1
|
Hu Q, Li Z, Li Y, Deng X, Chen Y, Ma X, Zeng J, Zhao Y. Natural products targeting signaling pathways associated with regulated cell death in gastric cancer: Recent advances and perspectives. Phytother Res 2023. [PMID: 37157181 DOI: 10.1002/ptr.7866] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Gastric cancer (GC) is one of the most serious gastrointestinal malignancies with high morbidity and mortality. The complexity of GC process lies in the multi-phenotypic linkage regulation, in which regulatory cell death (RCD) is the core link, which largely dominates the fate of GC cells and becomes a key determinant of GC development and prognosis. In recent years, increasing evidence has been reported that natural products can prevent and inhibit the development of GC by regulating RCDs, showing great therapeutic potential. In order to further clarify its key regulatory characteristics, this review focused on specific expressions of RCDs, combined with a variety of signaling pathways and their crosstalk characteristics, sorted out the key targets and action rules of natural products targeting RCD. It is highlighted that a variety of core biological pathways and core targets are involved in the decision of GC cell fate, including the PI3K/Akt signaling pathway, MAPK-related signaling pathways, p53 signaling pathway, ER stress, Caspase-8, gasdermin D (GSDMD), and so on. Moreover, natural products target the crosstalk of different RCDs by modulating above signaling pathways. Taken together, these findings suggest that targeting various RCDs in GC with natural products is a promising strategy, providing a reference for further clarifying the molecular mechanism of natural products treating GC, which warrants further investigations in this area.
Collapse
Affiliation(s)
- Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
2
|
Kong W, Wang Z, Wang B. Unveiling DNA damage repair-based molecular subtypes, tumor microenvironment and pharmacogenomic landscape in gastric cancer. Front Genet 2023; 14:1118889. [PMID: 37124627 PMCID: PMC10140566 DOI: 10.3389/fgene.2023.1118889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Objective: The current molecular classification system for gastric cancer covers genomic, molecular, and morphological characteristics. Non-etheless, classification of gastric cancer based upon DNA damage repair is still lacking. Here, we defined DNA damage repair-based subtypes across gastric cancer and identified clinicopathological, tumor microenvironment and pharmacogenomic features. Methods: Unsupervised clustering analysis was executed in the TCGA-STAD cohort based upon the transcriptional expression profiling of DNA damage repair genes. LASSO computational approach was adopted for generating a DNA damage repair-relevant gene signature. The identified subtypes or signature were externally verified in the GSE84426 or GSE84433 cohort. The transcriptional levels of immunomodulators, abundance of immune cells and somatic mutations were measured, respectively. Immunotherapeutic response, and drug sensitivity were investigated. The DNA damage repair-relevant genes were further experimentally verified. Results: Two DNA damage repair-based subtypes were identified, with the notable heterogeneity in prognostic stratification, tumor microenvironment and somatic mutations. The gene signature was generated for risk stratification and prognostic prediction, which was in relation to immunomodulators and immune cells. High-risk cases were more likely to respond to immunotherapy, with distinct pharmacogenomic landscapes between low- and high-risk groups. Higher levels of PAPPA2, MPO, MAGEA11, DEPP1, CPZ, and COLEC12 and lower level of CYTL1 were proven in gastric cancer cells versus controls. Silencing CYTL1 facilitated intracellular ROS accumulation and suppressed migration in gastric cancer cells. Conclusion: Collectively, the DNA damage repair-based classification is a suitable complement to existing molecular classification system, and the quantitative gene signature provides a robust tool in selecting specific therapeutic options.
