101
|
Chen L, Wang L, Shen H, Lin H, Li D. Anthelminthic drug niclosamide sensitizes the responsiveness of cervical cancer cells to paclitaxel via oxidative stress-mediated mTOR inhibition. Biochem Biophys Res Commun 2017; 484:416-421. [PMID: 28137584 DOI: 10.1016/j.bbrc.2017.01.140] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 01/25/2017] [Indexed: 01/07/2023]
Abstract
Drug repurposing represents an alternative therapeutic strategy to cancer treatment. The potent anti-cancer activities of a FDA-approved anthelminthic drug niclosamide have been demonstrated in various cancers. However, whether niclosamide is active against cervical cancer is unknown. In this study, we investigated the effects of niclosamide alone and its combination with paclitaxel in cervical cancer in vitro and in vivo. We found that niclosamide significantly inhibited proliferation and induced apoptosis of a panel of cervical cancer cell lines, regardless of their cellular origin and genetic pattern. Niclosamide also inhibited tumor growth in cervical cancer xenograft mouse model. Importantly, niclosamide significantly enhanced the responsiveness of cervical cancer cell to paclitaxel. We further found that niclosamide induced mitochondrial dysfunctions via inhibiting mitochondrial respiration, complex I activity and ATP generation, which led to oxidative stress. ROS scavenge agent N-acetyl-l-cysteine (NAC) completely reversed the effects of niclosamide in increasing cellular ROS, inhibiting proliferation and inducing apoptosis, suggesting that oxidative stress induction is the mechanism of action of niclosamide in cervical cancer cells. In addition, niclosamide significantly inhibited mammalian target of rapamycin (mTOR) signaling pathway in cervical cancer cells and its inhibitory effect on mTOR is modulated by oxidative stress. Our work suggests that niclosamide is a useful addition to the treatment armamentarium for cervical cancer and induction of oxidative stress may be a potential therapeutic strategy in cervical cancer.
Collapse
Affiliation(s)
- Liping Chen
- Department of Obstetrics and Gynecology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, China
| | - Haibin Shen
- Department of Obstetrics and Gynecology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, China
| | - Hui Lin
- Department of Obstetrics and Gynecology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, China.
| | - Dan Li
- Department of Obstetrics and Gynecology, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430033, China.
| |
Collapse
|
102
|
Wang C, Wu P, Shen XL, Wei XY, Jiang ZH. Synthesis, cytotoxic activity and drug combination study of tertiary amine derivatives of 2′,4′-dihydroxyl-6′-methoxyl-3′,5′-dimethylchalcone. RSC Adv 2017. [DOI: 10.1039/c7ra08639c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tertiary amine derivative of DMC (2b) exhibited broad spectrum of cytotoxicity and strong synergism with Taxol® against HeLa/Tax cells.
Collapse
Affiliation(s)
- Chen Wang
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization
- South China Botanical Garden
- Chinese Academy of Sciences
- Guangzhou 510650
- People's Republic of China
| | - Ping Wu
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization
- South China Botanical Garden
- Chinese Academy of Sciences
- Guangzhou 510650
- People's Republic of China
| | - Xiao-Ling Shen
- Laboratory of Herbal Drug Discovery
- Tropical Medicine Institute
- Guangzhou University of Chinese Medicine
- Guangzhou 510405
- People's Republic of China
| | - Xiao-Yi Wei
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization
- South China Botanical Garden
- Chinese Academy of Sciences
- Guangzhou 510650
- People's Republic of China
| | - Zi-Hua Jiang
- Department of Chemistry
- Lakehead University
- Thunder Bay
- Canada
| |
Collapse
|
103
|
Lin CS, Liu TC, Lee MT, Yang SF, Tsao TCY. Independent Prognostic Value of Hypoxia-inducible Factor 1-alpha Expression in Small Cell Lung Cancer. Int J Med Sci 2017; 14:785-790. [PMID: 28824314 PMCID: PMC5562133 DOI: 10.7150/ijms.19512] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/17/2017] [Indexed: 12/22/2022] Open
Abstract
Hypoxia is an important factor in tumor angiogenesis, metastasis, and resistance to chemotherapy or radiotherapy, and may be an indicator of poor prognosis. The transcription factor hypoxia-inducible factor 1 (HIF-1) is the key regulator of the hypoxic state. This study was designed to evaluate the prognostic value of HIF-1α expression in small cell lung cancer (SCLC). Forty-three paraffin-embedded biopsy materials were examined using immunohistochemistry. Our results indicated that the expression of HIF-1α was high in males, and patients with poor Eastern Cooperative Oncology Group (ECOG) performance status and metastases. To elucidate the prognostic value of HIF-1α expression, Kaplan-Meier analysis was carried out and the results showed that patients with high HIF-1α expression had a poorer prognosis than patients with low expression of HIF-1α. After adjusting clinical parameters by the Cox proportional hazards model, our results demonstrated that high HIF-1α expression is an independent prognostic factor for SCLC with a 39.2-fold risk of death (p<0.003). In conclusion, we have provided evidence that HIF-1α expression has significant value in predicting survival of patients with SCLC and is an independent prognostic factor beyond ECOG performance and metastasis status.
