51
|
Zhao X, Sun B, Zhang J, Zhang R, Zhang Q. Short-chain C6 ceramide sensitizes AT406-induced anti-pancreatic cancer cell activity. Biochem Biophys Res Commun 2016; 479:166-172. [PMID: 27562715 DOI: 10.1016/j.bbrc.2016.08.121] [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: 08/16/2016] [Accepted: 08/20/2016] [Indexed: 02/07/2023]
Abstract
Our previous study has shown that AT406, a first-in-class small molecular antagonist of IAPs (inhibitor of apoptosis proteins), inhibits pancreatic cancer cell proliferation in vitro and in vivo. The aim of this research is to increase AT406's sensitivity by adding short-chain C6 ceramide. We show that co-treatment of C6 ceramide dramatically potentiated AT406-induced caspase/apoptosis activation and cytotoxicity in established (Panc-1 and Mia-PaCa-2 lines) and primary human pancreatic cancer cells. Reversely, caspase inhibitors largely attenuated C6 ceramide plus AT406-induced above cancer cell death. Molecularly, C6 ceramide downregulated Bcl-2 to increase AT406's sensitivity in pancreatic cancer cells. Intriguingly, C6 ceramide-mediated AT406 sensitization was nullified with Bcl-2 shRNA knockdown or pretreatment of the Bcl-2 inhibitor ABT-737. In vivo, liposomal C6 ceramide plus AT406 co-administration dramatically inhibited Panc-1 xenograft tumor growth in severe combined immunodeficient (SCID) mice. The combined anti-tumor activity was significantly more potent than either single treatment. Expressions of IAPs (cIAP1/XIAP) and Bcl-2 were downregulated in Panc-1 xenografts with the co-administration. Together, we demonstrate that C6 ceramide sensitizes AT406-mediated anti-pancreatic cancer cell activity possibly via downregulating Bcl-2.
Collapse
Affiliation(s)
- Xiaoguang Zhao
- Department of Neurosurgery, CPLA Bethune International Peace Hospital, Shijiazhuang, Hebei, China.
| | - Baoyou Sun
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Jingjing Zhang
- Department of Thoracic Surgery III, The Chest Hospital of Hebei Province, Shijiazhuang, Hebei, China
| | - Ruishen Zhang
- Hospital of China Railway Electric Bureau Group First Engineering Company, Shijiazhuang, Hebei, China
| | - Qing Zhang
- Department of Neurology, The Chest Hospital of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
52
|
Jiang H, Liu W, Zhan SK, Pan YX, Bian LG, Sun B, Sun QF, Pan SJ. GSK621 Targets Glioma Cells via Activating AMP-Activated Protein Kinase Signalings. PLoS One 2016; 11:e0161017. [PMID: 27532105 PMCID: PMC4988667 DOI: 10.1371/journal.pone.0161017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/28/2016] [Indexed: 01/03/2023] Open
Abstract
Here, we studied the anti-glioma cell activity by a novel AMP-activated protein kinase (AMPK) activator GSK621. We showed that GSK621 was cytotoxic to human glioma cells (U87MG and U251MG lines), possibly via provoking caspase-dependent apoptotic cell death. Its cytotoxicity was alleviated by caspase inhibitors. GSK621 activated AMPK to inhibit mammalian target of rapamycin (mTOR) and downregulate Tetraspanin 8 (Tspan8) in glioma cells. AMPK inhibition, through shRNA knockdown of AMPKα or introduction of a dominant negative (T172A) AMPKα, almost reversed GSK621-induced AMPK activation, mTOR inhibition and Tspan8 degradation. Consequently, GSK621’s cytotoxicity in glioma cells was also significantly attenuated by AMPKα knockdown or mutation. Further studies showed that GSK621, at a relatively low concentration, significantly potentiated temozolomide (TMZ)’s sensitivity and lethality against glioma cells. We summarized that GSK621 inhibits human glioma cells possibly via activating AMPK signaling. This novel AMPK activator could be a novel and promising anti-glioma cell agent.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Wei Liu
- Department of Stereotactic and Functional Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Shi-Kun Zhan
- Department of Stereotactic and Functional Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Yi-Xin Pan
- Department of Stereotactic and Functional Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Liu-Guan Bian
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Bomin Sun
- Department of Stereotactic and Functional Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
| | - Qing-Fang Sun
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
- * E-mail: (Q-FS); (S-JP)
| | - Si-Jian Pan
- Department of Neurosurgery, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, 200025, P.R. China
- * E-mail: (Q-FS); (S-JP)
| |
Collapse
|
53
|
Wang J, Feng C, He Y, Ding W, Sheng J, Arshad M, Zhang X, Li P. Phosphorylation of apoptosis repressor with caspase recruitment domain by protein kinase CK2 contributes to chemotherapy resistance by inhibiting doxorubicin induced apoptosis. Oncotarget 2016; 6:27700-13. [PMID: 26172393 PMCID: PMC4695019 DOI: 10.18632/oncotarget.4392] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/17/2015] [Indexed: 01/15/2023] Open
Abstract
The development of cancer resistance to chemotherapy is the major obstacle to cancer therapy. Here, we identified that the phosphorylation of apoptosis repressor with caspase recruitment domain (ARC) at threonine 149 was essential to inhibit doxorubicin (DOX) induced apoptosis and mitochondrial fission in cancer cells. Our further study showed that casein kinase II (CK2) inhibitors could decrease the phosphorylation levels of ARC and make cancer cells sensitive to undergoing apoptosis. Furthermore, CK2α and CK2α', catalytic subunits of CK2, were observed to translocate into nuclear in cancer cells with the treatment of DOX. Finally, the synergistically therapeutic effect by combining DOX and CK2 inhibitor was confirmed in tumor xenograft model. Taken together, our results revealed that CK2-mediated phosphorylation of ARC contributed to chemotherapy resistance by inhibiting DOX induced apoptosis and combining DOX with CK2 inhibitor could induce apoptosis of cancer cells synergistically by down-regulating the phosphorylation of ARC. Therefore, development of new therapeutic strategies based on ARC and CK2, is promising for overcoming cancer resistance to chemotherapy.
Collapse
Affiliation(s)
- Jianxun Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao, 266021, China
| | - Chang Feng
- National Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuqi He
- Department of Gastroenterology, Beijing Military General Hospital, Beijing, 100700, China
| | - Wei Ding
- Affiliated Hospital, Medical College of Qingdao University, Qingdao, 266003, China
| | - Jianqiu Sheng
- Department of Gastroenterology, Beijing Military General Hospital, Beijing, 100700, China
| | - Muhammad Arshad
- National Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaojie Zhang
- National Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Peifeng Li
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao, 266021, China
| |
Collapse
|
54
|
Pan WY, Lin KJ, Huang CC, Chiang WL, Lin YJ, Lin WC, Chuang EY, Chang Y, Sung HW. Localized sequence-specific release of a chemopreventive agent and an anticancer drug in a time-controllable manner to enhance therapeutic efficacy. Biomaterials 2016; 101:241-50. [PMID: 27294541 DOI: 10.1016/j.biomaterials.2016.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/30/2016] [Accepted: 06/02/2016] [Indexed: 12/22/2022]
Abstract
Combination chemotherapy with multiple drugs commonly requires several injections on various schedules, and the probability that the drug molecules reach the diseased tissues at the proper time and effective therapeutic concentrations is very low. This work elucidates an injectable co-delivery system that is based on cationic liposomes that are adsorbed on anionic hollow microspheres (Lipos-HMs) via electrostatic interaction, from which the localized sequence-specific release of a chemopreventive agent (1,25(OH)2D3) and an anticancer drug (doxorubicin; DOX) can be thermally driven in a time-controllable manner by an externally applied high-frequency magnetic field (HFMF). Lipos-HMs can greatly promote the accumulation of reactive oxygen species (ROS) in tumor cells by reducing their cytoplasmic expression of an antioxidant enzyme (superoxide dismutase) by 1,25(OH)2D3, increasing the susceptibility of cancer cells to the cytotoxic action of DOX. In nude mice that bear xenograft tumors, treatment with Lipos-HMs under exposure to HFMF effectively inhibits tumor growth and is the most effective therapeutic intervention among all the investigated. These empirical results demonstrate that the synergistic anticancer effects of sequential release of 1,25(OH)2D3 and DOX from the Lipos-HMs may have potential for maximizing DOX cytotoxicity, supporting more effective cancer treatment.
Collapse
Affiliation(s)
- Wen-Yu Pan
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Kun-Ju Lin
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan, ROC; Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan, ROC
| | - Chieh-Cheng Huang
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Wei-Lun Chiang
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Yu-Jung Lin
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Wei-Chih Lin
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Er-Yuan Chuang
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC
| | - Yen Chang
- Department of Cardiovascular Surgery, Heart Center, Cheng Hsin General Hospital, Taipei, Taiwan, ROC; Collage of Medicine, National Yang Ming University, Taipei, Taiwan, ROC.
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan, ROC.