Collapse
|
3
|
Zhang X, Wang J, Fan Y, Zhao Z, Paraghamian SE, Hawkins GM, Buckingham L, O'Donnell J, Hao T, Suo H, Yin Y, Sun W, Kong W, Sun D, Zhao L, Zhou C, Bae-Jump VL. Asparagus officinalis combined with paclitaxel exhibited synergistic anti-tumor activity in paclitaxel-sensitive and -resistant ovarian cancer cells. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04276-8. [PMID: 36006482 DOI: 10.1007/s00432-022-04276-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Although paclitaxel is a promising first-line chemotherapeutic drug for ovarian cancer, acquired resistance to paclitaxel is one of the leading causes of treatment failure, limiting its clinical application. Asparagus officinalis has been shown to have anti-tumorigenic effects on cell growth, apoptosis, cellular stress and invasion of various types of cancer cells and has also been shown to synergize with paclitaxel to inhibit cell proliferation in ovarian cancer. METHODS Human ovarian cancer cell lines MES and its PTX-resistant counterpart MES-TP cell lines were used and were treated with Asparagus officinalis and paclitaxel alone as well as in combination. Cell proliferation, cellular stress, invasion and DMA damage were investigated and the synergistic effect of a combined therapy analyzed. RESULTS In this study, we found that Asparagus officinalis combined with low-dose paclitaxel synergistically inhibited cell proliferation, induced cellular stress and apoptosis and reduced cell invasion in paclitaxel-sensitive and -resistant ovarian cancer cell lines. The combined treatment effects were dependent on DNA damage pathways and suppressing microtubule dynamics, and the AKT/mTOR pathway and microtubule-associated proteins regulated the inhibitory effect through different mechanisms in paclitaxel-sensitive and -resistant cells. CONCLUSION These findings suggest that the combination of Asparagus officinalis and paclitaxel have potential clinical implications for development as a novel ovarian cancer treatment strategy.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, 100026, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Jiandong Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, 100026, People's Republic of China
| | - Yali Fan
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, 100026, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Ziyi Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, 100026, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Sarah E Paraghamian
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Gabrielle M Hawkins
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Lindsey Buckingham
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Jillian O'Donnell
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Tianran Hao
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Hongyan Suo
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, 100026, People's Republic of China.,Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, 100026, People's Republic of China
| | - Delin Sun
- Shandong Juxinyuan Asparagus Industry Development Research Institute, HeZe, 274400, Shandong, People's Republic of China
| | - Luyu Zhao
- Shandong Juxinyuan Agricultural Technology Co. LTD, HeZe, 274400, Shandong, People's Republic of China
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA. .,Division of Gynecologic Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Dr, Chapel Hill, NC, 27599, USA.
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, 170 Manning Dr, Chapel Hill, NC, 27599, USA. .,Division of Gynecologic Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 450 West Dr, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
4
|
Abe Y, Hirano H, Shoji H, Tada A, Isoyama J, Kakudo A, Gunji D, Honda K, Boku N, Adachi J, Tomonaga T. Comprehensive characterization of the phosphoproteome of gastric cancer from endoscopic biopsy specimens. Theranostics 2020; 10:2115-2129. [PMID: 32089736 PMCID: PMC7019165 DOI: 10.7150/thno.37623] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 12/09/2019] [Indexed: 12/26/2022] Open
Abstract
Rationale: Cancer phosphoproteomics can provide insights regarding kinases that can be targeted for therapeutic applications. Monitoring the phosphoproteomics in cancer is expected to play a key role in optimizing treatments with kinase inhibitors. Clinical phosphoproteomics in surgical tissues and patient-derived models has been studied intensively. However, the reported data may not accurately reflect the phosphosignaling status in patients due to the effect of ischemia occurring during surgery or changes in the characteristics of cancer cells when establishing the models. In contrast, endoscopic biopsies have an advantage for clinical phosphoproteomics because they can be rapidly cryo-preserved. We aimed to develop a highly sensitive method for phosphoproteomics in endoscopic biopsies of gastric cancer. Methods: Three tumor biopsies and three normal gastric biopsies were obtained by endoscopy at one time, and subjected to our optimized phosphoproteomics. Phosphopeptides were enriched with an immobilized metal affinity chromatography, and labeled with Tandem Mass Tag reagent. Quantified phosphosites were compared between the pairs of tumor/normal biopsies within same patient. Cancer-specific activated pathways and kinases were identified by pathway enrichment analysis and kinase-substrate enrichment analysis. Results: Our protocol enabled the identification of more than 10,000 class 1 phosphosites from endoscopic biopsies. A comparison between samples from cancer tissue and normal mucosa demonstrated differences in the phosphosignaling, including biomarkers of response to DNA damage. Finally, cancer-specific activation of DNA damage response signaling was validated by additional phosphoproteomics of other patients and western blotting of gastric cancer/normal cells. Conclusion: In summary, our pioneering approach will facilitate more accurate clinical phosphoproteomics in endoscopic biopsies, which can be applied to monitor the activities of therapeutic kinases and, ultimately, can be a useful tool to precision medicine.