Collapse
Affiliation(s)
- Chang-Sheng Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Chest Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Tu-Chen Liu
- Department of Chest Medicine, Cheng-Ching General Hospital, Taichung, Taiwan
| | - Ming-Tsung Lee
- Research Assistant Center, Chang Hua Show Chwan Health Care System, Changhua, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Thomas Chang-Yao Tsao
- Division of Chest, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
104
|
Casasampere M, Ordóñez YF, Casas J, Fabrias G. Dihydroceramide desaturase inhibitors induce autophagy via dihydroceramide-dependent and independent mechanisms. Biochim Biophys Acta Gen Subj 2016; 1861:264-275. [PMID: 27894925 DOI: 10.1016/j.bbagen.2016.11.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/10/2016] [Accepted: 11/23/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Autophagy consists on the delivery of cytoplasmic material and organelles to lysosomes for degradation. Research on autophagy is a growing field because deciphering the basic mechanisms of autophagy is key to understanding its role in health and disease, and to paving the way to discovering novel therapeutic strategies. Studies with chemotherapeutic drugs and pharmacological tools support a role for dihydroceramides as mediators of autophagy. However, their effect on the autophagy outcome (cell survival or death) is more controversial. METHODS We have examined the capacity of structurally varied Des1 inhibitors to stimulate autophagy (LC3-II analysis), to increase dihydroceramides (mass spectrometry) and to reduce cell viability (SRB) in T98G and U87MG glioblastoma cells under different experimental conditions. RESULTS The compounds activity on autophagy induction took place concomitantly with accumulation of dihydroceramides, which occurred by both stimulation of ceramide synthesis de novo and reduction of Des1 activity. However, autophagy was also induced by the test compounds after preincubation with myriocin and in cells with a reduced capacity to produce dihydroceramides (U87DND). Autophagy inhibition with 3-methyladenine in the de novo dihydroceramide synthesis competent U87MG cells increased cytotoxicity, while genetic inhibition of autophagy in U87DND cells, poorly efficient at synthesizing dihydroceramides, augmented resistance to the test compounds. CONCLUSION Dihydroceramide desaturase 1 inhibitors activate autophagy via both dihydroceramide-dependent and independent pathways and the balance between the two pathways influences the final cell fate. GENERAL SIGNIFICANCE The cells capacity to biosynthesize dihydroceramides must be taken into account in proautophagic Des1 inhibitors-including therapies.
Collapse
Affiliation(s)
- Mireia Casasampere
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Yadira F Ordóñez
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Josefina Casas
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Gemma Fabrias
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain..
| |
Collapse
|
105
|
Dower CM, Bhat N, Wang EW, Wang HG. Selective Reversible Inhibition of Autophagy in Hypoxic Breast Cancer Cells Promotes Pulmonary Metastasis. Cancer Res 2016; 77:646-657. [PMID: 28115361 DOI: 10.1158/0008-5472.can-15-3458] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 11/16/2022]
Abstract
Autophagy influences how cancer cells respond to nutrient deprivation and hypoxic stress, two hallmarks of the tumor microenvironment (TME). In this study, we explored the impact of autophagy on the pathophysiology of breast cancer cells using a novel hypoxia-dependent, reversible dominant-negative strategy to regulate autophagy at the cellular level within the TME. Suppression of autophagy via hypoxia-induced expression of the kinase-dead unc-51-like autophagy-activating kinase (ULK1) mutant K46N increased lung metastases in MDA-MB-231 xenograft mouse models. Consistent with this effect, expressing a dominant-negative mutant of ULK1 or ATG4b or a ULK1-targeting shRNA facilitated cell migration in vitro Functional proteomic and transcriptome analysis revealed that loss of hypoxia-regulated autophagy promotes metastasis via induction of the fibronectin integrin signaling axis. Indeed, loss of ULK1 function increased fibronectin deposition in the hypoxic TME. Together, our results indicated that hypoxia-regulated autophagy suppresses metastasis in breast cancer by preventing tumor fibrosis. These results also suggest cautions in the development of autophagy-based strategies for cancer treatment. Cancer Res; 77(3); 646-57. ©2016 AACR.
Collapse
Affiliation(s)
- Christopher M Dower
- Department of Pediatrics, Milton Hershey Medical Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Neema Bhat
- Department of Pediatrics, Milton Hershey Medical Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Edward W Wang
- Department of Pediatrics, Milton Hershey Medical Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Hong-Gang Wang
- Department of Pediatrics, Milton Hershey Medical Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
106
|
Tang ZH, Zhang LL, Li T, Lu JH, Ma DL, Leung CH, Chen XP, Jiang HL, Wang YT, Lu JJ. Glycyrrhetinic acid induces cytoprotective autophagy via the inositol-requiring enzyme 1α-c-Jun N-terminal kinase cascade in non-small cell lung cancer cells. Oncotarget 2016; 6:43911-26. [PMID: 26549806 PMCID: PMC4791276 DOI: 10.18632/oncotarget.6084] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 10/23/2015] [Indexed: 01/07/2023] Open
Abstract
Glycerrhetinic acid (GA), one of the main bioactive constituents of Glycyrrhiza uralensis Fisch, exerts anti-cancer effects on various cancer cells. We confirmed that GA inhibited cell proliferation and induced apoptosis in non-small cell lung cancer A549 and NCI-H1299 cells. GA also induced expression of autophagy marker phosphatidylethanolamine-modified microtubule-associated protein light-chain 3 (LC3-II) and punta formation of green fluorescent protein microtubule-associated protein light-chain 3. We further proved that expression of GA-increased autophagy marker was attributed to activation instead of suppression of autophagic flux. The c-jun N-terminal kinase (JNK) pathway was activated after incubation with GA. Pretreatment with the JNK inhibitor SP600125 or silencing of the JNK pathway by siRNA of JNK or c-jun decreased GA-induced autophagy. The endoplasmic reticulum (ER) stress responses were also apparently stimulated by GA by triggering the inositol-requiring enzyme 1α (IRE1α) pathway. The GA-induced JNK pathway activation and autophagy were decreased by IRE1α knockdown, and inhibition of autophagy or the JNK cascade increased GA-stimulated IRE1α expression. In addition, GA-induced cell proliferative inhibition and apoptosis were increased by inhibition of autophagy or the JNK pathway. Our study was the first to demonstrate that GA induces cytoprotective autophagy in non-small cell lung cancer cells by activating the IRE1α-JNK/c-jun pathway. The combined treatment of autophagy inhibitors markedly enhances the anti-neoplasmic activity of GA. Such combination shows potential as a strategy for GA or GA-contained prescriptions in cancer therapy.