| |
Collapse
|
55
|
Morimoto N, Wakamura M, Muramatsu K, Toita S, Nakayama M, Shoji W, Suzuki M, Winnik FM. Membrane Translocation and Organelle-Selective Delivery Steered by Polymeric Zwitterionic Nanospheres. Biomacromolecules 2016; 17:1523-35. [DOI: 10.1021/acs.biomac.6b00172] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nobuyuki Morimoto
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Masaru Wakamura
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Kanna Muramatsu
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Sayaka Toita
- Department
of Chemistry and Faculty of Pharmacy, University of Montreal, CP6128 Succursale
Center Ville, Montreal, QC H3C 3J7, Canada
| | - Masafumi Nakayama
- Frontier
Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai 980-8578, Japan
| | - Wataru Shoji
- Frontier
Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Aramaki aza Aoba 6-3, Aoba-ku, Sendai 980-8578, Japan
| | - Makoto Suzuki
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02 Aramaki-aza Aoba, Aoba-ku, Sendai, 980-8579, Japan
| | - Françoise M. Winnik
- Department
of Chemistry and Faculty of Pharmacy, University of Montreal, CP6128 Succursale
Center Ville, Montreal, QC H3C 3J7, Canada
- National Institute
for Materials Science, WPI International Center for Materials Nanoarchitectonics
(MANA), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| |
Collapse
|
56
|
C6 ceramide sensitizes the anti-hepatocellular carcinoma (HCC) activity by AZD-8055, a novel mTORC1/2 dual inhibitor. Tumour Biol 2016; 37:11039-48. [PMID: 26897748 DOI: 10.1007/s13277-015-4598-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022] Open
Abstract
Aberrant activation of mammalian target of rapamycin (mTOR) plays pivotal roles in promoting hepatocellular carcinoma (HCC) tumorigenesis and chemoresistance. Here, we tested the potential anti-HCC activity by a novel mTOR complex 1/2 (mTORC1/2) dual inhibitor AZD-8055 and, more importantly, the potential AZD-8055 sensitization effect by a cell-permeable short-chain ceramide (C6). We showed that AZD-8055 mainly exerted moderate cytotoxic effect against a panel of HCC cell lines (HepG2, Hep3B, and SMMC-7721). Co-treatment of C6 ceramide remarkably augmented AZD-8055-induced HCC cytotoxicity. Meanwhile, C6 ceramide dramatically potentiated AZD-8055-induced HCC cell apoptotic death. Further studies demonstrated that AZD-8055 and C6 ceramide synergistically induced anti-survival and pro-apoptotic activity in primary cultured human HCC cells, but not in the non-cancerous human hepatocytes. Signaling studies showed that AZD-8055 and C6 ceramide synergistically suppressed Akt-mTOR complex 1/2 cascade activation. In vivo, AZD-8055 oral administration suppressed HepG2 hepatoma xenograft growth in nude mice, while moderately improving mice survival. Its anti-tumor activity was dramatically potentiated with co-administration of a liposome-packed C6 ceramide. Together, these results demonstrate that concurrent targeting mTORC1/2 by AZD-8055 exerts anti-tumor ability in preclinical HCC models, and its activity is further sensitized with co-administration of C6 ceramide.
Collapse
|
57
|
Sriraman SK, Pan J, Sarisozen C, Luther E, Torchilin V. Enhanced Cytotoxicity of Folic Acid-Targeted Liposomes Co-Loaded with C6 Ceramide and Doxorubicin: In Vitro Evaluation on HeLa, A2780-ADR, and H69-AR Cells. Mol Pharm 2016; 13:428-37. [PMID: 26702994 DOI: 10.1021/acs.molpharmaceut.5b00663] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Current research in cancer therapy is beginning to shift toward the use of combinational drug treatment regimens. However, the efficient delivery of drug combinations is governed by a number of complex factors in the clinical setting. Therefore, the ability to synchronize the pharmacokinetics of the individual therapeutic agents present in combination not only to allow for simultaneous tumor accumulation but also to allow for a synergistic relationship at the intracellular level could prove to be advantageous. In this work, we report the development of a novel folic acid-targeted liposomal formulation simultaneously co-loaded with C6 ceramide and doxorubicin [FA-(C6+Dox)-LP]. In vitro cytotoxicity assays showed that the FA-(C6+Dox)-LP was able to significantly reduce the IC50 of Dox when compared to that after the treatment with the doxorubicin-loaded liposomes (Dox-LP) as well as the untargeted drug co-loaded (C6+Dox)-LP on HeLa, A2780-ADR, and H69-AR cells. The analysis of the cell cycle distribution showed that while the C6 liposomes (C6-LP) did not cause cell cycle arrest, all the Dox-containing liposomes mediated cell cycle arrest in HeLa cells in the G2 phase at Dox concentrations of 0.3 and 1 μM and in the S phase at the higher concentrations. It was also found that this arrest in the S phase precedes the progression of the cells to apoptosis. The targeted FA-(C6+Dox)-LP were able to significantly enhance the induction of apoptotic events in HeLa cell monolayers as compared to the other treatment groups. Next, using time-lapse phase holographic imaging microscopy, it was found that upon treatment with the FA-(C6+Dox)-LP, the HeLa cells underwent rapid progression to apoptosis after 21 h as evidenced by a drastic drop in the average area of the cells after loss of cell membrane integrity. Finally, upon evaluation in a HeLa spheroid cell model, treatment with the FA-(C6+Dox)-LP showed significantly higher levels of cell death compared to those with C6-LP and Dox-LP. Overall, this study clearly shows that the co-delivery of C6 ceramide and Dox using a liposomal platform significantly correlates with an antiproliferative effect due to cell cycle regulation and subsequent induction of apoptosis and thus warrants its further evaluation in preclinical animal models.
Collapse
Affiliation(s)
- Shravan Kumar Sriraman
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Jiayi Pan
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Can Sarisozen
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Ed Luther
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States
| | - Vladimir Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston, Massachusetts 02115, United States.,Department of Biochemistry, Faculty of Science, King Abdulaziz University , Jeddah, Saudi Arabia
| |
Collapse
|
58
|
AMPK Regulation of Cell Growth, Apoptosis, Autophagy, and Bioenergetics. EXPERIENTIA SUPPLEMENTUM (2012) 2016; 107:45-71. [PMID: 27812976 DOI: 10.1007/978-3-319-43589-3_3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In eukaryotic cells, AMP-activated protein kinase (AMPK) generally promotes catabolic pathways that produce ATP and at the same time inhibits anabolic pathways involved in different processes that consume ATP. As an energy sensor, AMPK is involved in the main cellular functions implicated in cell fate, such as cell growth and autophagy.Recently, AMPK has been connected with apoptosis regulation, although the molecular mechanism by which AMPK induces and/or inhibits cell death is not clear.This chapter reviews the essential role of AMPK in signaling pathways that respond to cellular stress and damage, highlighting the complex and reciprocal regulation between AMPK and their targets and effectors. The therapeutic implications of the role of AMPK in different pathologies such as diabetes, cancer, or mitochondrial dysfunctions are still controversial, and it is necessary to further investigate the molecular mechanisms underlying AMPK activation.
Collapse
|
59
|
Teixeira V, Costa V. Unraveling the role of the Target of Rapamycin signaling in sphingolipid metabolism. Prog Lipid Res 2015; 61:109-33. [PMID: 26703187 DOI: 10.1016/j.plipres.2015.11.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
Sphingolipids are important bioactive molecules that regulate basic aspects of cellular metabolism and physiology, including cell growth, adhesion, migration, senescence, apoptosis, endocytosis, and autophagy in yeast and higher eukaryotes. Since they have the ability to modulate the activation of several proteins and signaling pathways, variations in the relative levels of different sphingolipid species result in important changes in overall cellular functions and fate. Sphingolipid metabolism and their route of synthesis are highly conserved from yeast to mammalian cells. Studies using the budding yeast Saccharomyces cerevisiae have served in many ways to foster our understanding of sphingolipid dynamics and their role in the regulation of cellular processes. In the past decade, studies in S. cerevisiae have unraveled a functional association between the Target of Rapamycin (TOR) pathway and sphingolipids, showing that both TOR Complex 1 (TORC1) and TOR Complex 2 (TORC2) branches control temporal and spatial aspects of sphingolipid metabolism in response to physiological and environmental cues. In this review, we report recent findings in this emerging and exciting link between the TOR pathway and sphingolipids and implications in human health and disease.
Collapse
Affiliation(s)
- Vitor Teixeira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Porto, Portugal; ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Departamento de Biologia Molecular, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Vítor Costa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Porto, Portugal; ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Departamento de Biologia Molecular, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
60
|
Abstract
Studies over the past two decades have identified ceramide as a multifunctional central molecule in the sphingolipid biosynthetic pathway. Given its diverse tumor suppressive activities, molecular understanding of ceramide action will produce fundamental insights into processes that limit tumorigenesis and may identify key molecular targets for therapeutic intervention. Ceramide can be activated by a diverse array of stresses such as heat shock, genotoxic damage, oxidative stress and anticancer drugs. Ceramide triggers a variety of tumor suppressive and anti-proliferative cellular programs such as apoptosis, autophagy, senescence, and necroptosis by activating or repressing key effector molecules. Defects in ceramide generation and metabolism in cancer contribute to tumor cell survival and resistance to chemotherapy. The potent and versatile anticancer activity profile of ceramide has motivated drug development efforts to (re-)activate ceramide in established tumors. This review focuses on our current understanding of the tumor suppressive functions of ceramide and highlights the potential downstream targets of ceramide which are involved in its tumor suppressive action.
Collapse
|
61
|
Zhai L, Sun N, Han Z, Jin HC, Zhang B. Liposomal short-chain C6 ceramide induces potent anti-osteosarcoma activity in vitro and in vivo. Biochem Biophys Res Commun 2015; 468:274-80. [PMID: 26505795 DOI: 10.1016/j.bbrc.2015.10.113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 10/21/2015] [Indexed: 10/22/2022]
Abstract
Osteosarcoma (OS) remains one deadly disease for many affected patients. The search for novel and more efficient anti-OS agents is urgent. In the current study, we demonstrated that liposome-packed C6 ceramide exerted potent cytotoxic effect against established (U2OS and MG-63 lines) and primary human OS cells. Meanwhile, the liposomal C6 (ceramide) induced caspase-mediated apoptotic death in OS cells. Liposomal C6 was significantly more potent than conventional free C6 in inhibiting OS cells, yet it was safe to non-cancerous bone cells (primary murine osteoblasts or human MLO-Y4 osteocytic cells). At the signaling level, we showed that liposomal C6 potently inhibited Akt activation in OS cells. Further studies revealed that a low dose of liposomal C6 dramatically sensitized the in vitro anti-OS activity of two conventional chemodrugs: methotrexate (MTX) and doxorubicin. In vivo, intravenous injection of liposomal C6 inhibited Akt activation and suppressed U2OS xenograft growth in nude mice without causing apparent toxicities. Meanwhile, when given at a low-dose (5 mg/kg body weight), liposomal C6 dramatically sensitized MTX's anti-U2OS activity in vivo. Collectively, our data demonstrate that liposomal C6 exerts potent anti-tumor activity in preclinical OS models.
