy 2016; 11:2160-71. [PMID: 26735433 DOI: 10.1080/15548627.2015.1082024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autophagy is closely related to tumor cell sensitivity to anticancer drugs. The HDAC (histone deacetylase) inhibitor valproic acid (VPA) interacted synergistically with chemotherapeutic agents to trigger lymphoma cell autophagy, which resulted from activation of AMPK (AMP-activated protein kinase) and inhibition of downstream MTOR (mechanistic target of rapamycin [serine/threonine kinase]) signaling. In an HDAC-independent manner, VPA potentiated the effect of doxorubicin on lymphoma cell autophagy via reduction of cellular inositol 1,4,5 trisphosphate (IP3), blockade of calcium into mitochondria and modulation of PRKAA1/2-MTOR cascade. In murine xenograft models established with subcutaneous injection of lymphoma cells, dual treatment of VPA and doxorubicin initiated IP3-mediated calcium depletion and PRKAA1/2 activation, induced in situ autophagy and efficiently retarded tumor growth. Aberrant genes involving mitochondrial calcium transfer were frequently observed in primary tumors of lymphoma patients. Collectively, these findings suggested an HDAC-independent chemosensitizing activity of VPA and provided an insight into the clinical application of targeting autophagy in the treatment of lymphoma.
Collapse
Affiliation(s)
- Meng-Meng Ji
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Li Wang
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China.,b Pôle de Recherches Sino-Français en Science du Vivant et Génomique; Laboratory of Molecular Pathology ; Shanghai , China
| | - Qin Zhan
- b Pôle de Recherches Sino-Français en Science du Vivant et Génomique; Laboratory of Molecular Pathology ; Shanghai , China
| | - Wen Xue
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Yan Zhao
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Xia Zhao
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China.,b Pôle de Recherches Sino-Français en Science du Vivant et Génomique; Laboratory of Molecular Pathology ; Shanghai , China
| | - Peng-Peng Xu
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Yang Shen
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Han Liu
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Anne Janin
- b Pôle de Recherches Sino-Français en Science du Vivant et Génomique; Laboratory of Molecular Pathology ; Shanghai , China.,c U1165 Inserm/Université Paris 7; Hôpital Saint Louis ; Paris , France
| | - Shu Cheng
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China
| | - Wei-Li Zhao
- a State Key Laboratory of Medical Genomics; Shanghai Institute of Hematology; Shanghai Rui Jin Hospital; Shanghai Jiao Tong University School of Medicine ; Shanghai , China.,b Pôle de Recherches Sino-Français en Science du Vivant et Génomique; Laboratory of Molecular Pathology ; Shanghai , China
| |
Collapse
|
18
|
Sharma P. Biology and Management of Patients With Triple-Negative Breast Cancer. Oncologist 2016; 21:1050-62. [PMID: 27401886 PMCID: PMC5016071 DOI: 10.1634/theoncologist.2016-0067] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/17/2016] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED : Triple-negative breast cancer (TNBC) accounts for 15% of all breast cancers and is associated with poor long-term outcomes compared with other breast cancer subtypes. Because of the lack of approved targeted therapy, at present chemotherapy remains the mainstay of treatment for early and advanced disease. TNBC is enriched for germline BRCA mutation, providing a foundation for the use of this as a biomarker to identify patients suitable for treatment with DNA-damaging agents. Inherited and acquired defects in homologous recombination DNA repair, a phenotype termed "BRCAness," may be present in a large proportion of TNBC cases, making it an attractive selection and response biomarker for DNA-damaging therapy. Triple-negative breast cancer is a diverse entity for which additional subclassifications are needed. Increasing understanding of biologic heterogeneity of TNBC has provided insight into identifying potentially effective systemic therapies, including cytotoxic and targeted agents. Numerous experimental approaches are under way, and several encouraging drug classes, such as immune checkpoint inhibitors, poly(ADP-ribose) polymerase inhibitors, platinum agents, phosphatidylinositol-3-kinase pathway inhibitors, and androgen receptor inhibitors, are being investigated in TNBC. Molecular biomarker-based patient selection in early-phase trials has the potential to accelerate development of effective therapies for this aggressive breast cancer subtype. TNBC is a complex disease, and it is likely that several different targeted approaches will be needed to make meaningful strides in improving the outcomes. IMPLICATIONS FOR PRACTICE Triple-negative breast cancer (TNBC) is an aggressive subtype that is associated with poor outcomes. This article reviews clinical features and discusses the molecular diversity of this unique subtype. Current treatment paradigms, the role of germline testing, and platinum agents in TNBC are reviewed. Results and observations from pertinent clinical trials with potential implications for patient management are summarized. This article also discusses the clinical development and ongoing clinical trials of novel promising therapeutic agents in TNBC.