Collapse
|
5
|
Chen J, Tian B, Zhou C, Sun J, Lin L, Jin S, Liu Q, Fu S, Liu L, Liu H, Zhang Z, Li C, Wei H. A Novel Resveratrol-Arsenic Trioxide Combination Treatment Synergistically Induces Apoptosis of Adriamycin-Selected Drug-Resistant Leukemia K562 Cells. J Cancer 2019; 10:5483-5493. [PMID: 31632492 PMCID: PMC6775695 DOI: 10.7150/jca.34506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023] Open
Abstract
Leukemia cells can develop resistance to apoptosis induced by chemotherapeutic agents. Concomitant multidrug resistance of cells remains the greatest clinical obstacle in the effective treatment of blood and solid tumors. Natural products have been identified that possess the capacity to modulate chemotherapeutic resistance and induce apopotosis. In this study, we generated adriamycin-resistant K562 leukemia (K562/RA) cells and compared the responses of sensitive and resistant leukemia cells to the natural products arsenic trioxide (ATO) and resveratrol (Rsv), with a view to determining whether Rsv potentiates the sensitivity of drug-resistant cells to ATO-induced apoptosis and the associated molecular mechanisms. Our results showed that resistance of K562/RA cells induced by adriamycin treatment was significantly higher (115.81-fold) than that of parental K562 cells. Simultaneously, K562/RA cells were cross-resistant to multiple agents, with the exception of ATO. Rsv enhanced the sensitivity of K562/RA cells to ATO and reduced the required dose of ATO as well as associated adverse reactions by promoting the proliferation inhibitory and apoptosis-inducing effects of ATO, which may be associated with reduced expression of the drug resistance genes mdr1/P-gp, mrp1/MRP1 and bcrp/BCRP, as well as the apoptotic inhibitory genes bcl-2, NF-κB and P53, and conversely, activation of caspase-3. Our collective findings indicate that ATO and Rsv synergistically enhance the sensitivity of drug-resistant leukemia cells to apoptosis.
Collapse
Affiliation(s)
- Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Baoying Tian
- Hanzhong vocational and technical college, Hanzhong, Shanxi, 723000
| | - Cunmin Zhou
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Jingjing Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Li Lin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Shucheng Jin
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Quanrui Liu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Siyu Fu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Lian Liu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Hang Liu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Zhewen Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| | - Caili Li
- School of Medicine of Northwest University for Nationalities, Lanzhou, Gansu 730030, P.R. China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000
| |
Collapse
|
6
|
Narayan S, Ramisetti S, Jaiswal AS, Law BK, Singh-Pillay A, Singh P, Amin S, Sharma AK. ASR352, A potent anticancer agent: Synthesis, preliminary SAR, and biological activities against colorectal cancer bulk, 5-fluorouracil/oxaliplatin resistant and stem cells. Eur J Med Chem 2019; 161:456-467. [PMID: 30384048 PMCID: PMC7115410 DOI: 10.1016/j.ejmech.2018.10.052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/07/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
Despite new agent development and short-term benefits in patients with colorectal cancer (CRC), metastatic CRC cure rates have not improved due to high rates of 5-fluorouracil (5-FU)/leucovorin/oxaliplatin (FOLFOX)-resistance and a clinical therapeutic plateau. At the same time, this treatment regime leads to significant toxicity, cost, and patient inconvenience. Drug-resistance is linked to CRC stem cells, which are associated with the epidermal-to-mesenchymal transition (EMT) pathway. Thus, to optimally treat CRC, a therapy that can target the cell survival and EMT pathways in both CRC bulk and stem cell populations is critical. We recently identified a novel small molecule NSC30049 (7a) that is effective alone, and in combination potentiates 5-FU-mediated growth inhibition of CRC bulk, FOLFOX-resistant, and CRC stem cells both in vitro and in vivo models. In the present study, we report the synthesis and anti-CRC evaluation of several stable and effective 7a analogs. ASR352 (7b) was identified as one of the equipotent 7a analogs that inhibited the growth of CRC bulk cells, sensitized FOLFOX-resistant cells, and reduced the sphere formation capacity of CRC stem cells. It appears that the complex mechanism of cytotoxicity for 7b includes abrogation of 5-FU-induced the S phase, reduction of the phosphorylation of Chk1 at S317P, S345P and S296P, increased γH2AX staining, activation of caspase 3/PARP1 cleavage, and enhancement of Bax/Bcl2 ratio. Further 7b-mediated reduced phosphorylation of Chk1 was an indirect effect, since it did not inhibit Chk1 activity in an in vitro kinase assay. Our findings suggest that 7b as a single agent, or in combination with 5-FU can be developed as a therapeutic agent in CRC bulk, FOLFOX-resistant, and CRC stem cell populations for unmanageable metastatic CRC conditions.
Collapse
Affiliation(s)
- Satya Narayan
- Department of Anatomy and Cell Biology, University of Florida, Gainesville, FL, 32610, USA.
| | - Srinivasa Ramisetti
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA, 17033, USA
| | - Aruna S Jaiswal
- Department of Hematology and Oncology, University of Florida, Gainesville, FL, 32610, USA
| | - Brian K Law
- Department of Pharmacology and Experimental Therapeutics, University of Florida, Gainesville, FL, 32610, USA
| | - Ashona Singh-Pillay
- School of Chemistry and Physics, University of Kwa-Zulu Natal (UKZN), Westville Campus, Durban, 4000, South Africa
| | - Parvesh Singh
- School of Chemistry and Physics, University of Kwa-Zulu Natal (UKZN), Westville Campus, Durban, 4000, South Africa
| | - Shantu Amin
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA, 17033, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State University College of Medicine, Penn State Cancer Institute, Hershey, PA, 17033, USA.