Collapse
Affiliation(s)
- Lei Zhai
- Department of Orthopaedic Surgery, The Affiliated Hospital of Logistics College of Chinese People's Armed Police Force, Tianjin, China
| | - Nan Sun
- Department of Nephropathy, The Affiliated Hospital of Logistics College of Chinese People's Armed Police Force, Tianjin, China
| | - Zhe Han
- Department of Orthopaedic Surgery, Tianjin Hospital, Tianjin, China
| | - Hai-chao Jin
- Department of Orthopaedic Surgery, The Affiliated Hospital of Logistics College of Chinese People's Armed Police Force, Tianjin, China
| | - Bo Zhang
- Department of Immunology, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
62
|
LAPTM4B facilitates late endosomal ceramide export to control cell death pathways. Nat Chem Biol 2015; 11:799-806. [DOI: 10.1038/nchembio.1889] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/09/2015] [Indexed: 12/31/2022]
|
63
|
Song W, Mu H, Wu J, Liao M, Zhu H, Zheng L, He X, Niu B, Zhai Y, Bai C, Lei A, Li G, Hua J. miR-544 Regulates Dairy Goat Male Germline Stem Cell Self-Renewal via Targeting PLZF. J Cell Biochem 2015; 116:2155-65. [DOI: 10.1002/jcb.25172] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
Affiliation(s)
- Wencong Song
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Hailong Mu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Jiang Wu
- College of Agriculture; Guangdong Ocean University; Zhanjiang 524088 China
| | - Mingzhi Liao
- College of Life Science; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Haijing Zhu
- College of Life Science; Yulin College, Yulin University; 719000 China
| | - Liming Zheng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Xin He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Bowen Niu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Yuanxin Zhai
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Chunling Bai
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education; Inner Mongolia University; Hohhot 010021 China
| | - Anmin Lei
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| | - Guangpeng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education; Inner Mongolia University; Hohhot 010021 China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China; Northwest A&F University; Yangling Shaanxi 712100 China
| |
Collapse
|
64
|
Chen MB, Jiang Q, Liu YY, Zhang Y, He BS, Wei MX, Lu JW, Ji Y, Lu PH. C6 ceramide dramatically increases vincristine sensitivity both in vivo and in vitro, involving AMP-activated protein kinase-p53 signaling. Carcinogenesis 2015; 36:1061-70. [PMID: 26116623 DOI: 10.1093/carcin/bgv094] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 06/18/2015] [Indexed: 12/15/2022] Open
Abstract
Use of the conventional cancer chemotherapy (i.e. vincristine) is limited in tumor cells exhibiting pre-existing or acquired resistance. Here, we found that C6 ceramide (C6) dramatically sensitized vincristine's activity. In vitro, C6 and vincristine coadministration induced substantial necrosis and apoptosis in multiple human cancer cell lines, which were accompanied by a profound AMP-activated protein kinase (AMPK) activation, subsequent p53 activation, mTORC1 inactivation and Bcl-2/HIF-1α downregulation. Such synergistic effects were attenuated by AMPK inactivation through genetic mutation or short hairpin RNA silencing. Coadministration-activated p53 translocated to mitochondria, and formed a complex with cyclophilin-D, leading to mitochondrial permeability transition pore opening and cell necrosis. Disrupting p53-Cyp-D complexation through pharmacological or genetic means reduced costimulation-induced cytotoxicity. In vivo, a liposomal C6 was synthesized, which dramatically enhanced the antiproliferative activity of vincristine on HCT-116 or A2780 xenografts. Together, C6 sensitizes vincristine-induced anticancer activity in vivo and in vitro, involving activating AMPK-p53 signaling.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, No.91, Qianjin Road, Kunshan, Jiangsu 215300, China, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215021, China, Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China, Department of Traditional Chinese Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China, Department of Medical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Baiziting 42, Nanjing, Jiangsu 210009, China, Department of Thoracic Surgery and Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, Jiangsu 214023, China and Department of Medical Oncology Center, Institute of Integrated Traditional and Western Medicine Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yuan-yuan Liu
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215021, China
| | - Yan Zhang
- Department of Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, No.91, Qianjin Road, Kunshan, Jiangsu 215300, China, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215021, China, Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China, Department of Traditional Chinese Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China, Department of Medical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Baiziting 42, Nanjing, Jiangsu 210009, China, Department of Thoracic Surgery and Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, Jiangsu 214023, China and Department of Medical Oncology Center, Institute of Integrated Traditional and Western Medicine Affiliated to Nanjing Medical University, Nanjing 210029, China
| | - Bang-shun He
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Mu-Xin Wei
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jian-Wei Lu
- Department of Medical Oncology, Nanjing Medical University Affiliated Cancer Hospital, Baiziting 42, Nanjing, Jiangsu 210009, China, Department of Thoracic Surgery and
| | - Yong Ji
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, Jiangsu 214023, China and
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, No. 299, Qingyang Road, Wuxi, Jiangsu 214023, China and Department of Medical Oncology Center, Institute of Integrated Traditional and Western Medicine Affiliated to Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
65
|
Chen MB, Yang L, Lu PH, Fu XL, Zhang Y, Zhu YQ, Tian Y. MicroRNA-101 down-regulates sphingosine kinase 1 in colorectal cancer cells. Biochem Biophys Res Commun 2015; 463:954-60. [PMID: 26071354 DOI: 10.1016/j.bbrc.2015.06.041] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/05/2015] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRs) dysregulation is a general feature of colorectal cancer (CRC) and other solid tumors, and is associated cancer progression. In the current study, we demonstrate that microRNA-101 (miR-101) inhibits CRC cells probably through down-regulating sphingosine kinase 1 (SphK1). Our results showed that exogenously expressing miR-101 inhibited CRC cell (HT-29 and HCT-116 lines) growth in vitro. At the molecular level, miR-101 dramatically down-regulated SphK1 mRNA and protein expression, causing pro-apoptotic ceramide production in above CRC cells. On the other hand, inhibition of miR-101 through expressing antagomiR-101 increased SphK1 expression to down-regulate ceramide level in HT-29 cells. miR-101 expression increased the in vitro anti-CRC activity of conventional chemo-agents: paclitaxel and doxorubicin. CRC cells with SphK1-shRNA knockdown showed similar phenotypes as the miR-101-expressed CRC cells, presenting with elevated level of ceramide and high sensitivity to paclitaxel or doxorubicin. In vivo, HCT-116 xenograft growth in severe combined immuno-deficient (SCID) mice was dramatically inhibited by over-expressing miR-101. Further, miR-101 enhanced paclitaxel-induced anti-HCT-116 activity in vivo. Together, these results indicate that miR-101 exerts its anti-CRC activities probably through down-regulating SphK1.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Radiotherapy and Oncology, the Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China; Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Lan Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xing-Li Fu
- Jiangsu University Health Science Center, Jiangsu, China
| | - Yan Zhang
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Ya-Qun Zhu
- Department of Radiotherapy and Oncology, the Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.
| | - Ye Tian
- Department of Radiotherapy and Oncology, the Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou, China.
| |
Collapse
|
66
|
Ge YQ, Xu XF, Yang B, Chen Z, Cheng RB. Saponins from Rubus parvifolius L. induce apoptosis in human chronic myeloid leukemia cells through AMPK activation and STAT3 inhibition. Asian Pac J Cancer Prev 2015; 15:5455-61. [PMID: 25041018 DOI: 10.7314/apjcp.2014.15.13.5455] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Saponins are a major active component for the traditional Chinese medicine, Rubus parvifolius L., which has shown clear antitumor activities. However, the specific effects and mechanisms of saponins of Rubus parvifolius L. (SRP) remain unclear with regard to human chronic myeloid leukemia cells. The aim of this study was to investigate inhibition of proliferation and apoptosis induction effects of SRP in K562 cells and further elucidate its regulatory mechanisms. MATERIALS AND METHODS K562 cells were treated with different concentrations of SRP and MTT assays were performed to determine cell viability. Apoptosis induction by SRP was determined with FACS and DAPI staining analysis. Western blotting was used to detect expression of apoptosis and survival related genes. Specific inhibitors were added to confirm roles of STAT3 and AMPK pathways in SRP induction of apoptosis. RESULTS Our results indicated that SRP exhibited obvious inhibitory effects on the growth of K562 cells, and significantly induced apoptosis. Cleavage of pro-apoptotic proteins was dramatically increased after SRP exposure. SRP treatment also increased the activities of AMPK and JNK pathways, and inhibited the phosphorylation expression level of STAT3 in K562 cells. Inhibition of the AMPK pathway blocked the activation of JNK by SRP, indicating that SRP regulated the expression of JNK dependent on the AMPK pathway. Furthermore, inhibition of the latter significantly conferred resistance to SRP pro- apoptotic activity, suggesting involvement of the AMPK pathway in induction of apoptosis. Pretreatment with a STAT3 inhibitor also augmented SRP induced growth inhibition and cell apoptosis, further confirming roles of the STAT3 pathway after SRP treatment. CONCLUSIONS Our results demonstrated that SRP induce cell apoptosis through AMPK activation and STAT3 inhibition in K562 cells. This suggests the possibility of further developing SRP as an alternative treatment option, or perhaps using it as adjuvant chemotherapeutic agent for chronic myeloid leukemia therapy.
Collapse
Affiliation(s)
- Yu-Qing Ge
- Zhejiang Provincial Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China E-mail :
| | | | | | | | | |
Collapse
|
67
|
Liu Y, Luo Y, Li X, Zheng W, Chen T. Rational Design of Selenadiazole Derivatives to Antagonize Hyperglycemia-Induced Drug Resistance in Cancer Cells. Chem Asian J 2015; 10:642-52. [DOI: 10.1002/asia.201403409] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Indexed: 12/22/2022]
|
68
|
Chen B, Li R, Yan N, Chen G, Qian W, Jiang HL, Ji C, Bi ZG. Astragaloside IV controls collagen reduction in photoaging skin by improving transforming growth factor-β/Smad signaling suppression and inhibiting matrix metalloproteinase-1. Mol Med Rep 2015; 11:3344-8. [PMID: 25591734 PMCID: PMC4368092 DOI: 10.3892/mmr.2015.3212] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 11/12/2014] [Indexed: 01/03/2023] Open
Abstract
Exposure to ultraviolet (UV) light reduces levels of type I collagen in the dermis and results in human skin damage and premature skin aging (photoaging). This leads to a wrinkled appearance through the inhibition of transforming growth factor‑β (TGF‑β)/Smad signaling. UV irradiation increases type I collagen degradation through upregulating matrix metalloproteinase (MMP) expression. Astragaloside IV (AST) is one of the major active components extracted from Astragalus membranaceus. However, its multiple anti‑photoaging effects remain to be elucidated. In the present study, the effects of AST against collagen reduction in UV‑induced skin aging in human skin fibroblasts were investigated. The expression of type I procollagen (COL1), MMP‑1, TGF‑βRⅡ and Smad7 were determined using reverse transcription‑polymerase chain reaction, western blotting and ELISA, respectively. UV irradiation inhibits type I collagen production by suppressing the TGF‑β/Smad signaling pathway and increasing COL1 degradation by inducing MMP‑1 expression. Transforming growth factor‑β type II protein and COL1 mRNA decreased but MMP‑1 and Smad7 levels increased in the photoaging model group, which was reversed by topical application of AST. AST prevents collagen reduction from UV irradiation in photoaging skin by improving TGF‑β/Smad signaling suppression and inhibiting MMP‑1, thus AST may be a potential agent against skin photoaging.