Collapse
|
19
|
Thotala D, Karvas RM, Engelbach JA, Garbow JR, Hallahan AN, DeWees TA, Laszlo A, Hallahan DE. Valproic acid enhances the efficacy of radiation therapy by protecting normal hippocampal neurons and sensitizing malignant glioblastoma cells. Oncotarget 2016; 6:35004-22. [PMID: 26413814 PMCID: PMC4741505 DOI: 10.18632/oncotarget.5253] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 09/04/2015] [Indexed: 12/18/2022] Open
Abstract
Neurocognitive deficits are serious sequelae that follow cranial irradiation used to treat patients with medulloblastoma and other brain neoplasms. Cranial irradiation causes apoptosis in the subgranular zone of the hippocampus leading to cognitive deficits. Valproic acid (VPA) treatment protected hippocampal neurons from radiation-induced damage in both cell culture and animal models. Radioprotection was observed in VPA-treated neuronal cells compared to cells treated with radiation alone. This protection is specific to normal neuronal cells and did not extend to cancer cells. In fact, VPA acted as a radiosensitizer in brain cancer cells. VPA treatment induced cell cycle arrest in cancer cells but not in normal neuronal cells. The level of anti-apoptotic protein Bcl-2 was increased and the pro-apoptotic protein Bax was reduced in VPA treated normal cells. VPA inhibited the activities of histone deacetylase (HDAC) and glycogen synthase kinase-3β (GSK3β), the latter of which is only inhibited in normal cells. The combination of VPA and radiation was most effective in inhibiting tumor growth in heterotopic brain tumor models. An intracranial orthotopic glioma tumor model was used to evaluate tumor growth by using dynamic contrast-enhanced magnetic resonance (DCE MRI) and mouse survival following treatment with VPA and radiation. VPA, in combination with radiation, significantly delayed tumor growth and improved mouse survival. Overall, VPA protects normal hippocampal neurons and not cancer cells from radiation-induced cytotoxicity both in vitro and in vivo. VPA treatment has the potential for attenuating neurocognitive deficits associated with cranial irradiation while enhancing the efficiency of glioma radiotherapy.
Collapse
Affiliation(s)
- Dinesh Thotala
- Department of Radiation Oncology, Washington University in St. Louis, Missouri, USA.,Siteman Cancer Center, Washington University in St. Louis, Missouri, USA
| | - Rowan M Karvas
- Department of Radiation Oncology, Washington University in St. Louis, Missouri, USA
| | - John A Engelbach
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, Missouri, USA
| | - Joel R Garbow
- School of Medicine, Washington University in St. Louis, Missouri, USA.,Mallinckrodt Institute of Radiology, Washington University in St. Louis, Missouri, USA.,Siteman Cancer Center, Washington University in St. Louis, Missouri, USA
| | - Andrew N Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, Missouri, USA
| | - Todd A DeWees
- Department of Radiation Oncology, Washington University in St. Louis, Missouri, USA
| | - Andrei Laszlo
- Department of Radiation Oncology, Washington University in St. Louis, Missouri, USA
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University in St. Louis, Missouri, USA.,Mallinckrodt Institute of Radiology, Washington University in St. Louis, Missouri, USA.,Siteman Cancer Center, Washington University in St. Louis, Missouri, USA.,Hope Center, Washington University in St. Louis, Missouri, USA
| |
Collapse
|
20
|
Makita N, Ninomiya I, Tsukada T, Okamoto K, Harada S, Nakanuma S, Sakai S, Makino I, Kinoshita J, Hayashi H, Oyama K, Nakagawara H, Miyashita T, Tajima H, Takamura H, Fushida S, Ohta T. Inhibitory effects of valproic acid in DNA double-strand break repair after irradiation in esophageal squamous carcinoma cells. Oncol Rep 2015; 34:1185-92. [PMID: 26135807 DOI: 10.3892/or.2015.4089] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/15/2015] [Indexed: 12/24/2022] Open
Abstract
Radiation therapy is one of the most promising therapeutic strategies in unresectable esophageal squamous cell carcinoma (ESCC). The histone deacetylase (HDAC) inhibitor has been shown to enhance radiosensitivity. Valproic acid (VPA) is a well-known drug used to treat seizure disorders and epilepsy, and has been shown to inhibit HDACs. We recently reported that a clinically safe dose of VPA enhances radiation‑induced cytotoxicity in human ESCC cells. However, the mechanism of radiosensitizing effect of VPA has not yet been confirmed. The present study examined the effect of VPA on DNA double-strand break (DSB) repair after radiation in the human ESCC cell lines KES, TE9 and TE11 by examining H2AX phosphorylation (γH2AX) levels as a marker of radiation‑induced DSBs. The present study also examined whether VPA inhibited radiation-induced DNA DSB repair by suppressing non-homologous end joining (NHEJ), focusing particularly on the acetylation of Ku70. VPA was shown to prolong γH2AX levels after irradiation in all three ESCC cell lines. Moreover, prolonged γH2AX foci formation after irradiation was also observed by immunocytochemistry following VPA pretreatment in KES and TE9 cells. VPA was shown to induce Ku70 acetylation after irradiation in all three ESCC cell lines. Our results suggest that VPA prolonged radiation‑induced DSBs by inhibiting NHEJ in DSB repair pathways in ESCC. VPA could therefore be used as an effective radiosensitizer in ESCC radiotherapy.
