51
|
Stefanko A, Thiede C, Ehninger G, Simons K, Grzybek M. Lipidomic approach for stratification of acute myeloid leukemia patients. PLoS One 2017; 12:e0168781. [PMID: 28207743 PMCID: PMC5313223 DOI: 10.1371/journal.pone.0168781] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 12/06/2016] [Indexed: 12/19/2022] Open
Abstract
The pathogenesis and progression of many tumors, including hematologic malignancies is highly dependent on enhanced lipogenesis. De novo fatty-acid synthesis permits accelerated proliferation of tumor cells by providing membrane components but these may also alter physicochemical properties of lipid bilayers, which can impact signaling or even increase drug resistance in cancer cells. Cancer type-specific lipid profiles would permit us to monitor and interpret actual effects of lipid changes, potential fingerprints of individual tumors to be explored as diagnostic markers. We have used the shotgun MS approach to identify lipid patterns in different types of acute myeloid leukemia (AML) patients that either show no karyotype change or belong to t(8;21) or inv16 types. Differences in lipidomes of t(8;21) and inv(16) patients, as compared to AML patients without karyotype change, presented mostly as substantial modulation of ceramide/sphingolipid synthesis. Furthermore, between the t(8;21) and all other patients we observed significant changes in physicochemical membrane properties. These were related to a marked alteration in lipid saturation levels. The discovered differences in lipid profiles of various AML types improve our understanding of the pathobiochemical pathways involved and may serve in the development of diagnostic tools.
Collapse
Affiliation(s)
- Adam Stefanko
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Christian Thiede
- Medical Clinic and Polyclinic I, University Hospital TU Dresden, Dresden, Germany
| | - Gerhard Ehninger
- Medical Clinic and Polyclinic I, University Hospital TU Dresden, Dresden, Germany
| | - Kai Simons
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Lipotype GmbH, Dresden, Germany
| | - Michal Grzybek
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Clinic Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- * E-mail:
| |
Collapse
|
52
|
Morad SAF, Davis TS, MacDougall MR, Tan SF, Feith DJ, Desai DH, Amin SG, Kester M, Loughran TP, Cabot MC. Role of P-glycoprotein inhibitors in ceramide-based therapeutics for treatment of cancer. Biochem Pharmacol 2017; 130:21-33. [PMID: 28189725 DOI: 10.1016/j.bcp.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/01/2017] [Indexed: 10/20/2022]
Abstract
The anticancer properties of ceramide, a sphingolipid with potent tumor-suppressor properties, can be dampened via glycosylation, notably in multidrug resistance wherein ceramide glycosylation is characteristically elevated. Earlier works using the ceramide analog, C6-ceramide, demonstrated that the antiestrogen tamoxifen, a first generation P-glycoprotein (P-gp) inhibitor, blocked C6-ceramide glycosylation and magnified apoptotic responses. The present investigation was undertaken with the goal of discovering non-anti-estrogenic alternatives to tamoxifen that could be employed as adjuvants for improving the efficacy of ceramide-centric therapeutics in treatment of cancer. Herein we demonstrate that the tamoxifen metabolites, desmethyltamoxifen and didesmethyltamoxifen, and specific, high-affinity P-gp inhibitors, tariquidar and zosuquidar, synergistically enhanced C6-ceramide cytotoxicity in multidrug resistant HL-60/VCR acute myelogenous leukemia (AML) cells, whereas the selective estrogen receptor antagonist, fulvestrant, was ineffective. Active C6-ceramide-adjuvant combinations elicited mitochondrial ROS production and cytochrome c release, and induced apoptosis. Cytotoxicity was mitigated by introduction of antioxidant. Effective adjuvants markedly inhibited C6-ceramide glycosylation as well as conversion to sphingomyelin. Active regimens were also effective in KG-1a cells, a leukemia stem cell-like line, and in LoVo human colorectal cancer cells, a solid tumor model. In summary, our work details discovery of the link between P-gp inhibitors and the regulation and potentiation of ceramide metabolism in a pro-apoptotic direction in cancer cells. Given the active properties of these adjuvants in synergizing with C6-ceramide, independent of drug resistance status, stemness, or cancer type, our results suggest that the C6-ceramide-containing regimens could provide alternative, promising therapeutic direction, in addition to finding novel, off-label applications for P-gp inhibitors.