Collapse
Affiliation(s)
- Bin Chen
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, P.R. China
| | - Ran Li
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, P.R. China
| | - Ning Yan
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, P.R. China
| | - Gang Chen
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, P.R. China
| | - Wen Qian
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, P.R. China
| | - Hui-Li Jiang
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210024, P.R. China
| | - Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Zhi-Gang Bi
- Department of Dermatology, BenQ Medical Center, Nanjing Medical University, Nanjing, Jiangsu 210019, P.R. China
| |
Collapse
|
69
|
Yang L, Zheng LY, Tian Y, Zhang ZQ, Dong WL, Wang XF, Zhang XY, Cao C. C6 ceramide dramatically enhances docetaxel-induced growth inhibition and apoptosis in cultured breast cancer cells: a mechanism study. Exp Cell Res 2015; 332:47-59. [PMID: 25576381 DOI: 10.1016/j.yexcr.2014.12.017] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/14/2014] [Accepted: 12/26/2014] [Indexed: 01/26/2023]
Abstract
Here we reported that co-administration of docetaxel and a cell-permeable short-chain ceramide (C6) resulted in a striking increase in growth inhibition and apoptosis in primary and transformed breast cells (MCF-7 and MDA-231), which were associated with mitochondrial permeability transition pore (mPTP) opening, a significant reactive oxygen species (ROS) production and the pro-apoptotic AMP-Protein Kinase (AMPK) as well as c-Jun N-terminal kinases (JNK) activations. Contrarily, the mPTP blocker sanglifehrin A (SfA) or the ROS scavenger N-acetyl-l-cysteine (NAC) largely inhibited co-administration-induced cytotoxicity. Further, cyclosporin A (CsA), the inhibitor of cyclophilin-D (Cyp-D, the key mPTP component), as well as Cyp-D RNA silencing also suppressed breast cancer cell death by the co-treatment, while cells overexpressing Cyp-D showed hypersensitivity to docetaxel. Meanwhile, JNK and AMPK inhibition alleviated cell death induced by the co-administration in cultured breast cancer cells. Significantly, C6 ceramide plus docetaxel caused dramatic human epidermal growth factor receptor (HER)-1/-2 degradation and downstream Akt/Erk inhibition in HER-2 expressing MDA-231 cells. These in vitro findings provide confidence in support of further development of C6 ceramide as an adjunct of docetaxel for the treatment of the metastatic breast cancer.
Collapse
Affiliation(s)
- Lan Yang
- Department of Breast Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Li-Yun Zheng
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ye Tian
- Department of Radiotherapy and Oncology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhi-Qing Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215021, China
| | - Wan-Li Dong
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xu-Fen Wang
- Department of Breast Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiao-Ying Zhang
- Department of Cardiothoracic Surgery, the Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Cong Cao
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu 215021, China.
| |
Collapse
|
70
|
Simultaneous active intracellular delivery of doxorubicin and C6-ceramide shifts the additive/antagonistic drug interaction of non-encapsulated combination. J Control Release 2014; 196:122-31. [DOI: 10.1016/j.jconrel.2014.09.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/08/2014] [Accepted: 09/25/2014] [Indexed: 11/21/2022]
|
71
|
Wang S, Qiu J, Shi Z, Wang Y, Chen M. Nanoscale drug delivery for taxanes based on the mechanism of multidrug resistance of cancer. Biotechnol Adv 2014; 33:224-241. [PMID: 25447422 DOI: 10.1016/j.biotechadv.2014.10.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/15/2014] [Accepted: 10/23/2014] [Indexed: 02/07/2023]
Abstract
Taxanes are one type of the most extensively used chemotherapeutic agents to treat cancers. However, their clinical use is severely limited by intrinsic and acquired resistance. A diverse variety of mechanisms has been implicated about taxane resistance, such as alterations of drug targets, overexpression of efflux transporters, defective apoptotic machineries, and barriers in drug transport. The deepening understanding of molecular mechanisms of taxane resistance has spawned a number of targets for reversing resistance. However, circumvention of taxane resistance would not only possess therapeutic potential, but also face with clinical challenge, which accelerates the development of optimal nanoscale delivery systems. This review highlights the current understanding on the mechanisms of taxane resistance, and provides a comprehensive analysis of various nanoscale delivery systems to reverse taxane resistance.
Collapse
Affiliation(s)
- Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jiange Qiu
- Department of Cell Biology and Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhi Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Cell Biology and Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
72
|
Kai S, Lu JH, Hui PP, Zhao H. Pre-clinical evaluation of cinobufotalin as a potential anti-lung cancer agent. Biochem Biophys Res Commun 2014; 452:768-74. [PMID: 25201730 DOI: 10.1016/j.bbrc.2014.08.147] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 08/30/2014] [Indexed: 10/24/2022]
Abstract
Lung cancer is a major cause of cancer-related mortality in the United States and around the world. Due to the pre-existing or acquired chemo-resistance, the current standard chemotherapy regimens only show moderate activity against lung cancer. In the current study, we explored the potential anti-lung cancer activity of cinobufotalin in vivo and in vitro, and studied the underlying mechanisms. We demonstrated that cinobufotalin displayed considerable cytotoxicity against lung cancer cells (A549, H460 and HTB-58 lines) without inducing significant cell apoptosis. Our data suggest that mitochondrial protein cyclophilin D (Cyp-D)-dependent mitochondrial permeability transition pore (mPTP) opening mediates cinobufotalin-induced non-apoptotic death of lung cancer cells. The Cyp-D inhibitor cyclosporine A (CsA), the mPTP blocker sanglifehrin A (SfA), and Cyp-D shRNA-silencing significantly inhibited cinobufotalin-induced mitochondrial membrane potential (MMP) reduction and A549 cell death (but not apoptosis). Using a mice xenograft model, we found that cinobufotalin inhibited A549 lung cancer cell growth in vivo. Thus, cinobufotalin mainly induces Cyp-D-dependent non-apoptotic death in cultured lung cancer cells. The results of this study suggest that cinobufotalin might be further investigated as a novel anti-lung cancer agent.
Collapse
Affiliation(s)
- Sheng Kai
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China
| | - Jia-Huan Lu
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China
| | - Ping-Ping Hui
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China
| | - Hui Zhao
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China.
| |
Collapse
|
73
|
C6 ceramide sensitizes pemetrexed-induced apoptosis and cytotoxicity in osteosarcoma cells. Biochem Biophys Res Commun 2014; 452:72-8. [DOI: 10.1016/j.bbrc.2014.08.065] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/12/2014] [Indexed: 02/07/2023]
|
74
|
Cordycepin down-regulates multiple drug resistant (MDR)/HIF-1α through regulating AMPK/mTORC1 signaling in GBC-SD gallbladder cancer cells. Int J Mol Sci 2014; 15:12778-90. [PMID: 25046749 PMCID: PMC4139874 DOI: 10.3390/ijms150712778] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 06/13/2014] [Accepted: 07/04/2014] [Indexed: 12/12/2022] Open
Abstract
Gallbladder cancer is the most common malignancy of the bile duct, with low 5-year survival rate and poor prognosis. Novel effective treatments are urgently needed for the therapy of this disease. Here, we showed that cordycepin, the bioactive compound in genus Cordyceps, induced growth inhibition and apoptosis in cultured gallbladder cancer cells (Mz-ChA-1, QBC939 and GBC-SD lines). We found that cordycepin inhibited mTOR complex 1 (mTORC1) activation and down-regulated multiple drug resistant (MDR)/hypoxia-inducible factor 1α (HIF-1α) expression through activating of AMP-activated protein kinase (AMPK) signaling in gallbladder cancer GBC-SD cells. Contrarily, AMPKα1-shRNA depletion dramatically inhibited cordycepin-induced molecular changes as well as GBC-SD cell apoptosis. Further, our results showed that co-treatment with a low concentration cordycepin could remarkably enhance the chemosensitivity of GBC-SD cells to gemcitabine and 5-fluorouracil (5-FU), and the mechanism may be attributed to AMPK activation and MDR degradation. In summary, cordycepin induces growth inhibition and apoptosis in gallbladder cancer cells via activating AMPK signaling. Cordycepin could be a promising new drug or chemo-adjuvant for gallbladder cancer.
Collapse
|
75
|
Zhao Z, Yin JQ, Wu MS, Song G, Xie XB, Zou C, Tang Q, Wu Y, Lu J, Wang Y, Wang J, Kang T, Jia Q, Shen J. Dihydromyricetin activates AMP-activated protein kinase and P38(MAPK) exerting antitumor potential in osteosarcoma. Cancer Prev Res (Phila) 2014; 7:927-38. [PMID: 24894198 DOI: 10.1158/1940-6207.capr-14-0067] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Numerous patients with osteosarcoma either are not sensitive to chemotherapy or develop drug resistance to current chemotherapy regimens. Therefore, it is necessary to develop several potentially useful therapeutic agents. Dihydromyricetin is the major flavonoid component derived from Ampelopsis grossedentata, which has a long history of use in food and medicine. The present study examined the antitumor activity both in vitro and in vivo without noticeable side effects and the underlying mechanism of action of dihydromyricetin in osteosarcoma cells. We found that dihydromyricetin induced increased p21 expression and G2-M cell-cycle arrest, caused DNA damage, activated ATM-CHK2-H2AX signaling pathways, and induced apoptosis in osteosarcoma cells as well as decreasing the sphere formation capability by downregulating Sox2 expression. Mechanistic analysis showed that the antitumor potential of dihydromyricetin may be due to the activation of AMPKα and p38(MAPK), as the activating AMPKα led to the inactivation of GSK3β in osteosarcoma cells. Moreover, GSK3β deletion or GSK3β inhibition by LiCl treatment resulted in increased p21 expression and reduced Sox2 expression in osteosarcoma cells. Taken together, our results strongly indicate that the antitumor potential of dihydromyricetin is correlated with P38(MAPK) and the AMPKα-GSK3β-Sox2 signaling pathway. Finally, immunohistochemical analysis indicated that some patients had a lower p-AMPK expression after chemotherapy, which supports that the combination of dihydromyricetin and chemotherapy drug will be beneficial for patients with osteosarcoma. In conclusion, our results are the first to suggest that dihydromyricetin may be a therapeutic candidate for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Zhiqiang Zhao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Jun-qiang Yin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Man-si Wu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou; and
| | - Guohui Song
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Xian-biao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Changye Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou; and
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Yongqian Wang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Jin Wang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou; and
| | - Qiang Jia
- The Institute of Biology, Guizhou Academy of Sciences, Guiyang, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University;
| |
Collapse
|
76
|
Don AS, Lim XY, Couttas TA. Re-configuration of sphingolipid metabolism by oncogenic transformation. Biomolecules 2014; 4:315-53. [PMID: 24970218 PMCID: PMC4030989 DOI: 10.3390/biom4010315] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 02/11/2014] [Accepted: 02/27/2014] [Indexed: 12/15/2022] Open
Abstract
The sphingolipids are one of the major lipid families in eukaryotes, incorporating a diverse array of structural variants that exert a powerful influence over cell fate and physiology. Increased expression of sphingosine kinase 1 (SPHK1), which catalyses the synthesis of the pro-survival, pro-angiogenic metabolite sphingosine 1-phosphate (S1P), is well established as a hallmark of multiple cancers. Metabolic alterations that reduce levels of the pro-apoptotic lipid ceramide, particularly its glucosylation by glucosylceramide synthase (GCS), have frequently been associated with cancer drug resistance. However, the simple notion that the balance between ceramide and S1P, often referred to as the sphingolipid rheostat, dictates cell survival contrasts with recent studies showing that highly potent and selective SPHK1 inhibitors do not affect cancer cell proliferation or survival, and studies demonstrating higher ceramide levels in some metastatic cancers. Recent reports have implicated other sphingolipid metabolic enzymes such as acid sphingomyelinase (ASM) more strongly in cancer pathogenesis, and highlight lysosomal sphingolipid metabolism as a possible weak point for therapeutic targeting in cancer. This review describes the evidence implicating different sphingolipid metabolic enzymes and their products in cancer pathogenesis, and suggests how newer systems-level approaches may improve our overall understanding of how oncogenic transformation reconfigures sphingolipid metabolism.