Collapse
Affiliation(s)
- Naoki Makita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tomoya Tsukada
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Koichi Okamoto
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Shinichi Harada
- Center for Biomedical Research and Education, School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Shinichi Nakanuma
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Seisho Sakai
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hironori Hayashi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Katsunobu Oyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hisatoshi Nakagawara
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa 920-8641, Japan
| |
Collapse
|
21
|
Zhang D, Tang B, Xie X, Xiao YF, Yang SM, Zhang JW. The interplay between DNA repair and autophagy in cancer therapy. Cancer Biol Ther 2015; 16:1005-13. [PMID: 25985143 DOI: 10.1080/15384047.2015.1046022] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
DNA is the prime target of anticancer treatments. DNA damage triggers a series of signaling cascades promoting cellular survival, including DNA repair, cell cycle arrest, and autophagy. The elevated basal and/or stressful levels of both DNA repair and autophagy observed in tumor cells, in contrast to normal cells, have been identified as the most important drug-responsive programs that impact the outcome of anticancer therapy. The exact relationship between DNA repair and autophagy in cancer cells remains unclear. On one hand, autophagy has been shown to regulate some of the DNA repair proteins after DNA damage by maintaining the balance between their synthesis, stabilization, and degradation. One the other hand, some evidence has demonstrated that some DNA repair molecular have a crucial role in the initiation of autophagy. In this review, we mainly discuss the interplay between DNA repair and autophagy in anticancer therapy and expect to enlighten some effective strategies for cancer treatment.
Collapse
Key Words
- AMPK, adenosine monophosphate-activated protein kinase
- ATG5, autophagy-related gene 5
- ATM, ataxia-telangiectasia mutated
- ATR, ATM and Rad3-related
- BER, base excision repair
- Chk1, check-point kinase 1
- Chk2, check-point kinase 2
- DDR, DNA damage response
- DNA damage
- DNA damage response
- DNA repair
- DNA-PKcs, DNA-dependent protein kinase catalytic subunit
- DSBs, double-strand breaks
- HDAC, histone deacetylases
- HR, homologous recombination
- IR, ionizing radiation
- MGMT, O6 methylguanine –DNA methyltransferase
- MMR, mismatch repair
- MRN, Mre11-Rad50-Nbs1
- NER, nucleotide excision recombination
- NHEJ, non-homologous end joining
- OGG1, 8-oxoguannine DNA glycosidase
- PARP-1, poly (ADP-ribose) polymerase 1
- PI3K, phosphoinositide 3-kinase
- PML, promyelocytic leukemia
- SSBs, single-strand break
- TMZ, temozolomide
- TSC2, tuberous sclerosis complex 2
- anticancer therapy
- apoptosis
- autophagy
- cell cycle arrest
- mTOR, mammalian target of rapamycin
- γ-H2AX, phosphorylated histone
Collapse
Affiliation(s)
- Dan Zhang
- a Department of Gastroenterology; Xinqiao Hospital; Third Military Medical University ; Chongqing , China
| | | | | | | | | | | |
Collapse
|
22
|
Thaler F, Mercurio C. Towards selective inhibition of histone deacetylase isoforms: what has been achieved, where we are and what will be next. ChemMedChem 2014; 9:523-6. [PMID: 24730063 DOI: 10.1002/cmdc.201300413] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Histone deacetylases (HDACs) are widely studied targets for the treatment of cancer and other diseases. Up to now, over twenty HDAC inhibitors have entered clinical studies and two of them have already reached the market, namely the hydroxamic acid derivative SAHA (vorinostat, Zolinza) and the cyclic depsipeptide FK228 (romidepsin, Istodax) that have been approved for the treatment of cutaneous T-cell lymphoma (CTCL). A common aspect of the first HDAC inhibitors is the absence of any particular selectivity towards specific isozymes. Some of molecules resulted to be “pan”-HDAC inhibitors, while others are class I selective. In the meantime, the knowledge of HDAC biology has continuously progressed. Key advances in the structural biology of various isozymes, reliable molecular homology models as well as suitable biological assays have provided new tools for drug discovery activities. This Minireview aims at surveying these recent developments as well as the design, synthesis and biological characterization of isoform-selective derivatives.