Collapse
Affiliation(s)
- Zheng-Hai Tang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Le-Le Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jia-Hong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiu-Ping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Yi-Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
107
|
Regina C, Panatta E, Candi E, Melino G, Amelio I, Balistreri CR, Annicchiarico-Petruzzelli M, Di Daniele N, Ruvolo G. Vascular ageing and endothelial cell senescence: Molecular mechanisms of physiology and diseases. Mech Ageing Dev 2016; 159:14-21. [DOI: 10.1016/j.mad.2016.05.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/12/2016] [Accepted: 05/03/2016] [Indexed: 01/21/2023]
|
108
|
He Z, Agostini M, Liu H, Melino G, Simon HU. p73 regulates basal and starvation-induced liver metabolism in vivo. Oncotarget 2016; 6:33178-90. [PMID: 26375672 PMCID: PMC4741757 DOI: 10.18632/oncotarget.5090] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/26/2015] [Indexed: 12/30/2022] Open
Abstract
As a member of the p53 gene family, p73 regulates cell cycle arrest, apoptosis, neurogenesis, immunity and inflammation. Recently, p73 has been shown to transcriptionally regulate selective metabolic enzymes, such as cytochrome c oxidase subunit IV isoform 1, glucose 6-phosphate dehydrogenase and glutaminase-2, resulting in significant effects on metabolism, including hepatocellular lipid metabolism, glutathione homeostasis and the pentose phosphate pathway. In order to further investigate the metabolic effect of p73, here, we compared the global metabolic profile of livers from p73 knockout and wild-type mice under both control and starvation conditions. Our results show that the depletion of all p73 isoforms cause altered lysine metabolism and glycolysis, distinct patterns for glutathione synthesis and Krebs cycle, as well as an elevated pentose phosphate pathway and abnormal lipid accumulation. These results indicate that p73 regulates basal and starvation-induced fuel metabolism in the liver, a finding that is likely to be highly relevant for metabolism-associated disorders, such as diabetes and cancer.
Collapse
Affiliation(s)
- Zhaoyue He
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Massimiliano Agostini
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - He Liu
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester, United Kingdom.,Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
109
|
Liu J, Pan C, Guo L, Wu M, Guo J, Peng S, Wu Q, Zuo Q. A new mechanism of trastuzumab resistance in gastric cancer: MACC1 promotes the Warburg effect via activation of the PI3K/AKT signaling pathway. J Hematol Oncol 2016; 9:76. [PMID: 27581375 PMCID: PMC5007850 DOI: 10.1186/s13045-016-0302-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/11/2016] [Indexed: 12/13/2022] Open
Abstract
Background Trastuzumab, a humanized antibody targeting HER2, exhibits remarkable therapeutic efficacy against HER2-positive gastric cancer. However, recurrent therapeutic resistance presents revolutionary claims. Warburg effect and AKT signaling pathway was involved in the resistance to trastuzumab. Our previous studies have demonstrated that overexpression of metastasis associated with the colon cancer 1 (MACC1) predicted poor prognosis of GC and promoted tumor cells proliferation and invasion. In this study, we found that MACC1 was significantly upregulated in trastuzumab-resistant cell lines. Besides, downregulation of MACC1 reversed this resistance. Methods The effect of trastuzumab and glycolysis inhibitor combination on cell viability, apoptosis, and cell metabolism was investigated in vitro using established trastuzumab-resistant GC cell lines. We assessed the impact of trastuzumab combined with oxamate on tumor growth and metabolism in an established xenograft model of HER2-positive GC cell lines. Results Here, we found that MACC1 was significantly upregulated in trastuzumab-resistant cell lines. Besides, downregulation of MACC1 in trastuzumab-resistant cells reversed this resistance. Overexpression of MACC1-induced trastuzumab resistance, enhanced the Warburg effect, and activated the PI3K/AKT signaling pathway, while downregulation of MACC1 presented the opposite effects. Moreover, when the PI3K/AKT signaling pathway was inhibited, the effects of MACC1 on resistance and glycolysis were diminished. Our findings indicated that MACC1 promoted the Warburg effect mainly through the PI3K/AKT signaling pathway, which further enhanced GC cells trastuzumab resistance. Conclusions Our results indicate that co-targeting of HER2 and the Warburg effect reversed trastuzumab resistance in vitro and in vivo, suggesting that the combination might overcome trastuzumab resistance in MACC1-overexpressed, HER2-positive GC patients. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0302-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Changqie Pan
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lihong Guo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Mengwan Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jing Guo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Sheng Peng
- Department of ICU, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qianying Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qiang Zuo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
110
|
Kuwahara Y, Roudkenar MH, Suzuki M, Urushihara Y, Fukumoto M, Saito Y, Fukumoto M. The Involvement of Mitochondrial Membrane Potential in Cross-Resistance Between Radiation and Docetaxel. Int J Radiat Oncol Biol Phys 2016; 96:556-65. [PMID: 27681752 DOI: 10.1016/j.ijrobp.2016.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 01/18/2023]