Collapse
Affiliation(s)
- Anthony S Don
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Xin Y Lim
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Timothy A Couttas
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
77
|
Wang M, Yu T, Zhu C, Sun H, Qiu Y, Zhu X, Li J. Resveratrol triggers protective autophagy through the ceramide/Akt/mTOR pathway in melanoma B16 cells. Nutr Cancer 2014; 66:435-40. [PMID: 24579778 DOI: 10.1080/01635581.2013.878738] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Resveratrol (3,5,4'-trihydroxy-trans-stilbene, RSV), a natural polyphenolic compound, is known as a promising anti-cancer agent. In this study, we showed that RSV could inhibit the growth of B16 cells via induction of apoptosis. Moreover, our results showed for the first time that RSV induced autophagy in B16 cells, which might occur through ceramide accumulation and Akt/mTOR pathway inhibition. Inhibition of autophagy by an autophagic inhibitor 3-methyladenine (3-MA) or si-Beclin 1 enhanced RSV-induced cytotoxicity and apoptosis. Thus, autophagy inhibition represents a promising approach to improve the efficacy of RSV in the treatment of patients with melanoma.
Collapse
Affiliation(s)
- Meng Wang
- a Department of Oncology , Shandong Jining No.1 People's Hospital , Jining , China
| | | | | | | | | | | | | |
Collapse
|
78
|
Hu M, Huang H, Zhao R, Li P, Li M, Miao H, Chen N, Chen M. AZD8055 induces cell death associated with autophagy and activation of AMPK in hepatocellular carcinoma. Oncol Rep 2014; 31:649-56. [PMID: 24297300 DOI: 10.3892/or.2013.2890] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/07/2013] [Indexed: 02/07/2023] Open
Abstract
AZD8055 is a potent inhibitor of mTORC1 and mTOR2 and shows inhibitory effects in several types of cancer cells in vitro and in vivo. However, the effect of AZD8055 on hepatocellular carcinoma (HCC) cells has not been studied. We report that AZD8055 inhibits cell proliferation and colony formation of Hep3B and Huh7 cells but does not cause PARP cleavage, or caspase activation, suggesting that classical apoptosis is not its main mechanism of cell death. By contrast, AZD8055-induced cell death was associated with several characteristics of autophagy, including an increase in acidic vesicular organelle content, conversion of cytosolic LC3-I to membrane-bound LC3-II and elevation of the levels of Atg-5/12, BECN1 and LC3-II. Inhibition of autophagy by 3-methyladenine (3-MA) partially inhibited AZD8055-induced cell death. Furthermore, AZD8055 caused the activation of AMPK and co-treatment with the AMPK inhibitor dorsomorphin also caused a partial but significant reduction of AZD8055-induced cell death. In conclusion, AZD8055-induced HCC cell death is associated with induction of autophagy and activation of AMPK.
Collapse
Affiliation(s)
- Min Hu
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Haili Huang
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Rui Zhao
- Institute of Genetic Engineering, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Peng Li
- Clinical Research Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Mingyi Li
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Huilai Miao
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Nianping Chen
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| | - Ming Chen
- Laboratory of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
79
|
Yang W, Park IJ, Yun H, Im DU, Ock S, Kim J, Seo SM, Shin HY, Viollet B, Kang I, Choe W, Kim SS, Ha J. AMP-activated protein kinase α2 and E2F1 transcription factor mediate doxorubicin-induced cytotoxicity by forming a positive signal loop in mouse embryonic fibroblasts and non-carcinoma cells. J Biol Chem 2014; 289:4839-52. [PMID: 24398673 DOI: 10.1074/jbc.m113.496315] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Doxorubicin is one of the most widely used anti-cancer drugs, but its clinical application is compromised by severe adverse effects in different organs including cardiotoxicity. In the present study we explored mechanisms of doxorubicin-induced cytotoxicity by revealing a novel role for the AMP-activated protein kinase α2 (AMPKα2) in mouse embryonic fibroblasts (MEFs). Doxorubicin robustly induced the expression of AMPKα2 in MEFs but slightly reduced AMPKα1 expression. Our data support the previous notion that AMPKα1 harbors survival properties under doxorubicin treatment. In contrast, analyses of Ampkα2(-/-) MEFs, gene knockdown of AMPKα2 by shRNA, and inhibition of AMPKα2 activity with an AMPK inhibitor indicated that AMPKα2 functions as a pro-apoptotic molecule under doxorubicin treatment. Doxorubicin induced AMPKα2 at the transcription level via E2F1, a transcription factor that regulates apoptosis in response to DNA damage. E2F1 directly transactivated the Ampkα2 gene promoter. In turn, AMPKα2 significantly contributed to stabilization and activation of E2F1 by doxorubicin, forming a positive signal amplification loop. AMPKα2 directly interacted with and phosphorylated E2F1. This signal loop was also detected in H9c2, C2C12, and ECV (human epithelial cells) cells as well as mouse liver under doxorubicin treatment. Resveratrol, which has been suggested to attenuate doxorubicin-induced cytotoxicity, significantly blocked induction of AMPKα2 and E2F1 by doxorubicin, leading to protection of these cells. This signal loop appears to be non-carcinoma-specific because AMPKα2 was not induced by doxorubicin in five different tested cancer cell lines. These results suggest that AMPKα2 may serve as a novel target for alleviating the cytotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Wookyeom Yang
- From the Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Truman JP, García-Barros M, Obeid LM, Hannun YA. Evolving concepts in cancer therapy through targeting sphingolipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:1174-88. [PMID: 24384461 DOI: 10.1016/j.bbalip.2013.12.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/18/2013] [Accepted: 12/21/2013] [Indexed: 12/29/2022]
Abstract
Traditional methods of cancer treatment are limited in their efficacy due to both inherent and acquired factors. Many different studies have shown that the generation of ceramide in response to cytotoxic therapy is generally an important step leading to cell death. Cancer cells employ different methods to both limit ceramide generation and to remove ceramide in order to become resistant to treatment. Furthermore, sphingosine kinase activity, which phosphorylates sphingosine the product of ceramide hydrolysis, has been linked to multidrug resistance, and can act as a strong survival factor. This review will examine several of the most frequently used cancer therapies and their effect on both ceramide generation and the mechanisms employed to remove it. The development and use of inhibitors of sphingosine kinase will be focused upon as an example of how targeting sphingolipid metabolism may provide an effective means to improve treatment response rates and reduce associated treatment toxicity. This article is part of a Special Issue entitled Tools to study lipid functions.
Collapse
Affiliation(s)
- Jean-Philip Truman
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794 Stony Brook, NY, USA.
| | - Mónica García-Barros
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794 Stony Brook, NY, USA.
| | - Lina M Obeid
- Northport Veterans Affairs Medical Center, Northport, NY 11768, USA; Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794 Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794 Stony Brook, NY, USA.
| |
Collapse
|
81
|
Tu Y, Ji C, Yang B, Yang Z, Gu H, Lu CC, Wang R, Su ZL, Chen B, Sun WL, Xia JP, Bi ZG, He L. DNA-dependent protein kinase catalytic subunit (DNA-PKcs)-SIN1 association mediates ultraviolet B (UVB)-induced Akt Ser-473 phosphorylation and skin cell survival. Mol Cancer 2013; 12:172. [PMID: 24365180 PMCID: PMC3922905 DOI: 10.1186/1476-4598-12-172] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/10/2013] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The exposure of skin keratinocytes to Ultraviolet (UV) irradiation leads to Akt phosphorylation at Ser-473, which is important for the carcinogenic effects of excessive sun exposure. The present study investigated the underlying mechanism of Akt Ser-473 phosphorylation by UVB radiation. RESULTS We found that DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and mammalian target of rapamycin (mTOR) complex 2 (mTORC2) were both required for UVB-induced Akt Ser-473 phosphorylation in keratinocytes. Inhibition of DNA-PKcs activity via its inhibitor NU7026, a dominant-negative kinase-dead mutation, RNA interference (RNAi) or gene depletion led to the attenuation of UVB-induced Akt Ser-473 phosphorylation. Meanwhile, siRNA silencing or gene depletion of SIN1, a key component of mTORC2, abolished Akt Ser-473 phosphorylation by UVB. Significantly, we discovered that DNA-PKcs was associated with SIN1 in cytosol upon UVB radiation, and this complexation appeared required for Akt Ser-473 phosphorylation. Meanwhile, this DNA-PKcs-SIN1 complexation by UVB was dependent on epidermal growth factor receptor (EGFR) activation, and was disrupted by an EGFR inhibitor (AG1478) or by EGFR depletion. UVB-induced complexation between DNA-PKcs and mTORC2 components was also abolished by NU7026 and DNA-PKcs mutation. Finally, we found that both DNA-PKcs and SIN1 were associated with apoptosis resistance of UVB radiation, and inhibition of them by NU7026 or genetic depletion significantly enhanced UVB-induced cell death and apoptosis. CONCLUSION Taken together, these results strongly suggest that DNA-PKcs-mTORC2 association is required for UVB-induced Akt Ser-473 phosphorylation and cell survival, and might be important for tumor cell transformation.