| |
Collapse
|
7
|
Pajuelo-Lozano N, Bargiela-Iparraguirre J, Dominguez G, Quiroga AG, Perona R, Sanchez-Perez I. XPA, XPC, and XPD Modulate Sensitivity in Gastric Cisplatin Resistance Cancer Cells. Front Pharmacol 2018; 9:1197. [PMID: 30386247 PMCID: PMC6199368 DOI: 10.3389/fphar.2018.01197] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Cisplatin is an election drug widely used in clinic for the treatment of advanced gastric cancer. However, the heterogeneity of the gastric tumors and its resistance to the drugs, make in some cases the response very low and the prognosis unpredictable. In this manuscript we aim to find the molecular processes involved in cisplatin-induced apoptosis in two gastric cancer cell lines with different sensitivity to the treatment: AGS and MKN45. The apoptosis induction is higher in MKN45 than in AGS cells in response to CDDP. The intrinsic apoptotic pathway study revealed that MKN45 cells undergo degradation of Mcl-1 together with an increase of Bid and Bad levels, which results in sensitivity to CDDP. In addition, DNA repair NER pathway is impair in MKN45 cells due to low levels of XPC and the absence of translocation of XPA and XPD to the nucleus after stimuli. Altogether, these results suggest that NER and Bcl-2 protein family proteins are potential targets to improve the response to cisplatin treatment.
Collapse
Affiliation(s)
- Natalia Pajuelo-Lozano
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas de Madrid, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain
| | - Jone Bargiela-Iparraguirre
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas de Madrid, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain
| | - Gemma Dominguez
- Departamento de Medicina, Facultad de Medicina, Instituto de Investigaciones Biomédicas de Madrid, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain
| | - Adoracion G Quiroga
- Departamento de Quimica Inorganica, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rosario Perona
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain.,CIBER of Rare Diseases, Valencia, Spain
| | - Isabel Sanchez-Perez
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas de Madrid, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain.,Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid, Madrid, Spain.,CIBER of Rare Diseases, Valencia, Spain.,Unidad Asociada de Biomedicina, University of Castilla-La Mancha, Consejo Superior de Investigaciones Científicas, Albacete, Spain
| |
Collapse
|
8
|
Li Y, Zhang D, Yu K, Hu Y, Wu Q, Qian F, Wang Z. CMPD1 inhibited human gastric cancer cell proliferation by inducing apoptosis and G2/M cell cycle arrest. Biol Res 2018; 51:11. [PMID: 29661232 PMCID: PMC5901880 DOI: 10.1186/s40659-018-0159-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 04/10/2018] [Indexed: 01/02/2023] Open
Abstract
Background Gastric cancer occupies the fourth highest morbidity rate of cancers worldwide. Clinical therapies of gastric cancer remain limited because of uncertainty of mechanisms and shortness of effective medicine. Thus, new drug candidates for gastric cancer treatment is urgently needed. Results In this study, CMPD1 as a wildly used MK2 phosphorylation inhibitor was employed to find its impact on gastric cancer cell proliferation, apoptosis and cell cycle using colony formation assay and flow cytometry analysis. Along with its anti-proliferation effect on gastric cancer cell line MKN-45 and SGC7901, CMPD1 also induced massive apoptosis and significant G2/M phase arrest in a time-dependent and dose-dependent manner in MKN-45 cells respectively. Furthermore, Western blot confirmed that the expression of anti-apoptotic proteins Bcl-2 was decreased while BAX, cytochrome c release and cleaved PARP were increased. In addition, oncogene c-Myc was downregulated in response to CMPD1 treatment. Conclusions Our results demonstrated that CMPD1 has anti-tumor effect on human gastric cancer cell line MKN-45 possibly via downregulating oncogene c-Myc expression and CMPD1 could be applied as a potential candidate for treating gastric malignancy. To the best of our knowledge, it is the first report of anti-tumor effect of CMPD-1 on human gastric cancer cells.
Collapse
Affiliation(s)
- Yu Li
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Center for Cancer Precision Medicine, Bengbu Medical College, Bengbu, 233003, Anhui, People's Republic of China
| | - Depeng Zhang
- Engineering Research Center of Cell, & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Kaikai Yu
- Engineering Research Center of Cell, & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yudong Hu
- Engineering Research Center of Cell, & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qiong Wu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China.,Center for Cancer Precision Medicine, Bengbu Medical College, Bengbu, 233003, Anhui, People's Republic of China
| | - Feng Qian
- Center for Cancer Precision Medicine, Bengbu Medical College, Bengbu, 233003, Anhui, People's Republic of China. .,Engineering Research Center of Cell, & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - Zishu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, 233004, Anhui, People's Republic of China. .,Center for Cancer Precision Medicine, Bengbu Medical College, Bengbu, 233003, Anhui, People's Republic of China.