Abstract
PURPOSE To understand the molecular mechanisms underlying cancer cell radioresistance, clinically relevant radioresistant (CRR) cells that continue to proliferate during exposure to 2 Gy/day X-rays for more than 30 days were established. A modified high-density survival assay for anticancer drug screening revealed that CRR cells were resistant to an antimicrotubule agent, docetaxel (DTX). The involvement of reactive oxygen species (ROS) from mitochondria (mtROS) in the cross-resistance to X-rays and DTX was studied. METHODS AND MATERIALS Sensitivity to anticancer agents was determined by a modified high-density cell survival or water-soluble tetrazolium salt assay. DTX-induced mtROS generation was determined by MitoSOX red staining. JC-1 staining was used to visualize mitochondrial membrane potential. DTX-induced DNA double-strand breaks were determined by γ-H2AX staining. To obtain mitochondrial DNA-lacking (ρ(0)) cells, the cells were cultured for 3 to 4 weeks in medium containing ethidium bromide. RESULTS Treatment with DTX increased mtROS in parental cells but not in CRR cells. DTX induced DNA double-strand breaks in parental cells. The mitochondrial membrane potential of CRR cells was lower in CRR cells than in parental cells. Depletion of mtDNA induced DTX resistance in parental cells. Treatment with dimethyl sulfoxide also induced DTX resistance in parental cells. CONCLUSIONS The mitochondrial dysfunction observed in CRR cells contributes to X-ray and DTX cross-resistance. The activation of oxidative phosphorylation in CRR cells may represent an effective approach to overcome radioresistant cancers. In general, the overexpression of β-tubulin or multidrug efflux pumps is thought to be involved in DTX resistance. In the present study, we discovered another DTX resistant mechanism by investigating CRR cells.
Collapse
Affiliation(s)
- Yoshikazu Kuwahara
- Department of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan; Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Mehryar Habibi Roudkenar
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Masatoshi Suzuki
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yusuke Urushihara
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Motoi Fukumoto
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yohei Saito
- Department of Radiopharmacy, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Manabu Fukumoto
- Department of Pathology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan; Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
111
|
Inhibition of JNK-mediated autophagy enhances NSCLC cell sensitivity to mTORC1/2 inhibitors. Sci Rep 2016; 6:28945. [PMID: 27358039 PMCID: PMC4928093 DOI: 10.1038/srep28945] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
As the activation of autophagy contributes to the efficacy of many anticancer therapies, deciphering the precise role of autophagy in cancer therapy is critical. Here, we report that the dual mTORC1/2 inhibitors PP242 and OSI-027 decreased cell viability but did not induce apoptosis in the non-small cell lung cancer (NSCLC) cell lines H460 and A549. PP242 induced autophagy in NSCLC cells as demonstrated by the formation of massive vacuoles and acidic vesicular organelles and the accumulation of LC3-II. JNK was activated by PP242, and PP242-induced autophagy was blocked by inhibiting JNK pathway with SP600125 or JNK siRNA, suggesting that JNK activation is required for the mTORC1/2 inhibitor-mediated induction of autophagy in NSCLC cells. Inhibiting JNK or autophagy increased the sensitivity of H460 cells to mTORC1/2 inhibitors, indicating that JNK or autophagy promoted survival in NSCLC cells treated with mTORC1/2 inhibitors. Together, these data suggest that combining mTORC1/2 inhibitors with inhibitors of JNK or autophagy might be an effective approach for improving therapeutic outcomes in NSCLC.
Collapse
|
112
|
Nakayama Y, Inoue T. Antiproliferative Fate of the Tetraploid Formed after Mitotic Slippage and Its Promotion; A Novel Target for Cancer Therapy Based on Microtubule Poisons. Molecules 2016; 21:molecules21050663. [PMID: 27213315 PMCID: PMC6274067 DOI: 10.3390/molecules21050663] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/20/2022] Open
Abstract
Microtubule poisons inhibit spindle function, leading to activation of spindle assembly checkpoint (SAC) and mitotic arrest. Cell death occurring in prolonged mitosis is the first target of microtubule poisons in cancer therapies. However, even in the presence of microtubule poisons, SAC and mitotic arrest are not permanent, and the surviving cells exit the mitosis without cytokinesis (mitotic slippage), becoming tetraploid. Another target of microtubule poisons-based cancer therapy is antiproliferative fate after mitotic slippage. The ultimate goal of both the microtubule poisons-based cancer therapies involves the induction of a mechanism defined as mitotic catastrophe, which is a bona fide intrinsic oncosuppressive mechanism that senses mitotic failure and responds by driving a cell to an irreversible antiproliferative fate of death or senescence. This mechanism of antiproliferative fate after mitotic slippage is not as well understood. We provide an overview of mitotic catastrophe, and explain new insights underscoring a causal association between basal autophagy levels and antiproliferative fate after mitotic slippage, and propose possible improved strategies. Additionally, we discuss nuclear alterations characterizing the mitotic catastrophe (micronuclei, multinuclei) after mitotic slippage, and a possible new type of nuclear alteration (clustered micronuclei).