Collapse
Affiliation(s)
- Samy A F Morad
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States; Department of Pharmacology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Traci S Davis
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States
| | - Matthew R MacDougall
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States
| | - Su-Fern Tan
- Department of Medicine, Hematology/Oncology, University of Virginia, Charlottesville, VA, United States
| | - David J Feith
- Department of Medicine, Hematology/Oncology, University of Virginia, Charlottesville, VA, United States; University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Dhimant H Desai
- Penn State University College of Medicine, Department of Pharmacology, University Drive, Hershey, PA, United States
| | - Shantu G Amin
- Penn State University College of Medicine, Department of Pharmacology, University Drive, Hershey, PA, United States
| | - Mark Kester
- University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Thomas P Loughran
- Department of Medicine, Hematology/Oncology, University of Virginia, Charlottesville, VA, United States; University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, Greenville, NC, United States.
| |
Collapse
|
53
|
Poon C, Duan X, Chan C, Han W, Lin W. Nanoscale Coordination Polymers Codeliver Carboplatin and Gemcitabine for Highly Effective Treatment of Platinum-Resistant Ovarian Cancer. Mol Pharm 2016; 13:3665-3675. [PMID: 27712076 PMCID: PMC5673481 DOI: 10.1021/acs.molpharmaceut.6b00466] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Due to the ability of ovarian cancer (OCa) to acquire drug resistance, it has been difficult to develop efficient and safe chemotherapy for OCa. Here, we examined the therapeutic use of a new self-assembled core-shell nanoscale coordination polymer nanoparticle (NCP-Carbo/GMP) that delivers high loadings of carboplatin (28.0 ± 2.6 wt %) and gemcitabine monophosphate (8.6 ± 1.5 wt %). A strong synergistic effect was observed between carboplatin and gemcitabine against platinum-resistant OCa cells, SKOV-3 and A2780/CDPP, in vitro. The coadministration of carboplatin and gemcitabine in the NCP led to prolonged blood circulation half-life (11.8 ± 4.8 h) and improved tumor uptake of the drugs (10.2 ± 4.4% ID/g at 24 h), resulting in 71% regression and 80% growth inhibition of SKOV-3 and A2780/CDDP tumors, respectively. Our findings demonstrate that NCP particles provide great potential for the codelivery of multiple chemotherapeutics for treating drug-resistant cancer.
Collapse
Affiliation(s)
| | | | - Christina Chan
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Wenbo Han
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| |
Collapse
|
54
|
Lipidomic Profiling of Lung Pleural Effusion Identifies Unique Metabotype for EGFR Mutants in Non-Small Cell Lung Cancer. Sci Rep 2016; 6:35110. [PMID: 27739449 PMCID: PMC5064315 DOI: 10.1038/srep35110] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022] Open
Abstract
Cytology and histology forms the cornerstone for the diagnosis of non-small cell lung cancer (NSCLC) but obtaining sufficient tumour cells or tissue biopsies for these tests remains a challenge. We investigate the lipidome of lung pleural effusion (PE) for unique metabolic signatures to discriminate benign versus malignant PE and EGFR versus non-EGFR malignant subgroups to identify novel diagnostic markers that is independent of tumour cell availability. Using liquid chromatography mass spectrometry, we profiled the lipidomes of the PE of 30 benign and 41 malignant cases with or without EGFR mutation. Unsupervised principal component analysis revealed distinctive differences between the lipidomes of benign and malignant PE as well as between EGFR mutants and non-EGFR mutants. Docosapentaenoic acid and Docosahexaenoic acid gave superior sensitivity and specificity for detecting NSCLC when used singly. Additionally, several 20- and 22- carbon polyunsaturated fatty acids and phospholipid species were significantly elevated in the EGFR mutants compared to non-EGFR mutants. A 7-lipid panel showed great promise in the stratification of EGFR from non-EGFR malignant PE. Our data revealed novel lipid candidate markers in the non-cellular fraction of PE that holds potential to aid the diagnosis of benign, EGFR mutation positive and negative NSCLC.