| |
Collapse
|
9
|
Echeverri M, Alvarez-Valdés A, Navas F, Perles J, Sánchez-Pérez I, Quiroga AG. Using phosphine ligands with a biological role to modulate reactivity in novel platinum complexes. ROYAL SOCIETY OPEN SCIENCE 2018; 5:171340. [PMID: 29515851 PMCID: PMC5830740 DOI: 10.1098/rsos.171340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/16/2018] [Indexed: 05/04/2023]
Abstract
Three platinum complexes with cis and trans configuration cis-[Pt(TCEP)2Cl2], cis-[Pt(tmTCEP)2Cl2] and trans-[Pt(TCEP)2Cl2], where TCEP is tris(2-carboxyethyl)phosphine, have been synthesized and fully characterized by usual techniques including single-crystal X-ray diffraction for trans-[Pt(TCEP)2Cl2] and cis-[Pt(tmTCEP)2Cl2]. Here, we also report on an esterification process of TCEP, which takes place in the presence of alcohols, leading to a platinum complex coordinated to an ester tmTCEP (2-methoxycarbonylethyl phosphine) ligand. The stability in solution of the three compounds and their interaction with biological models such as DNA (pBR322 and calf thymus DNA) and proteins (lysozyme and RNase) have also been studied.
Collapse
Affiliation(s)
- Marcelo Echeverri
- Inorganic Chemistry Department, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Francisco Navas
- Inorganic Chemistry Department, Universidad Autónoma de Madrid, Madrid, Spain
| | - Josefina Perles
- Single Crystal XRD Laboratory, SIdI, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - A. G. Quiroga
- Inorganic Chemistry Department, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
10
|
Arai H, Wada R, Ishino K, Kudo M, Uchida E, Naito Z. Expression of DNA damage response proteins in gastric cancer: Comprehensive protein profiling and histological analysis. Int J Oncol 2018; 52:978-988. [PMID: 29328366 DOI: 10.3892/ijo.2018.4238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/15/2017] [Indexed: 11/05/2022] Open
Abstract
Gastric cancer is the third major cause of cancer-related mortality in Japan. The aim of this study was to identify a factor implicated in the biology of gastric cancer by comprehensive protein profiling. Protein profiling was carried out by liquid chromatography-tandem mass spectrometry, using formalin-fixed paraffin-embedded specimens of 17 gastric cancer cases. Pathway analysis and orthogonal partial least square-discriminant analysis suggested the significant expression of ribonucleoproteins, heterogeneous nuclear ribonucleoproteins, interleukin binding factor 2 (ILF2), KU70 and KU80, which are involved in DNA damage response (DDR). Thus, the expression and phosphorylation levels of KU70, ILF2, CHK1 and CHK2 were examined by immunohistochemistry in 42 cases of gastric cancer. The expressions of ILF2 and CHK1 were unaffected in all cases. The expression and phosphorylation of CHK2 were absent in 2 cases. Despite the expression of proteins, the phosphorylation of KU70 and CHK2 appeared to be impaired in 1 and 4 cases, respectively. In 7 out of 42 cases (17%), DDR appeared to be impaired. Recurrence was noted in 2 out of these 7 cases (29%), whereas the recurrence was noted in 2 out of the remaining 35 cases (6%). The expression levels of KU70, ILF2, CHK1, CHK2 and TP53 were further examined in 4 gastric cancer cell lines. The expression and phosphorylation levels following exposure to ultraviolet radiation were abnormal in the 3 cell lines. The normal consecutive phosphorylation of CHK1 and CHK2, the upregulation of TP53 and an increase in apoptotic cell death following exposure to ultraviolet radiation was detected only in one cell line, suggesting that the preserved functions of DDR and TP53 are necessary for the determination of cell fate. It is thus suggested that DDR plays an important role in the pathobiology of gastric cancers.