Collapse
|
23
|
Wang Y, Kuramitsu Y, Tokuda K, Baron B, Kitagawa T, Akada J, Maehara SI, Maehara Y, Nakamura K. Gemcitabine induces poly (ADP-ribose) polymerase-1 (PARP-1) degradation through autophagy in pancreatic cancer. PLoS One 2014; 9:e109076. [PMID: 25271986 PMCID: PMC4182782 DOI: 10.1371/journal.pone.0109076] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 09/08/2014] [Indexed: 01/02/2023] Open
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) and autophagy play increasingly important roles in DNA damage repair and cell death. Gemcitabine (GEM) remains the first-line chemotherapeutic drug for pancreatic cancer (PC). However, little is known about the relationship between PARP-1 expression and autophagy in response to GEM. Here we demonstrate that GEM induces DNA-damage response and degradation of mono-ADP ribosylated PARP-1 through the autophagy pathway in PC cells, which is rescued by inhibiting autophagy. Hypoxia and serum starvation inhibit autophagic activity due to abrogated GEM-induced mono-ADP-ribosylated PARP-1 degradation. Activation of extracellular regulated protein kinases (ERK) induced by serum starvation shows differences in intracellular localization as well as modulation of autophagy and PARP-1 degradation in GEM-sensitive KLM1 and -resistant KLM1-R cells. Our study has revealed a novel role of autophagy in PARP-1 degradation in response to GEM, and the different impacts of MEK/ERK signaling pathway on autophagy between GEM-sensitive and -resistant PC cells.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Yasuhiro Kuramitsu
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- * E-mail:
| | - Kazuhiro Tokuda
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Byron Baron
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Takao Kitagawa
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Junko Akada
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Shin-ichiro Maehara
- Department of Surgery and Science, Graduate School of Medical Science, Kyusyu University, Fukuokashi, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Science, Kyusyu University, Fukuokashi, Fukuoka, Japan
| | - Kazuyuki Nakamura
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
- Centre of Clinical Laboratories in Tokuyama Medical Association Hospital, Shunan, Japan
| |
Collapse
|
24
|
Pan DS, Yang QJ, Fu X, Shan S, Zhu JZ, Zhang K, Li ZB, Ning ZQ, Lu XP. Discovery of an orally active subtype-selective HDAC inhibitor, chidamide, as an epigenetic modulator for cancer treatment. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00350k] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tumorigenesis is maintained through a complex interplay of multiple cellular biological processes and is regulated to some extent by epigenetic control of gene expression.
Collapse
Affiliation(s)
- De-Si Pan
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Qian-Jiao Yang
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Xin Fu
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Song Shan
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Jing-Zhong Zhu
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Kun Zhang
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Zhi-Bin Li
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Zhi-Qiang Ning
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| | - Xian-Ping Lu
- Shenzhen Chipscreen Biosciences Ltd
- BIO-Incubator
- Shenzhen
- P. R. China
| |
Collapse
|
25
|
Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, Han W, Lou F, Yang J, Zhang Q, Wang X, He C, Pan H. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 2013; 4:e838. [PMID: 24113172 PMCID: PMC3824660 DOI: 10.1038/cddis.2013.350] [Citation(s) in RCA: 915] [Impact Index Per Article: 83.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/25/2013] [Accepted: 08/27/2013] [Indexed: 01/11/2023]
Abstract
Induction of cell death and inhibition of cell survival are the main principles of cancer therapy. Resistance to chemotherapeutic agents is a major problem in oncology, which limits the effectiveness of anticancer drugs. A variety of factors contribute to drug resistance, including host factors, specific genetic or epigenetic alterations in the cancer cells and so on. Although various mechanisms by which cancer cells become resistant to anticancer drugs in the microenvironment have been well elucidated, how to circumvent this resistance to improve anticancer efficacy remains to be defined. Autophagy, an important homeostatic cellular recycling mechanism, is now emerging as a crucial player in response to metabolic and therapeutic stresses, which attempts to maintain/restore metabolic homeostasis through the catabolic lysis of excessive or unnecessary proteins and injured or aged organelles. Recently, several studies have shown that autophagy constitutes a potential target for cancer therapy and the induction of autophagy in response to therapeutics can be viewed as having a prodeath or a prosurvival role, which contributes to the anticancer efficacy of these drugs as well as drug resistance. Thus, understanding the novel function of autophagy may allow us to develop a promising therapeutic strategy to enhance the effects of chemotherapy and improve clinical outcomes in the treatment of cancer patients.