Collapse
|
55
|
Chiu WH, Su WC, Li CL, Chen CL, Lin CF. An increase in glucosylceramide synthase induces Bcl-xL-mediated cell survival in vinorelbine-resistant lung adenocarcinoma cells. Oncotarget 2016; 6:20513-24. [PMID: 26001295 PMCID: PMC4653022 DOI: 10.18632/oncotarget.4109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/23/2015] [Indexed: 02/07/2023] Open
Abstract
Reversing drug resistance with concurrent treatment confers anticancer benefits. In this study, we investigated the potential mechanism of glucosylceramide synthase (GCS)-mediated vinca alkaloid vinorelbine (VNR) resistance in human lung adenocarcinoma cells. Compared with PC14PE6/AS2 (AS2) and CL1-0 cells, apoptotic analysis showed that both A549 and CL1-5 cells were VNR-resistant, while these cells highly expressed GCS at the protein level. VNR treatment significantly converts ceramide to glucosylceramide in VNR-resistant cells; however, pharmacologically inhibiting GCS with (±)-threo-1-Phenyl-2-decanoylamino-3-morpholino-1-propanol hydrochloride (PDMP) induced ceramide accumulation, accompanied by a decrease in glucosylceramide. Under concurrent treatment with VNR and PDMP, an increase in cell apoptosis could be identified; furthermore, genetically silencing GCS confirmed these effects. In VNR-resistant cells, Bcl-xL expression was aberrantly increased, while pharmacologically inhibiting Bcl-xL with ABT-737 sensitized cells to VNR-induced apoptosis. Conversely, enforced expression of Bcl-xL strengthened the survival response of the VNR-susceptible cells AS2 and CL1-0. Without changes in mRNA expression, Bcl-xL was overexpressed independent of β-catenin-mediated transcriptional regulation in VNR-resistant cells. Simultaneous GCS inhibition and VNR treatment caused a decrease in Bcl-xL expression. According to these findings, an increase in GCS caused Bcl-xL augmentation, facilitating VNR resistance in lung adenocarcinoma cells.
Collapse
Affiliation(s)
- Wei-Hsin Chiu
- Division of Hemato-Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wu-Chou Su
- Division of Hemato-Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chia-Ling Li
- Division of Hemato-Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chia-Ling Chen
- Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chiou-Feng Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
56
|
Morad SAF, Ryan TE, Neufer PD, Zeczycki TN, Davis TS, MacDougall MR, Fox TE, Tan SF, Feith DJ, Loughran TP, Kester M, Claxton DF, Barth BM, Deering TG, Cabot MC. Ceramide-tamoxifen regimen targets bioenergetic elements in acute myelogenous leukemia. J Lipid Res 2016; 57:1231-42. [PMID: 27140664 PMCID: PMC4918852 DOI: 10.1194/jlr.m067389] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/29/2016] [Indexed: 01/01/2023] Open
Abstract
The objective of our study was to determine the mechanism of action of the short-chain ceramide analog, C6-ceramide, and the breast cancer drug, tamoxifen, which we show coactively depress viability and induce apoptosis in human acute myelogenous leukemia cells. Exposure to the C6-ceramide-tamoxifen combination elicited decreases in mitochondrial membrane potential and complex I respiration, increases in reactive oxygen species (ROS), and release of mitochondrial proapoptotic proteins. Decreases in ATP levels, reduced glycolytic capacity, and reduced expression of inhibitors of apoptosis proteins also resulted. Cytotoxicity of the drug combination was mitigated by exposure to antioxidant. Cells metabolized C6-ceramide by glycosylation and hydrolysis, the latter leading to increases in long-chain ceramides. Tamoxifen potently blocked glycosylation of C6-ceramide and long-chain ceramides. N-desmethyltamoxifen, a poor antiestrogen and the major tamoxifen metabolite in humans, was also effective with C6-ceramide, indicating that traditional antiestrogen pathways are not involved in cellular responses. We conclude that cell death is driven by mitochondrial targeting and ROS generation and that tamoxifen enhances the ceramide effect by blocking its metabolism. As depletion of ATP and targeting the "Warburg effect" represent dynamic metabolic insult, this ceramide-containing combination may be of utility in the treatment of leukemia and other cancers.