Collapse
Affiliation(s)
- Yuji Nakayama
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| | - Toshiaki Inoue
- Division of Human Genome Science, Department of Molecular and Cellular Biology, School of Life Sciences, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan.
| |
Collapse
|
113
|
Periplogenin induces necroptotic cell death through oxidative stress in HaCaT cells and ameliorates skin lesions in the TPA- and IMQ-induced psoriasis-like mouse models. Biochem Pharmacol 2016; 105:66-79. [DOI: 10.1016/j.bcp.2016.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022]
|
114
|
Tan S, Shi H, Ba M, Lin S, Tang H, Zeng X, Zhang X. miR-409-3p sensitizes colon cancer cells to oxaliplatin by inhibiting Beclin-1-mediated autophagy. Int J Mol Med 2016; 37:1030-8. [PMID: 26935807 DOI: 10.3892/ijmm.2016.2492] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 01/19/2016] [Indexed: 11/06/2022] Open
Abstract
The chemoresistance of colon cancer cells limits the efficacy of chemotherapy. miR-409-3p has been shown to be downregulated in various types of cancer. In the present study, we examined the role of miR-409-3p in colon cancer as well as the effects of miR‑409-3p on the sensitivity of colon cancer cells to oxaliplatin. The expression of miR-409 was significantly downregulated in the human colon cancer cell lines compared with the normal colon epithelial cells. Importantly, the miR-409-3p expression levels were lower in human colon cancer patient samples than in normal colon tissues. Moreover, we observed a negative correlation between the miR‑409-3p levels and resistance to oxaliplatin: the oxaliplatin-resistant colon cancer cells exhibited significantly downregulated miR‑409-3p levels, but higher autophagic activity than the oxaliplatin-sensitive cells. Using bioinformatics analysis, we predicted that miR‑409-3p miRNA binds to the key autophagy gene encoding Beclin-1. Our findings indicated that the overexpression of miR‑409-3p inhibited Beclin-1 expression and autophagic activity by binding to the 3'-untranslated region of Beclin-1 mRNA. In addition, the overexpression of miR‑409-3p enhanced the chemosensitivity of the oxaliplatin-sensitive and oxaliplatin-resistant colon cancer cells. The restoration of Beclin-1 abrogated these effects of miR‑409-3p. In a xenograft model using nude mice, we examined the effects of miR‑409-3p on tumor growth during chemotherapy. miR‑409-3p overexpression sensitized the tumor to chemotherapy, while inhibiting chemotherapy-induced autophagy in a manner dependent on Beclin-1. The findings of our study suggest that miR-409-3p is capable of enhancing the chemosensitivity of colon cancer cells by inhibiting Beclin-1-mediated autophagy.
Collapse
Affiliation(s)
- Shifan Tan
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Huijuan Shi
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Mingchen Ba
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Shengqv Lin
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Hongsheng Tang
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Xiaoqi Zeng
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Xiangliang Zhang
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| |
Collapse
|
115
|
Fan Z, Cui H, Yu H, Ji Q, Kang L, Han B, Wang J, Dong Q, Li Y, Yan Z, Yan X, Zhang X, Lin Z, Hu Y, Jiao S. MiR-125a promotes paclitaxel sensitivity in cervical cancer through altering STAT3 expression. Oncogenesis 2016; 5:e197. [PMID: 26878391 PMCID: PMC5154343 DOI: 10.1038/oncsis.2016.1] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/07/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022] Open
Abstract
Cervical cancer (CC) is one of the most common malignancies in women. Paclitaxel is the front-line chemotherapeutic agent for treating CC. However, its therapeutic efficacy is limited because of chemoresistance, the mechanism of which remains poorly understood. Here, we used microRNA (miRNA) arrays to compare miRNA expression levels in the CC cell lines, HeLa and CaSki, with their paclitaxel resistance counterparts, HeLa/PR and CaSki/PR. We demonstrate that miR-125a was one of most significantly downregulated miRNAs in paclitaxel-resistant cells, which also acquired cisplatin resistance. And that the upregulation of miR-125a sensitized HeLa/PR and CaSki/PR cells to paclitaxel both in vitro and in vivo and to cisplatin in vitro. Moreover, we determined that miR-125a increased paclitaxel and cisplatin sensitivity by downregulating STAT3. MiR-125a enhanced paclitaxel and cisplatin sensitivity by promoting chemotherapy-induced apoptosis. Clinically, miR-125a expression was associated with an increased responsiveness to paclitaxel combined with cisplatin and a more favorable outcome. These data indicate that miR-125a may be a useful method to enable treatment of chemoresistant CC and may also provide a biomarker for predicting paclitaxel and cisplatin responsiveness in CC.