Collapse
Affiliation(s)
- X Sui
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
HDACs (histone deacetylases) are a group of enzymes that deacetylate histones as well as non-histone proteins. They are known as modulators of gene transcription and are associated with proliferation and differentiation of a variety of cell types and the pathogenesis of some diseases. Recently, HDACs have come to be considered crucial targets in various diseases, including cancer, interstitial fibrosis, autoimmune and inflammatory diseases, and metabolic disorders. Pharmacological inhibitors of HDACs have been used or tested to treat those diseases. In the present review, we will examine the application of HDAC inhibitors in a variety of diseases with the focus on their effects of anti-cancer, fibrosis, anti-inflammatory, immunomodulatory activity and regulating metabolic disorders.
Collapse
|
27
|
Fortini P, Ferretti C, Dogliotti E. The response to DNA damage during differentiation: pathways and consequences. Mutat Res 2013; 743-744:160-168. [PMID: 23562804 DOI: 10.1016/j.mrfmmm.2013.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 02/17/2013] [Accepted: 03/12/2013] [Indexed: 11/25/2022]
Abstract
Damage to genomic DNA triggers a prompt set of signaling events known as the DNA damage response (DDR) which coordinates DNA repair, cell cycle arrest and ultimately cell death or senescence. Although activation of adequate DNA damage signaling and repair systems depends on the type of lesion and the cell-cycle phase in which it occurs, emerging evidence indicates that DNA repair and DDR function differently in different cellular contexts. Depending on the time maintenance and function of a specific cell type the risk of accumulating DNA damage may vary. For instance, damage to stem cells if not repaired can lead to mutation amplification or propagation through the processes of self-renewal and differentiation, respectively, whereas damage to post-mitotic cells can affect mostly tissue homeostasis. Stem cells are therefore expected to address DNA damage differently from their somatic counterparts. In this review the information available on the common and distinct mechanisms of control of genome integrity utilized by different cell types along the self-renewal/differentiation program will be reviewed, with special emphasis on their roles in the prevention of aging and disease.
Collapse
Affiliation(s)
- Paola Fortini
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Chiara Ferretti
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Eugenia Dogliotti
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
28
|
Martin FL. Epigenetic influences in the aetiology of cancers arising from breast and prostate: a hypothesised transgenerational evolution in chromatin accessibility. ISRN ONCOLOGY 2013; 2013:624794. [PMID: 23431470 PMCID: PMC3574745 DOI: 10.1155/2013/624794] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 12/26/2012] [Indexed: 01/12/2023]
Abstract
Epidemiological studies have consistently supported the notion that environmental and/or dietary factors play a central role in the aetiology of cancers of the breast and prostate. However, for more than five decades investigators have failed to identify a single cause-and-effect factor, which could be implicated; identification of a causative entity would allow the implementation of an intervention strategy in at-risk populations. This suggests a more complex pathoaetiology for these cancer sites, compared to others. When one examines the increases or decreases in incidence of specific cancers amongst migrant populations, it is notable that disease arising in colon or stomach requires one or at most two generations to exhibit a change in incidence to match that of high-incidence regions, whereas for breast or prostate cancer, at least three generations are required. This generational threshold could suggest a requirement for nonmutation-driven epigenetic alterations in the F0/F1 generations (parental/offspring adopting a more westernized lifestyle), which then predisposes the inherited genome of subsequent generations to mutagenic/genotoxic alterations leading to the development of sporadic cancer in these target sites. As such, individual susceptibility to carcinogen insult would not be based per se on polymorphisms in activating/detoxifying/repair enzymes, but on elevated accessibility of crucial target genes (e.g., oncogenes, tumour suppressor genes) or hotspots therein to mutation events. This could be termed a genomic susceptibility organizational structure (SOS). Several exposures including alcohol and heavy metals are epigens (i.e., modifiers of the epigenome), whereas others are mutagenic/genotoxic, for example, heterocyclic aromatic amines; humans are continuously and variously exposed to mixtures of these agents. Within such a transgenerational multistage model of cancer development, determining the interaction between epigenetic modification to generate a genomic SOS and genotoxic insult will facilitate a new level of understanding in the aetiology of cancer.
Collapse
Affiliation(s)
- Francis L. Martin
- Centre for Biophotonics, Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster LA1 4YQ, UK
| |
Collapse
|