Collapse
Affiliation(s)
| | - Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Kunming Medical University, Yunnan Provincial Institute of Dermatology, Kunming 650032, Yunnan, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Doxorubicin, DNA torsion, and chromatin dynamics. Biochim Biophys Acta Rev Cancer 2013; 1845:84-9. [PMID: 24361676 DOI: 10.1016/j.bbcan.2013.12.002] [Citation(s) in RCA: 336] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/16/2013] [Accepted: 12/10/2013] [Indexed: 11/23/2022]
Abstract
Doxorubicin is one of the most important anti-cancer chemotherapeutic drugs, being widely used for the treatment of solid tumors and acute leukemias. The action of doxorubicin and other anthracycline drugs has been intensively investigated during the last several decades, but the mechanisms that have been proposed for cell killing remain disparate and controversial. In this review, we examine the proposed models for doxorubicin action from the perspective of the chromatin landscape, which is altered in many types of cancer due to recurrent mutations in chromatin modifiers. We highlight recent evidence for effects of anthracyclines on DNA torsion and chromatin dynamics that may underlie basic mechanisms of doxorubicin-mediated cell death and suggest new therapeutic strategies for cancer treatment.
Collapse
|
83
|
Sanli T, Steinberg GR, Singh G, Tsakiridis T. AMP-activated protein kinase (AMPK) beyond metabolism: a novel genomic stress sensor participating in the DNA damage response pathway. Cancer Biol Ther 2013; 15:156-69. [PMID: 24100703 DOI: 10.4161/cbt.26726] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
AMP-activated protein kinase (AMPK), an established metabolic stress sensor, has gained popularity in cancer biology due to its ability to control cellular growth and mediate cell cycle checkpoints in cancer cells in response to low energy levels. AMPK is a key effector of the tumor suppressor liver kinase B 1 (LKB1) which inhibits the cellular growth mediator mammalian target of rapamycin (mTOR) and activates checkpoint mediators such as p53 and the cyclin dependent kinase inhibitors p21(cip1) and p27(kip1). However, recent work describes a novel function for AMPK as a sensor of genomic stress and a participant of the DNA damage response (DDR) pathway. Ionizing radiation and chemotherapy activate AMPK in cancer cells to mediate signal transduction downstream of ataxia telangiectasia mutated (ATM) to activate p53- p21(cip1)/p27(kip1) and inhibit mTOR. We discuss evidence on the transcriptional and post-translational regulation of AMPK by ionizing radiation and the role of the enzyme as a mediator of chemo- and radiation sensitivity in epithelial cancer cells. Furthermore, we review data on the participation of AMPK in cytokinesis and observations suggesting a physical association of this enzyme with the mitotic apparatus. The evidence available to date suggests that AMPK is a point of convergence of metabolic and genomic stress signals, which (1) control the activity of growth mediators, (2) propagate DDR, and (3) mediate the anti-proliferative effects of common cytotoxic cancer therapy such as radiation and chemotherapy. This highlights the importance of targeting AMPK with novel cancer therapeutics.
Collapse
Affiliation(s)
- Toran Sanli
- Translational Radiation Biology Laboratory; Juravinski Cancer Center; Hamilton, ON Canada; Department of Oncology; McMaster University; Hamilton, ON Canada
| | | | - Gurmit Singh
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton, ON Canada
| | - Theodoros Tsakiridis
- Translational Radiation Biology Laboratory; Juravinski Cancer Center; Hamilton, ON Canada; Department of Oncology; McMaster University; Hamilton, ON Canada
| |
Collapse
|
84
|
Qin LS, Yu ZQ, Zhang SM, Sun G, Zhu J, Xu J, Guo J, Fu LS. The short chain cell-permeable ceramide (C6) restores cell apoptosis and perifosine sensitivity in cultured glioblastoma cells. Mol Biol Rep 2013; 40:5645-55. [DOI: 10.1007/s11033-013-2666-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 09/14/2013] [Indexed: 12/26/2022]
|
85
|
Adiseshaiah PP, Clogston JD, McLeland CB, Rodriguez J, Potter TM, Neun BW, Skoczen SL, Shanmugavelandy SS, Kester M, Stern ST, McNeil SE. Synergistic combination therapy with nanoliposomal C6-ceramide and vinblastine is associated with autophagy dysfunction in hepatocarcinoma and colorectal cancer models. Cancer Lett 2013; 337:254-65. [PMID: 23664889 PMCID: PMC3722309 DOI: 10.1016/j.canlet.2013.04.034] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 03/14/2013] [Accepted: 04/29/2013] [Indexed: 11/24/2022]
Abstract
Autophagy, a catabolic survival pathway, is gaining attention as a potential target in cancer. In human liver and colon cancer cells, treatment with an autophagy inducer, nanoliposomal C6-ceramide, in combination with the autophagy maturation inhibitor, vinblastine, synergistically enhanced apoptotic cell death. Combination treatment resulted in a marked increase in autophagic vacuole accumulation and decreased autophagy maturation, without diminution of the autophagy flux protein P62. In a colon cancer xenograft model, a single intravenous injection of the drug combination significantly decreased tumor growth in comparison to the individual treatments. Most importantly, the combination treatment did not result in increased toxicity as assessed by body weight loss. The mechanism of combination treatment-induced cell death both in vitro and in vivo appeared to be apoptosis. Supportive of autophagy flux blockade as the underlying synergy mechanism, treatment with other autophagy maturation inhibitors, but not autophagy initiation inhibitors, were similarly synergistic with C6-ceramide. Additionally, knockout of the autophagy protein Beclin-1 suppressed combination treatment-induced apoptosis in vitro. In conclusion, in vitro and in vivo data support a synergistic antitumor activity of the nanoliposomal C6-ceramide and vinblastine combination, potentially mediated by an autophagy mechanism.
Collapse
Affiliation(s)
- Pavan P. Adiseshaiah
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Jeffrey D. Clogston
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Christopher B. McLeland
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Jamie Rodriguez
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Timothy M. Potter
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Barry W. Neun
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Sarah L. Skoczen
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | | | - Mark Kester
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Stephan T. Stern
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Scott E. McNeil
- Nanotechnology Characterization Laboratory, Advanced Technology Program, SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| |
Collapse
|
86
|
Park B, Lee YM, Kim JS, Her Y, Kang JH, Oh SH, Kim HM. Neutral sphingomyelinase 2 modulates cytotoxic effects of protopanaxadiol on different human cancer cells. Altern Ther Health Med 2013; 13:194. [PMID: 23889969 PMCID: PMC3729373 DOI: 10.1186/1472-6882-13-194] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 07/16/2013] [Indexed: 11/25/2022]
Abstract
Background Some of ginsenosides, root extracts from Panax ginseng, exert cytotoxicity against cancer cells through disruption of membrane subdomains called lipid rafts. Protopanaxadiol (PPD) exhibits the highest cytotoxic effect among 8 ginsenosides which we evaluated for anti-cancer activity. We investigated if PPD disrupts lipid rafts in its cytotoxic effects and also the possible mechanisms. Methods Eight ginsenosides were evaluated using different cancer cells and cell viability assays. The potent ginsenoside, PPD was investigated for its roles in lipid raft disruption and downstream pathways to apoptosis of cancer cells. Anti-cancer effects of PPD was also investigated in vivo using mouse xenograft model. Results PPD consistently exerts its potent cytotoxicity in 2 cell survival assays using 5 different cancer cell lines. PPD disrupts lipid rafts in different ways from methyl-β-cyclodextrin (MβCD) depleting cholesterol out of the subdomains, since lipid raft proteins were differentially modulated by the saponin. During disruption of lipid rafts, PPD activated neutral sphingomyelinase 2 (nSMase 2) hydrolyzing membrane sphingomyelins into pro-apoptotic intracellular ceramides. Furthermore, PPD demonstrated its anti-cancer activities against K562 tumor cells in mouse xenograft model, confirming its potential as an adjunct or chemotherapeutic agent by itself in vivo. Conclusions This study demonstrates that neutral sphingomyelinase 2 is responsible for the cytotoxicity of PPD through production of apoptotic ceramides from membrane sphingomyelins. Thus neutral sphingomyelinase 2 and its relevant mechanisms may potentially be employed in cancer chemotherapies.
Collapse
|
87
|
Wang B, Wang XB, Chen LY, Huang L, Dong RZ. Belinostat-induced apoptosis and growth inhibition in pancreatic cancer cells involve activation of TAK1-AMPK signaling axis. Biochem Biophys Res Commun 2013; 437:1-6. [PMID: 23743198 DOI: 10.1016/j.bbrc.2013.05.090] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer accounts for more than 250,000 deaths worldwide each year. Recent studies have shown that belinostat, a novel pan histone deacetylases inhibitor (HDACi) induces apoptosis and growth inhibition in pancreatic cancer cells. However, the underlying mechanisms are not fully understood. In the current study, we found that AMP-activated protein kinase (AMPK) activation was required for belinostat-induced apoptosis and anti-proliferation in PANC-1 pancreatic cancer cells. A significant AMPK activation was induced by belinostat in PANC-1 cells. Inhibition of AMPK by RNAi knockdown or dominant negative (DN) mutation significantly inhibited belinostat-induced apoptosis in PANC-1 cells. Reversely, AMPK activator AICAR and A-769662 exerted strong cytotoxicity in PANC-1 cells. Belinostat promoted reactive oxygen species (ROS) production in PANC-1 cells, increased ROS induced transforming growth factor-β-activating kinase 1 (TAK1)/AMPK association to activate AMPK. Meanwhile, anti-oxidants N-Acetyl-Cysteine (NAC) and MnTBAP as well as TAK1 shRNA knockdown suppressed belinostat-induced AMPK activation and PANC-1 cell apoptosis. In conclusion, we propose that belinostat-induced apoptosis and growth inhibition require the activation of ROS-TAK1-AMPK signaling axis in cultured pancreatic cancer cells.