Collapse
Affiliation(s)
- Z Fan
- Department of Oncology, PLA General Hospital, Beijing, China
| | - H Cui
- Department of Oncology, 309th Hospital of PLA, Beijing, China
| | - H Yu
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Q Ji
- Department of Orthopedics, PLA General Hospital, Beijing, China
| | - L Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - B Han
- Department of Endocrinology and Metabolism, 264th Hospital of PLA, Shanxi, China
| | - J Wang
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Q Dong
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Y Li
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Z Yan
- Department of Oncology, PLA General Hospital, Beijing, China
| | - X Yan
- Department of Oncology, PLA General Hospital, Beijing, China
| | - X Zhang
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Z Lin
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Y Hu
- Department of Oncology, PLA General Hospital, Beijing, China
| | - S Jiao
- Department of Oncology, PLA General Hospital, Beijing, China
| |
Collapse
|
116
|
Yue X, Zhao P, Wu K, Huang J, Zhang W, Wu Y, Liang X, He X. GRIM-19 inhibition induced autophagy through activation of ERK and HIF-1α not STAT3 in Hela cells. Tumour Biol 2016; 37:9789-96. [PMID: 26810068 DOI: 10.1007/s13277-016-4877-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/15/2016] [Indexed: 12/13/2022] Open
Abstract
Gene associated with retinoid-interferon-induced mortality (GRIM-19), an important subunit of mitochondrial complex I, has been identified as a tumor suppressor, and its reduced expression has been reported to be associated with tumorigenesis and metastasis. Autophagy has been proposed as a protective mechanism for cell survival under various stresses, including chemotherapy. However, it remains unknown whether GRIM-19 is linked to autophagy and chemotherapy resistance. Here, we showed that suppression of GRIM-19 by shRNA enhanced cell-type-dependent autophagy by activating extracellular regulated protein kinase (ERK) and hypoxia inducible factor-1a (HIF-1a) in a reactive oxygen species (ROS)-mediated manner, and thereby conferred resistance to paclitaxel. Besides, the antioxidant N-acetyl-L-cysteine (NAC) and autophagy inhibitor 3-MA could in part overcome this resistance. We also found that GRIM-19 expression was significantly correlated with clinical stage and grade in patients with cervical cancers. Taken together, our results indicated that GRIM-19 inhibition induced autophagy and chemotherapy resistance, which could affect prognosis of cervical cancers. Our study has identified new function of GRIM-19 and its underlying mechanism, and it will provide possible avenues for therapeutic targeting in cervical cancers.
Collapse
MESH Headings
- Adult
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/drug effects
- Apoptosis Regulatory Proteins/antagonists & inhibitors
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Autophagy/drug effects
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Case-Control Studies
- Cell Proliferation/drug effects
- Drug Resistance, Neoplasm
- Female
- Fluorescent Antibody Technique
- Follow-Up Studies
- HeLa Cells
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lymphatic Metastasis
- Male
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- NADH, NADPH Oxidoreductases/antagonists & inhibitors
- NADH, NADPH Oxidoreductases/genetics
- NADH, NADPH Oxidoreductases/metabolism
- Neoplasm Invasiveness
- Neoplasm Staging
- Paclitaxel/pharmacology
- Prognosis
- RNA, Small Interfering/genetics
- STAT3 Transcription Factor/metabolism
- Survival Rate
- Tumor Cells, Cultured
- Uterine Cervical Neoplasms/drug therapy
- Uterine Cervical Neoplasms/metabolism
- Uterine Cervical Neoplasms/pathology
Collapse
Affiliation(s)
- Xin Yue
- Clinical Research Center, Wuhan Medical and Healthcare Center for Women and Children, No. 100 Hongkong Rd, Wuhan, Hubei, China
| | - Peiwei Zhao
- Clinical Research Center, Wuhan Medical and Healthcare Center for Women and Children, No. 100 Hongkong Rd, Wuhan, Hubei, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, China
| | - Juan Huang
- Department of Pathology, Wuhan Medical and Healthcare Center for Women and Children, Wuhan, Hubei, China
| | - Wen Zhang
- Department of Pathology, Wuhan Medical and Healthcare Center for Women and Children, Wuhan, Hubei, China
| | - Yaogui Wu
- School of Public Health, Wuhan University, Wuhan, China
| | - Xiaohui Liang
- Department of Oncology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xuelian He
- Clinical Research Center, Wuhan Medical and Healthcare Center for Women and Children, No. 100 Hongkong Rd, Wuhan, Hubei, China.