Collapse
Affiliation(s)
- Hiroki Arai
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Ryuichi Wada
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Kousuke Ishino
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Mitsuhiro Kudo
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Eiji Uchida
- Department of Gastrointestinal Hepato-Biliary-Pancreatic Surgery, Nippon Medical School, Tokyo 113-8602, Japan
| | - Zenya Naito
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
11
|
Chen J, Wei H, Cheng J, Xie B, Wang B, Yi J, Tian B, Liu Z, Wang F, Zhang Z. Characteristics of doxorubicin-selected multidrug-resistant human leukemia HL-60 cells with tolerance to arsenic trioxide and contribution of leukemia stem cells. Oncol Lett 2017; 15:1255-1262. [PMID: 29399180 DOI: 10.3892/ol.2017.7353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 06/09/2017] [Indexed: 01/26/2023] Open
Abstract
The present study selected and characterized a multidrug-resistant HL-60 human acute promyelocytic leukemia cell line, HL-60/RS, by exposure to stepwise incremental doses of doxorubicin. The drug-resistant HL-60/RS cells exhibited 85.68-fold resistance to doxorubicin and were cross-resistant to other chemotherapeutics, including cisplatin, daunorubicin, cytarabine, vincristine and etoposide. The cells over-expressed the transporters P-glycoprotein, multidrug-resistance-related protein 1 and breast-cancer-resistance protein, encoded by the adenosine triphosphate-binding cassette (ABC)B1, ABCC1 and ABCG2 genes, respectively. Unlike other recognized chemoresistant leukemia cell lines, HL-60/RS cells were also strongly cross-resistant to arsenic trioxide. The proportion of leukemia stem cells (LSCs) increased synchronously with increased of drug resistance in the doxorubicin-induced HL-60 cell population. The present study confirmed that doxorubicin-induced HL-60 cells exhibited multidrug-resistance and high arsenic-trioxide resistance. Drug-resistance in these cells may be due to surviving chemoresistant LSCs in the HL-60 population, which have been subjected to long and consecutive selection by doxorubicin.
Collapse
Affiliation(s)
- Jing Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Jie Cheng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bei Xie
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Juan Yi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Baoying Tian
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhuan Liu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Feifei Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zhewen Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
12
|
Li Y, Zhou W, Tang K, Chen X, Feng Z, Chen J. Silencing Aurora A leads to re-sensitization of breast cancer cells to Taxol through downregulation of SRC-mediated ERK and mTOR pathways. Oncol Rep 2017; 38:2011-2022. [PMID: 28849180 PMCID: PMC5652950 DOI: 10.3892/or.2017.5908] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/31/2017] [Indexed: 12/11/2022] Open
Abstract
While Taxol has been reported to improve the clinical survival of breast cancer patients, subsequently developed drug-resistance of the cancer cells limits its final efficacy and applications. Previous studies suggested that Aurora A is involved in the development of the Taxol-resistance of breast cancer. We established Taxol-resistant breast cancer MCF-7/T cells and xenograft models to explore the role of Aurora A in Taxol resistant ER-positive breast cancer. Compared with their parental MCF-7/C cells, the Taxol-resistant MCF-7/T cells exhibited enhanced colony formation, less cell death and higher invasive ability. The resistant cells presented overexpressed Aurora A, elevated phosphorylated SRC and upregulated Ras/Raf/ERK and Akt/mTOR pathways. Silencing of Aurora A reduced the activity of SRC and downregulated the ERK and Akt/mTOR pathways, which led to re-sensitization of the resistant MCF-7/T cells to Taxol in vitro. These results suggested that the activation of Aurora A and the subsequent upregulation of ERK and Akt through SRC induced Taxol-resistance in breast cancer cells, and inhibiting Aurora A and the related SRC/EKT/Akt pathway could restore the sensitivity of breast cancer cells to Taxol. These results might shed light on the development of strategies to circumvent Taxol-related chemoresistance in breast cancer clinical practice.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Wanqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Ke Tang
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Nature Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Zhiqiang Feng
- Beijing Key Laboratory of Active Substance Discovery and Drugability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Jindong Chen
- Department of Urology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
13
|
Abstract
The development of drug resistance has largely limited the clinical outcome of anti-cancer treatment. Recent work has highlighted the involvement of non-coding RNAs, microRNAs (miRNAs), in cancer development. The present study aimed to investigate the role of miR-21 in the development of drug resistance to paclitaxel in gastric cancer cells. Our study found that the expression of miR-21 upregulated in the paclitaxel resistant cell line SGC7901/paclitaxel compared to its parental line SGC7901. Moreover, over-expression of miR-21 significantly decreased antiproliferative effects and apoptosis induced by paclitaxel, while knockdown of miR-21 dramatically increased antiproliferative effects and apoptosis induction by paclitaxel. Moreover, our results demonstrated that miR-21 may modulate the sensitivity to PTX, at least in part, by regulating the expression of P-glycoprotein.