Collapse
Affiliation(s)
- Bing Wang
- Department of Abdominal Surgical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, China.
| | | | | | | | | |
Collapse
|
88
|
Gambogic acid induces EGFR degradation and Akt/mTORC1 inhibition through AMPK dependent-LRIG1 upregulation in cultured U87 glioma cells. Biochem Biophys Res Commun 2013; 435:397-402. [DOI: 10.1016/j.bbrc.2013.04.099] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 04/29/2013] [Indexed: 11/24/2022]
|
89
|
Yang X, Huang N. Berberine induces selective apoptosis through the AMPK‑mediated mitochondrial/caspase pathway in hepatocellular carcinoma. Mol Med Rep 2013; 8:505-10. [PMID: 23732865 DOI: 10.3892/mmr.2013.1506] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 04/23/2013] [Indexed: 01/11/2023] Open
Abstract
Advanced hepatocellular carcinoma (HCC) is insensitive to traditional chemotherapeutic approaches, which causes difficulty in the development of novel agents for the treatment of HCC. Berberine is the main component of Coptidis Rhizoma, a plant alkaloid with a long history of use in Chinese medicine, and has become a potential candidate for the treatment of HCC due to its high antitumor activity and low toxicity. In this study, we investigated the mechanism via which berberine exerts its inhibitory effects on HCC. The data demonstrated that berberine selectively decreased cell viability in a time‑ and dose‑dependent manner in the HepG2, SMMC‑7721 and Bel‑7402 HCC cell lines compared with normal hepatocytes (HL‑7702 cells), as determined by a sulforhodamine B assay. Flow cytometric analysis revealed that berberine increased the number of late apoptotic cells. Pretreatment with berberine in HepG2 cells resulted in a significant increase in phosphorylated AMP‑activated protein kinase (AMPK), as well as a marked elevation in phosphorylated Akt levels. In addition, the activation of AMPK was accompanied by apoptotic effects that occurred in a caspase‑dependent manner through the mitochondrial pathway, and was coupled with the release of cytochrome c from the mitochondria and the activation of caspase‑9 and ‑3. Furthermore, the ratio of Bax/Bcl‑2 was increased in a dose‑dependent manner. Our study supports the theory that berberine selectively inhibits the growth of human hepatocellular cancer cells by inducing AMPK‑mediated caspase‑dependent mitochondrial pathway cell apoptosis, and rarely causes cytotoxicity in normal cells. Therefore, berberine is a promising novel agent for the treatment of HCC.
Collapse
Affiliation(s)
- Xiaolong Yang
- Research Unit of Infection and Immunity, Department of Pathophysiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | |
Collapse
|
90
|
Chen MB, Zhang Y, Wei MX, Shen W, Wu XY, Yao C, Lu PH. Activation of AMP-activated protein kinase (AMPK) mediates plumbagin-induced apoptosis and growth inhibition in cultured human colon cancer cells. Cell Signal 2013; 25:1993-2002. [PMID: 23712032 DOI: 10.1016/j.cellsig.2013.05.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/18/2013] [Indexed: 10/26/2022]
Abstract
Here we report that activation of AMP-activated protein kinase (AMPK) mediates plumbagin-induced apoptosis and growth inhibition in both primary cultured human colon cancer cells and cell lines. Knocking-down of AMPKα by the target shRNA significantly inhibits plumbagin-induced cytotoxicity in cultured colon cancer cells, while forced activation of AMPK by introducing a constitutively active AMPK (CA-AMPK), or by the AMPK activator, inhibits HT-29 colon cancer cell growth. Our Western-blots and immunoprecipitation (IP) results demonstrate that plumbagin induces AMPK/Apoptosis signal regulating kinase 1 (ASK1)/TNF receptor-associated factor 2 (TRAF2) association to activate pro-apoptotic c-Jun N-terminal kinases (JNK)-p53 signal axis. Further, after plumbagin treatment, activated AMPK directly phosphorylates Raptor to inhibit mTOR complex 1 (mTORC1) activation and Bcl-2 expression in colon cancer cells. Finally, we found that exogenously-added short-chain ceramide (C6) enhances plumbagin-induced AMPK activation and facilitates cell apoptosis and growth inhibition. Our results suggest that AMPK might be the key mediator of plumbagin's anti-tumor activity.
Collapse
Affiliation(s)
- Min-Bin Chen
- Department of Medical Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, 91 Qianjin Road, Kunshan 215300, Jiangsu Province, China
| | | | | | | | | | | | | |
Collapse
|
91
|
Yao J, Bi HE, Sheng Y, Cheng LB, Wendu RL, Wang CH, Cao GF, Jiang Q. Ultraviolet (UV) and hydrogen peroxide activate ceramide-ER stress-AMPK signaling axis to promote retinal pigment epithelium (RPE) cell apoptosis. Int J Mol Sci 2013; 14:10355-68. [PMID: 23685869 PMCID: PMC3676843 DOI: 10.3390/ijms140510355] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 04/15/2013] [Accepted: 05/02/2013] [Indexed: 12/25/2022] Open
Abstract
Ultraviolet (UV) radiation and reactive oxygen species (ROS) impair the physiological functions of retinal pigment epithelium (RPE) cells by inducing cell apoptosis, which is the main cause of age-related macular degeneration (AMD). The mechanism by which UV/ROS induces RPE cell death is not fully addressed. Here, we observed the activation of a ceramide-endoplasmic reticulum (ER) stress-AMP activated protein kinase (AMPK) signaling axis in UV and hydrogen peroxide (H2O2)-treated RPE cells. UV and H2O2 induced an early ceramide production, profound ER stress and AMPK activation. Pharmacological inhibitors against ER stress (salubrinal), ceramide production (fumonisin B1) and AMPK activation (compound C) suppressed UV- and H2O2-induced RPE cell apoptosis. Conversely, cell permeable short-chain C6 ceramide and AMPK activator AICAR (5-amino-1-β-D-ribofuranosyl-imidazole-4-carboxamide) mimicked UV and H2O2’s effects and promoted RPE cell apoptosis. Together, these results suggest that UV/H2O2 activates the ceramide-ER stress-AMPK signaling axis to promote RPE cell apoptosis.
Collapse
Affiliation(s)
- Jin Yao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
- Authors to whom correspondence should be addressed: E-Mails: (J.Y.); (Q.J.); Tel./Fax: +86-025-8667-7699 (J.Y. & Q.J.)
| | - Hui-E Bi
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
| | - Yi Sheng
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
| | - Li-Bo Cheng
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
- Eye Department, Li-Yang City Hospital of Traditional Chinese Medicine, Li-Yang City 213300, China
| | - Ri-Le Wendu
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
| | - Cheng-Hu Wang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
| | - Guo-Fan Cao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing 210029, China; E-Mails: (H.-E.B.); (Y.S.); (L.-B.C.); (R.-L.W.); (C.-H.W.); (G.-F.C.)
- Authors to whom correspondence should be addressed: E-Mails: (J.Y.); (Q.J.); Tel./Fax: +86-025-8667-7699 (J.Y. & Q.J.)
| |
Collapse
|
92
|
AMP-activated protein kinase (AMPK)/Ulk1-dependent autophagic pathway contributes to C6 ceramide-induced cytotoxic effects in cultured colorectal cancer HT-29 cells. Mol Cell Biochem 2013; 378:171-81. [PMID: 23508272 DOI: 10.1007/s11010-013-1608-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 02/23/2013] [Indexed: 10/27/2022]
Abstract
Colorectal cancer is the second leading cause of cancer-related deaths. Drug resistance and/or off-target toxicity against normal cells limit the effectiveness of current chemotherapies for the treatment of colorectal cancer. In the current study, we studied the potential cytotoxic effects of short-chain and cell-permeable C6 ceramide in cultured colorectal cancer HT-29 cells and focused on the underlying mechanisms. We observed that C6 ceramide-induced HT-29 cell death and growth inhibition in a dose- and time-dependent manner. However, no significant apoptosis was observed in C6 ceramide-treated HT-29 cells. Our data support that autophagy contributed to C6 ceramide-induced cytotoxic effects, as autophagy inhibitors, 3-methyladenine (3-MA) and hydroxychloroquine, inhibited C6 ceramide's effect; however, autophagy activators, everolimus (RAD001) and temsirolimus, mimicked C6 ceramide effects and induced HT-29 cell death. Further, we indentified that AMP-activated protein kinase (AMPK)/Ulk1 signaling was required for autophagy induction by C6 ceramide, and AMPK silencing by a specific short hairpin RNA suppressed C6 ceramide-induced autophagy and cytotoxic effects. Reversely, forced activation of AMPK by its activator AICAR or by genetic manipulation caused autophagic death in HT-29 cells, which was inhibited by 3-MA. Our results suggest that autophagy, but not apoptosis, is a major contributor for C6 ceramide-induced cytotoxic effects in HT-29 cells, and activation of AMPK/Ulk1 is required for the process.