| |
Collapse
|
117
|
Wang L, Li H, Ren Y, Zou S, Fang W, Jiang X, Jia L, Li M, Liu X, Yuan X, Chen G, Yang J, Wu C. Targeting HDAC with a novel inhibitor effectively reverses paclitaxel resistance in non-small cell lung cancer via multiple mechanisms. Cell Death Dis 2016; 7:e2063. [PMID: 26794658 PMCID: PMC4816165 DOI: 10.1038/cddis.2015.328] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 09/25/2015] [Accepted: 10/06/2015] [Indexed: 11/25/2022]
Abstract
Chemotherapy paclitaxel yields significant reductions in tumor burden in the majority of advanced non-small cell lung cancer (NSCLC) patients. However, acquired resistance limits its clinical use. Here we demonstrated that the histone deacetylase (HDAC) was activated in paclitaxel-resistant NSCLC cells, and its activation promoted proliferation and tumorigenesis of paclitaxel-resistant NSCLC cells in vitro and in vivo. By contrast, knockdown of HDAC1, a primary isoform of HDAC, sensitized resistant cells to paclitaxel in vitro. Furthermore, we observed that overexpression of HDAC1 was associated with the downregulation of p21, a known HDAC target, in advanced NSCLC patients with paclitaxel treatment, and predicted chemotherapy resistance and bad outcome. In addition, we also identified a novel HDACs inhibitor, SNOH-3, which inhibited HDAC expression and activity, induced cell apoptosis, and suppressed cell migration, invasion and angiogenesis. Notably, co-treatment with SNOH-3 and paclitaxel overcome paclitaxel resistance through inhibiting HDAC activity, leading to the induction of apoptosis and suppression of angiogenesis in vitro and in preclinical model. In summary, our data demonstrate a role of HDAC in paclitaxel-resistant NSCLC and provide a promising therapeutic strategy to overcome paclitaxel-acquired resistance.
Collapse
Affiliation(s)
- L Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - H Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Y Ren
- Department of Pathology, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, PR China
| | - S Zou
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, PR China
| | - W Fang
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, PR China
| | - X Jiang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - L Jia
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - M Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - X Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - X Yuan
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - G Chen
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, PR China
| | - J Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| | - C Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
- Benxi Institute of Pharmaceutical Research, Shenyang Pharmaceutical University, Shenyang, PR China
| |
Collapse
|
118
|
Altuntas S, Rossin F, Marsella C, D'Eletto M, Hidalgo LD, Farrace MG, Campanella M, Antonioli M, Fimia GM, Piacentini M. The transglutaminase type 2 and pyruvate kinase isoenzyme M2 interplay in autophagy regulation. Oncotarget 2015; 6:44941-54. [PMID: 26702927 PMCID: PMC4792602 DOI: 10.18632/oncotarget.6759] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/20/2015] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a self-degradative physiological process by which the cell removes worn-out or damaged components. Constant at basal level it may become highly active in response to cellular stress. The type 2 transglutaminase (TG2), which accumulates under stressful cell conditions, plays an important role in the regulation of autophagy and cells lacking this enzyme display impaired autophagy/mitophagy and a consequent shift their metabolism to glycolysis. To further define the molecular partners of TG2 involved in these cellular processes, we analysed the TG2 interactome under normal and starved conditions discovering that TG2 interacts with various proteins belonging to different functional categories. Herein we show that TG2 interacts with pyruvate kinase M2 (PKM2), a rate limiting enzyme of glycolysis which is responsible for maintaining a glycolytic phenotype in malignant cells and displays non metabolic functions, including transcriptional co-activation and protein kinase activity. Interestingly, the ablation of PKM2 led to the decrease of intracellular TG2's transamidating activity paralleled by an increase of its tyrosine phosphorylation. Along with this, a significant decrease of ULK1 and Beclin1 was also recorded, thus suggesting a block in the upstream regulation of autophagosome formation. These data suggest that the PKM2/TG2 interplay plays an important role in the regulation of autophagy in particular under cellular stressful conditions such as those displayed by cancer cells.
Collapse
Affiliation(s)
- Sara Altuntas
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Federica Rossin
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Claudia Marsella
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Manuela D'Eletto
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | | | | | - Michelangelo Campanella
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- Department of Comparative Biomedical Sciences, The Royal Veterinary College London and UCL Consortium for Mitochondrial Research, London, UK
| | - Manuela Antonioli
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
| | - Gian Maria Fimia
- National Institute for Infectious Diseases, IRCCS “Lazzaro Spallanzani”, Rome, Italy
- Department of Biological and Environmental Science and Technology (Di.S.Te.B.A.), University of Salento, Lecce, Italy
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- National Institute for Infectious Diseases, IRCCS “Lazzaro Spallanzani”, Rome, Italy
| |
Collapse
|
119
|
Fisher CM, Schefter TE. Profile of bevacizumab and its potential in the treatment of cervical cancer. Onco Targets Ther 2015; 8:3425-31. [PMID: 26640382 PMCID: PMC4657807 DOI: 10.2147/ott.s73251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Blocking angiogenesis is an effective antitumor strategy proven in many disease sites. Anti-angiogenic therapies are fulfilling the promise of improved outcomes in cervical cancer as demonstrated in several recent trials. With its overall survival improvement in metastatic or recurrent cervical cancer, a frame shift in the management of these patients has occurred. The US Food and Drug Administration approval of bevacizumab in advanced cervical cancer has led to national guidelines, including the US National Comprehensive Cancer Network guidelines for cervical cancer, including systemic regimens containing bevacizumab as first line combination therapy. Future trials will build on this anti-angiogenesis backbone via targeting additional novel pathways and potentially leading to further improved outcomes in cervical cancer.