Collapse
|
14
|
Shen J, Lu X, Du W, Zhou J, Qiu H, Chen J, Shen X, Zhong M. Lobetyol activate MAPK pathways associated with G 1 /S cell cycle arrest and apoptosis in MKN45 cells in vitro and in vivo. Biomed Pharmacother 2016; 81:120-127. [DOI: 10.1016/j.biopha.2016.03.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 03/29/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022] Open
|
15
|
CHK1 expression in Gastric Cancer is modulated by p53 and RB1/E2F1: implications in chemo/radiotherapy response. Sci Rep 2016; 6:21519. [PMID: 26867682 PMCID: PMC4751465 DOI: 10.1038/srep21519] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/26/2016] [Indexed: 12/19/2022] Open
Abstract
Radiation has a limited but relevant role in the adjuvant therapy of gastric cancer (GC) patients. Since Chk1 plays a critical function in cellular response to genotoxic agents, we aimed to analyze the role of Chk1 in GC as a biomarker for radiotherapy resistance. We analyzed Chk1 expression in AGS and MKN45 human GC cell lines by RT-QPCR and WB and in a small cohort of human patient’s samples. We demonstrated that Chk1 overexpression specifically increases resistance to radiation in GC cells. Accordingly, abrogation of Chk1 activity with UCN-01 and its expression with shChk1 increased sensitivity to bleomycin and radiation. Furthermore, when we assessed Chk1 expression in human samples, we found a correlation between nuclear Chk1 accumulation and a decrease in progression free survival. Moreover, using a luciferase assay we found that Chk1’s expression is controlled by p53 and RB/E2F1 at the transcriptional level. Additionally, we present preliminary data suggesting a posttranscriptional regulation mechanism, involving miR-195 and miR-503, which are inversely correlated with expression of Chk1 in radioresistant cells. In conclusion, Chk1/microRNA axis is involved in resistance to radiation in GC, and suggests Chk1 as a potential tool for optimal stratification of patients susceptible to receive adjuvant radiotherapy after surgery.
Collapse
|
16
|
Bargiela-Iparraguirre J, Prado-Marchal L, Pajuelo-Lozano N, Jiménez B, Perona R, Sánchez-Pérez I. Mad2 and BubR1 modulates tumourigenesis and paclitaxel response in MKN45 gastric cancer cells. Cell Cycle 2015; 13:3590-601. [PMID: 25483095 DOI: 10.4161/15384101.2014.962952] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Aneuploidy and chromosomal instability (CIN) are common features of gastric cancer (GC), but their contribution to carcinogenesis and antitumour therapy response is still poorly understood. Failures in the mitotic checkpoint induced by changes in expression levels of the spindle assembly checkpoint (SAC) proteins cause the missegregation of chromosomes in mitosis as well as aneuploidy. To evaluate the possible contribution of SAC to GC, we analyzed the expression levels of proteins of the mitotic checkpoint complex in a cohort of GC cell lines. We found that the central SAC proteins, Mad2 and BubR1, were the more prominently expressed members in disseminated GC cell lines. Silencing of Mad2 and BubR1 in MKN45 and ST2957 cells decreased their cell proliferation, migration and invasion abilities, indicating that Mad2 and BubR1 could contribute to cellular transformation and tumor progression in GC. We next evaluated whether silencing of SAC proteins could affect the response to microtubule poisons. We discovered that paclitaxel treatment increased cell survival in MKN45 cells interfered for Mad2 or BubR1 expression. However, apoptosis (assessed by caspase-3 activation, PARP proteolysis and levels of antiapoptotic Bcl 2-family members), the DNA damage response (assessed by H2Ax phosphorylation) and exit from mitosis (assessed by Cyclin B degradation and Cdk1 regulation) were activated equally between cells, independently of Mad2 or BubR1-protein levels. In contrast, we observed that the silencing of Mad2 or BubR1 in MKN45 cells showed the induction of a senescence-like phenotype accompanied by cell enlargement, increased senescence-associated β-galactosidase activity and increased IL-6 and IL-8 expression. In addition, the senescent phenotype is highly increased after treatment with PTX, indicating that senescence could prevent tumorigenesis in GC. In conclusion, the results presented here suggest that Mad2 and BubR1 could be used as prognostic markers of tumor progression and new pharmacological targets in the treatment for GC.