Collapse
|
93
|
He M, Sun HG, Hao JY, Li YL, Yu JK, Yan YY, Zhao L, Li N, Wang Y, Bai XF, Yu ZJ, Zheng ZH, Mi XY, Wang EH, Wei MJ. RNA interference-mediated FANCF silencing sensitizes OVCAR3 ovarian cancer cells to adriamycin through increased adriamycin-induced apoptosis dependent on JNK activation. Oncol Rep 2013; 29:1721-9. [PMID: 23440494 PMCID: PMC3658863 DOI: 10.3892/or.2013.2295] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 02/04/2013] [Indexed: 12/25/2022] Open
Abstract
In the present study, we downregulated FANCF expression by small interfering RNA (siRNA) in OVCAR ovarian cancer cells to address the effects of decreased FANCF expression on the function of the Fanconi anemia (FA)/breast cancer susceptibility gene (BRCA) pathway. Furthermore, we investigated whether this method increases the sensitivity of OVCAR3 cells to adriamycin (ADM) and the possible mechanism(s). We found that silencing of FANCF inactivated the FA/BRCA pathway by decreasing the monoubiquitination and focus formation of FANCD2 and reduced the function of the FA/BRCA pathway, resulting in the inhibition of cell proliferation, increased cell apoptosis and DNA damage in OVCAR3 cells. Moreover, we observed that silencing of FANCF enhanced the antiproliferative effect of ADM in OVCAR3 cells and increased ADM intracellular accumulation consequently sensitizing OVCAR3 cells to ADM. Furthermore, silencing of FANCF increased cell apoptosis of OVCAR3 cells which was caused by decreased mitochondrial membrane potential (MMP)-induced DNA damage, activated Jun N-terminal kinase (JNK), increased release of cytochrome c, increased expression of cleaved caspase-3 and poly(ADP-ribose) polymerase (PARP) dependent on JNK activation following treatment of ADM. Collectively, we confirm that silencing of FANCF sensitizes OVCAR3 ovarian cancer cells to ADM, suggesting that FANCF may serve as a potential target for therapeutic strategies in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Miao He
- Department of Pharmacology, China Medical University, Heping, Shenyang, Liaoning 110001, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Chen Z, Shen X, Shen F, Zhong W, Wu H, Liu S, Lai J. TAK1 activates AMPK-dependent cell death pathway in hydrogen peroxide-treated cardiomyocytes, inhibited by heat shock protein-70. Mol Cell Biochem 2013; 377:35-44. [PMID: 23378049 DOI: 10.1007/s11010-013-1568-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/18/2013] [Indexed: 12/25/2022]
Abstract
The aim of this current study is to investigate the potential role of AMP-activated protein kinase (AMPK) in hydrogen peroxide (H2O2)-induced cardiomyocyte death, and focused on the signaling mechanisms of AMPK activation by H2O2. We observed a significant AMPK activation in H2O2-treated cardiomyocytes (both primary cells and H9c2 line). Inhibition of AMPK by its inhibitor or RNAi-reduced H2O2-induced cardiomyocyte death. We here proposed that transforming growth factor-β-activating kinase 1 (TAK1) might be the upstream kinase for AMPK activation by H2O2. H2O2-induced TAK1 activation, which recruited and activated AMPK. TAK1 inhibitor significantly suppressed H2O2-induced AMPK activation and following cardiomyocyte death, while over-expression of TAK1-facilitated AMPK activation and aggregated cardiomyocyte death. Importantly, heat shock protein-70 (HSP-70)-reduced H2O2-induced reactive oxygen species (ROS) accumulation, the TAK1/AMPK activation and cardiomyocyte death. In conclusion, we here suggest that TAK1 activates AMPK-dependent cell death pathway in H2O2-treated cardiomyocytes, and HSP-70 inhibits the signaling pathway by reducing ROS content.
Collapse
Affiliation(s)
- Zhiyu Chen
- Division of Anesthesia and Critical Care Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
95
|
Deguelin induces both apoptosis and autophagy in cultured head and neck squamous cell carcinoma cells. PLoS One 2013; 8:e54736. [PMID: 23372762 PMCID: PMC3553079 DOI: 10.1371/journal.pone.0054736] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 12/14/2012] [Indexed: 12/16/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) represents more than 5% of all cancers diagnosed annually in United States and around the world. Despite advances in the management of patients with this disease, the survival has not been significantly improved, and the search for potential alternative therapies is encouraging. Here we demonstrate that deguelin administration causes a significant HNSCC cell death. Deguelin induces both cell apoptosis and autophagy by modulating multiple signaling pathways in cultured HNSCC cells. Deguelin inhibits Akt signaling, and down-regulates survivin and cyclin-dependent kinase 4 (Cdk4) expressions, by disrupting their association with heat shock protein-90 (Hsp-90). Deguelin induces ceramide production through de novo synthase pathway to promote HNSCC cell death. Importantly, increased ceramide level activates AMP-activated protein kinase (AMPK), which then directly phosphorylates Ulk1 and eventually leads to cell autophagy. We found that a low dose of deguelin sensitized HNSCC cells to 5-FU. Finally, using a nude mice Hep-2 xenograft model, we also showed a significant anti-tumor ability of deguelin in vivo. Together, we suggest that deguelin may represent a novel and effective chemo-agent against HNSCC.
Collapse
|
96
|
Wu CH, Cao C, Kim JH, Hsu CH, Wanebo HJ, Bowen WD, Xu J, Marshall J. Trojan-horse nanotube on-command intracellular drug delivery. NANO LETTERS 2012; 12:5475-5480. [PMID: 23030797 PMCID: PMC4106035 DOI: 10.1021/nl301865c] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
A major challenge to nanomaterial-based medicine is the ability to release drugs on-command. Here, we describe an innovative drug delivery system based on carbon nanotubes (CNTs), in which compounds can be released inside cells from within the nanotube "on-command" by inductive heating with an external alternating current or pulsed magnetic field. Without inductive heating the drug remains safely inside the CNTs, showing no toxicity in cell viability tests. Similar to the "Trojan-Horse" in function, we demonstrate the delivery of a combination of chemotherapeutic agents with low aqueous solubility, paclitaxel (Taxol), and C6-ceramide, to multidrug resistant pancreatic cancer cells. Nanotube encapsulation permitted the drugs to be used at a 100-fold lower concentration compared to exogenous treatment yet achieve a comparable ~70% cancer kill rate.
Collapse
Affiliation(s)
- Chia-Hsuan Wu
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Cong Cao
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island 02912, United States
- The Second Affiliated Hospital of Soochow University, Institute of Neuroscience of Soochow University, Suzhou, Jiangsu, 215000, P.R. China
| | - Jin Ho Kim
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Chih-Hsun Hsu
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Harold J. Wanebo
- Division of Surgical Oncology, Landmark Medical Center, Woonsocket, Rhode Island 02895, United States
| | - Wayne D. Bowen
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island 02912, United States
| | - Jimmy Xu
- School of Engineering, Brown University, Providence, Rhode Island 02912, United States
- World Class University Program, Seoul National University, Seoul 151-742, Korea
| | - John Marshall
- Department of Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
97
|
Wogonin induces apoptosis by activating the AMPK and p53 signaling pathways in human glioblastoma cells. Cell Signal 2012; 24:2216-25. [DOI: 10.1016/j.cellsig.2012.07.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/10/2012] [Accepted: 07/24/2012] [Indexed: 01/14/2023]
|
98
|
Ji C, Yang B, Yang Z, Tu Y, Yang YL, He L, Bi ZG. Ultra-violet B (UVB)-induced skin cell death occurs through a cyclophilin D intrinsic signaling pathway. Biochem Biophys Res Commun 2012; 425:825-9. [PMID: 22892127 DOI: 10.1016/j.bbrc.2012.07.160] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 07/28/2012] [Indexed: 12/22/2022]
Abstract
UVB-induced skin cell damage involves the opening of mitochondrial permeability transition pore (mPTP), which leads to both apoptotic and necrotic cell death. Cyclophilin D (Cyp-D) translocation to the inner membrane of mitochondrion acts as a key component to open the mPTP. Our Western-Blot results in primary cultured human skin keratinocytes and in HaCaT cell line demonstrated that UVB radiation and hydrogen peroxide (H(2)O(2)) induced Cyp-D expression, which was inhibited by anti-oxidant N-acetyl cysteine (NAC). We created a stable Cyp-D deficiency skin keratinocytes by expressing Cyp-D-shRNA through lentiviral infection. Cyp-D-deficient cells were significantly less susceptible than their counterparts to UVB- or H(2)O(2)-induced cell death. Further, cyclosporine A (Cs-A), a Cyp-D inhibitor, inhibited UVB- or H(2)O(2)-induced keratinocytes cell death. Reversely, over-expression of Cyp-D in primary keratinocytes caused spontaneous keratinocytes cell death. These results suggest Cyp-D's critical role in UVB/oxidative stress-induced skin cell death.
Collapse
Affiliation(s)
- Chao Ji
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210024, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
99
|
Luo L, Huang W, Tao R, Hu N, Xiao ZX, Luo Z. ATM and LKB1 dependent activation of AMPK sensitizes cancer cells to etoposide-induced apoptosis. Cancer Lett 2012; 328:114-9. [PMID: 22960274 DOI: 10.1016/j.canlet.2012.08.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 08/10/2012] [Accepted: 08/29/2012] [Indexed: 01/24/2023]
Abstract
The present study aims to determine the effect of AMPK on etoposide-induced apoptosis of cancer cells. Our results revealed that etoposide induced AMPK activation in prostate C4-2 cancer cells, an event that was attenuated by ATM siRNA. In A549 cells that lack LKB1, AMPK was unable to be activated by etoposide, which was restored by introduction of LKB1. Likewise, silencing LKB1 in C4-2 cells impaired AMPK activation. Finally, etoposide displayed a potent pro-apoptotic effect in cancer cells with functional LKB1 and AMPK. Thus, our results establish a linear relationship of ATM, LKB1 and AMPK in response to the DNA damage drug.
Collapse
Affiliation(s)
- Lingyu Luo
- Research Institute of Digestive Diseases, The First Hospital, Nanchang University, Nanchang, Jiangxi, China
| | | | | | | | | | | |
Collapse
|
100
|
Yang YL, Ji C, Cheng L, He L, Lu CC, Wang R, Bi ZG. Sphingosine kinase-1 inhibition sensitizes curcumin-induced growth inhibition and apoptosis in ovarian cancer cells. Cancer Sci 2012; 103:1538-45. [PMID: 22594559 DOI: 10.1111/j.1349-7006.2012.02335.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 05/01/2012] [Accepted: 05/02/2012] [Indexed: 01/11/2023] Open
Abstract
Recent published studies suggest that increasing levels of ceramides enhance the chemo-sensitivity of curcumin. Using in vitro approaches, we analyzed the impact of sphingosine kinase-1 (SphK-1) inhibition on ceramide production, and evaluated SphK1 inhibitor II (SKI-II) as a potential curcumin chemo-sensitizer in ovarian cancer cells. We found that SphK1 is overexpressed in ovarian cancer patients' tumor tissues and in cultured ovarian cancer cell lines. Inhibition of SphK1 by SKI-II or by RNA interference (RNAi) knockdown dramatically enhanced curcumin-induced apoptosis and growth inhibition in ovarian cancer cells. SKI-II facilitated curcumin-induced ceramide production, p38 activation and Akt inhibition. Inhibition of p38 by the pharmacological inhibitor (SB 203580), a dominant-negative expression vector, or by RNAi diminished curcumin and SKI-II co-administration-induced ovarian cancer cell apoptosis. In addition, restoring Akt activation introducing a constitutively active Akt, or inhibiting ceramide production by fumonisin B1 also inhibited the curcumin plus SKI-II co-administration-induced in vitro anti-ovarian cancer effect, suggesting that ceramide accumulation, p38 activation and Akt inhibition are downstream effectors. Our findings suggest that low, well-tolerated doses of SKI-II may offer significant improvement to the clinical curcumin treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yan-li Yang
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|