Collapse
Affiliation(s)
- Christine M Fisher
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| | - Tracey E Schefter
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| |
Collapse
|
120
|
Petrova V, Mancini M, Agostini M, Knight RA, Annicchiarico-Petruzzelli M, Barlev NA, Melino G, Amelio I. TAp73 transcriptionally represses BNIP3 expression. Cell Cycle 2015; 14:2484-93. [PMID: 25950386 PMCID: PMC4612661 DOI: 10.1080/15384101.2015.1044178] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 03/26/2015] [Accepted: 04/18/2015] [Indexed: 01/07/2023] Open
Abstract
TAp73 is a tumor suppressor transcriptional factor, belonging to p53 family. Alteration of TAp73 in tumors might lead to reduced DNA damage response, cell cycle arrest and apoptosis. Carcinogen-induced TAp73(-/-) tumors display also increased angiogenesis, associated to hyperactivition of hypoxia inducible factor signaling. Here, we show that TAp73 suppresses BNIP3 expression, directly binding its gene promoter. BNIP3 is a hypoxia responsive protein, involved in a variety of cellular processes, such as autophagy, mitophagy, apoptosis and necrotic-like cell death. Therefore, through different cellular process altered expression of BNIP3 may differently contribute to cancer development and progression. We found a significant upregulation of BNIP3 in human lung cancer datasets, and we identified a direct association between BNIP3 expression and survival rate of lung cancer patients. Our data therefore provide a novel transcriptional target of TAp73, associated to its antagonistic role on HIF signaling in cancer, which might play a role in tumor suppression.
Collapse
Affiliation(s)
- Varvara Petrova
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Molecular Pharmacology Laboratory; Saint-Petersburg Institute of Technology; Saint-Petersburg, Russia
| | - Mara Mancini
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
| | - Massimiliano Agostini
- Department of Experimental Medicine and Surgery; University of Rome “Tor Vergata”; Rome, Italy
| | - Richard A Knight
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
| | | | - Nikolai A Barlev
- Molecular Pharmacology Laboratory; Saint-Petersburg Institute of Technology; Saint-Petersburg, Russia
- Gene Expression Laboratory; Institute of Cytology; Saint-Petersburg, Russia
| | - Gerry Melino
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
- Molecular Pharmacology Laboratory; Saint-Petersburg Institute of Technology; Saint-Petersburg, Russia
- Department of Experimental Medicine and Surgery; University of Rome “Tor Vergata”; Rome, Italy
- Biochemistry Laboratory IDI-IRCC; Rome, Italy
| | - Ivano Amelio
- Medical Research Council; Toxicology Unit; Leicester University; Leicester, UK
| |
Collapse
|
121
|
Rahier NJ, Molinier N, Long C, Deshmukh SK, Kate AS, Ranadive P, Verekar SA, Jiotode M, Lavhale RR, Tokdar P, Balakrishnan A, Meignan S, Robichon C, Gomes B, Aussagues Y, Samson A, Sautel F, Bailly C. Anticancer activity of koningic acid and semisynthetic derivatives. Bioorg Med Chem 2015; 23:3712-21. [PMID: 25937235 DOI: 10.1016/j.bmc.2015.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/28/2015] [Accepted: 04/02/2015] [Indexed: 10/23/2022]
Abstract
A screening program aimed at discovering novel anticancer agents based on natural products led to the selection of koningic acid (KA), known as a potent inhibitor of glycolysis. A method was set up to produce this fungal sesquiterpene lactone in large quantities by fermentation, thus allowing (i) an extensive analysis of its anticancer potential in vitro and in vivo and (ii) the semi-synthesis of analogues to delineate structure-activity relationships. KA was characterized as a potent, but non-selective cytotoxic agent, active under both normoxic and hypoxic conditions and inactive in the A549 lung cancer xenograft model. According to our SAR, the acidic group could be replaced to keep bioactivity but an intact epoxide is essential.
Collapse
Affiliation(s)
- Nicolas J Rahier
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France.
| | - Nicolas Molinier
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - Christophe Long
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - Sunil Kumar Deshmukh
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Abhijeet S Kate
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Prafull Ranadive
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Shilpa Amit Verekar
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Mangesh Jiotode
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Rahul R Lavhale
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Pradipta Tokdar
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Arun Balakrishnan
- Piramal Enterprises Limited, 1 Nirlon Complex, Off Western Express Highway, Goregaon East, Mumbai, Maharashtra 400 063, India
| | - Samuel Meignan
- Antitumoral Pharmacology Unit, Centre Oscar Lambret, Inserm U908, BP 307, 59020 Lille cedex, France
| | - Céline Robichon
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - Bruno Gomes
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - Yannick Aussagues
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - Arnaud Samson
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - François Sautel
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France
| | - Christian Bailly
- Institut de Recherche Pierre Fabre, Centre de Recherche et Développement, 3 Avenue Hubert Curien-BP 13562, 31035 Toulouse cedex 1, France.
| |
Collapse
|
122
|
Jäger A, Jäger E, Surman F, Höcherl A, Angelov B, Ulbrich K, Drechsler M, Garamus VM, Rodriguez-Emmenegger C, Nallet F, Štěpánek P. Nanoparticles of the poly([N-(2-hydroxypropyl)]methacrylamide)-b-poly[2-(diisopropylamino)ethyl methacrylate] diblock copolymer for pH-triggered release of paclitaxel. Polym Chem 2015. [DOI: 10.1039/c5py00567a] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The potential of self-assembled nanoparticles for in vitro cytostatic activity has been explored on cancer cells.
Collapse
|