Collapse
Key Words
- BMC, bleomycin
- BubR1
- BubR1, budding uninhibited by benzimidazoles 1 homolog B protein (gene BUB1B)
- CDDP, cisplatin
- CIN, chromosome instability
- DDR, DNA damage response
- Mad2
- Mad2, mitotic arrest deficient-like-1 protein (gene Mad2L1)
- Monopolar Spindle kinase, MPS1
- PTX, paclitaxel
- SAC, spindle assembly checkpoint
- SASP, senescence associate secretory phenotype
- apoptosis
- gastric cancer
- mitosis
- paclitaxel
- senescence
- γH2AX, phosphorylated H2AX
Collapse
|
17
|
Valero ML, Cimas FJ, Arias L, Melgar-Rojas P, García E, Callejas-Valera JL, García-Cano J, Serrano-Oviedo L, Ángel de la Cruz-Morcillo M, Sánchez-Pérez I, Sánchez-Prieto R. E1a promotes c-Myc-dependent replicative stress: implications in glioblastoma radiosensitization. Cell Cycle 2013; 13:52-61. [PMID: 24196438 PMCID: PMC3925735 DOI: 10.4161/cc.26754] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/08/2013] [Accepted: 10/08/2013] [Indexed: 12/27/2022] Open
Abstract
The E1a gene from adenovirus is known to be a potent inducer of chemo/radiosensitivity in a wide range of tumors. However, the molecular bases of its radiosensitizer properties are still poorly understood. In an attempt to study this effect, U87MG cells, derived from a radio-resistant tumor as glioblastoma, where infected with lentivirus carrying E1a gene developing an acute sensitivity to ionizing radiation. The induction of radiosensitivity correlated with a marked G 2/M phase accumulation and a potent apoptotic response. Our findings demonstrate that c-Myc plays a pivotal role in E1a-associated radiosensitivity through the induction of a replicative stress situation, as our data support by genetic approaches, based in interference and overexpression in U87MG cells. In fact, we present evidence showing that Chk1 is a novel transcriptional target of E1a gene through the effect exerted by this adenoviral protein onto c-Myc. Moreover, c-Myc upregulation also explains the marked phosphorylation of H2AX associated to E1a expression in the absence of DNA damage. Indeed, all these observations were applicable to other experimental models, such as T98G, LN-405 and A172, rendering the same pattern in terms of radiosensitivity, cell cycle distribution, upregulation of Chk1, c-Myc, and phosphorylation pattern of H2AX. In summary, our data propose a novel mechanism to explain how E1a mediates radiosensitivity through the signaling axis E1a→c-Myc→ replicative stress situation. This novel mechanism of E1a-mediated radiosensitivity could be the key to open new possibilities in the current therapy of glioblastoma.
Collapse
Affiliation(s)
- María Llanos Valero
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Francisco Jose Cimas
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Laura Arias
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Pedro Melgar-Rojas
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Elena García
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Juan Luis Callejas-Valera
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Jesús García-Cano
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Leticia Serrano-Oviedo
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Miguel Ángel de la Cruz-Morcillo
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| | - Isabel Sánchez-Pérez
- Department of Biochemistry; School of Medicine;Biomedical Research Institute of Madrid CSIC/UAM; Madrid, Spain
| | - Ricardo Sánchez-Prieto
- Laboratorio de Oncología Molecular; Centro Regional de Investigaciones Biomédicas; Universidad de Castilla-La Mancha/PCyTA/ Unidad de Biomédicina UCLM-CSIC; Albacete, Spain
| |
Collapse
|
18
|
Lamore SD, Kamendi HW, Scott CW, Dragan YP, Peters MF. Cellular impedance assays for predictive preclinical drug screening of kinase inhibitor cardiovascular toxicity. Toxicol Sci 2013; 135:402-13. [PMID: 23897988 DOI: 10.1093/toxsci/kft167] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular (CV) toxicity is a leading contributor to drug attrition. Implementing earlier testing has successfully reduced human Ether-à-go-go-Related Gene-related arrhythmias. How- ever, analogous assays targeting functional CV effects remain elusive. Demand to address this gap is particularly acute for kinase inhibitors (KIs) that suffer frequent CV toxicity. The drug class also presents some particularly challenging requirements for assessing functional CV toxicity. Specifically, an assay must sense a downstream response that integrates diverse kinase signaling pathways. In addition, sufficient throughput is essential for handling inherent KI nonselectivity. A new opportunity has emerged with cellular impedance technology, which detects spontaneous beating cardiomyocytes. Impedance assays sense morphology changes downstream of cardiomyocyte contraction. To evaluate cardiomyocyte impedance assays for KI screening, we investigated two distinct KI classes where CV toxicity was discovered late and target risks remain unresolved. Microtubule-associated protein/microtubule affinity regulating kinase (MARK) inhibitors decrease blood pressure in dogs, whereas checkpoint kinase (Chk) inhibitors (AZD7762, SCH900776) exhibit dose-limiting CV toxicities in clinical trials. These in vivo effects manifested in vitro as cardiomyocyte beat cessation. MARK effects were deemed mechanism associated because beat inhibition potencies correlated with kinase inhibition, and gene knockdown and microtubule-targeting agents suppressed beating. MARK inhibitor impedance and kinase potencies aligned with rat blood pressure effects. Chk inhibitor effects were judged off-target because Chk and beat inhibition potencies did not correlate and knockdowns did not alter beating. Taken together, the data demonstrate that cardiomyocyte impedance assays can address three unmet needs-detecting KI functional cardiotoxicity in vitro, determining mechanism of action, and supporting safety structure-activity relationships.
Collapse
Affiliation(s)
- Sarah D Lamore
- Molecular Toxicology,Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Harriet W Kamendi
- Molecular Toxicology and Safety Pharmacology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Clay W Scott
- Molecular Toxicology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Yvonne P Dragan
- Molecular Toxicology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Matthew F Peters
- Molecular Toxicology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| |
Collapse
|