Collapse
Affiliation(s)
- Samy A F Morad
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Terence E Ryan
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - P Darrell Neufer
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Tonya N Zeczycki
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Traci S Davis
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Matthew R MacDougall
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Todd E Fox
- Cancer Center, Division of Hematology Oncology, Department of Medicine Department of Pharmacology, University of Virginia, Charlottesville, VA
| | - Su-Fern Tan
- Department of Pharmacology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - David J Feith
- Cancer Center, Division of Hematology Oncology, Department of Medicine Oncology, Department of Medicine
| | - Thomas P Loughran
- Cancer Center, Division of Hematology Oncology, Department of Medicine Department of Pharmacology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Mark Kester
- Cancer Center, Division of Hematology Oncology, Department of Medicine
| | - David F Claxton
- Penn State Hershey Cancer Institute, The Pennsylvania State University, Hershey, PA
| | - Brian M Barth
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH
| | - Tye G Deering
- East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC
| |
Collapse
|
57
|
Ma YY, Mou XZ, Ding YH, Zou H, Huang DS. Delivery systems of ceramide in targeted cancer therapy: ceramide alone or in combination with other anti-tumor agents. Expert Opin Drug Deliv 2016; 13:1397-406. [PMID: 27168034 DOI: 10.1080/17425247.2016.1188803] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Ying-Yu Ma
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
- Medical School and Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Ya-Hui Ding
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Dong-Sheng Huang
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
- Department of Hepatobiliary Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| |
Collapse
|
58
|
Yang J, Yang C, Zhang S, Mei Z, Shi M, Sun S, Shi L, Wang Z, Wang Y, Li Z, Xie C. ABC294640, a sphingosine kinase 2 inhibitor, enhances the antitumor effects of TRAIL in non-small cell lung cancer. Cancer Biol Ther 2016; 16:1194-204. [PMID: 26054751 DOI: 10.1080/15384047.2015.1056944] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Evidences suggest that tumor microenvironment may play an important role in cancer drug resistance. Sphingosine kinase 2 (SphK2) is proposed to be the key regulator of sphingolipid signaling. This study is aimed to investigate whether the combination of molecular targeting therapy using a specific inhibitor of SphK2 (ABC294640), with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can enhance the apoptosis of non-small cell lung cancer (NSCLC) cells. Our results revealed that NSCLC cells' sensitivity to TRAIL is correlated with the level of SphK2. Compared with TRAIL alone, the combination therapy enhanced the apoptosis induced by TRAIL, and knockdown of SphK2 by siRNA presented a similar effect. Combination therapy with ABC294640 increased the activity of caspase-3/8 and up-regulated the expression of death receptors (DR). Additional investigations revealed that translocation of DR4/5 to the cell membrane surface was promoted by adding ABC294640. However, expression of anti-apoptosis proteins such as Bcl(-)2 and IAPs was not significantly modified by this SphK2 inhibitor. Overall, this work demonstrates that SphK2 may contribute to the apoptosis resistance in NSCLC, thus indicating a new therapeutic target for resistant NSCLC cells.
Collapse
Key Words
- ABC294640, 3-(4-chlorophenyl)-adamantane-1-carboxylic acid (pyridin-4-ylmethyl) amide
- Bcl-2, B-cell lymphoma 2
- Cer, ceramide
- DISC, death-induced signaling complex
- DR4, death receptor 4
- DR5, death receptor 5
- MTT, (3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide
- NSCLC
- NSCLC, non-small cell lung cancer
- S1P, sphingosine-1-phosphate
- SphK2, sphingosine kinase 2
- TRAIL
- TRAIL, tumor necrosis factor-related apoptosis inducing ligand
- death receptor
- resistance
- sphingosine kinase 2
Collapse
Affiliation(s)
- Jie Yang
- a Department of Radiation and Medical Oncology ; Zhongnan Hospital of Wuhan University ; Wuhan , PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Li YL, Lin ML, He SQ, Jin JF. Sphingolipid metabolism affects the anticancer effect of cisplatin. World J Transl Med 2016; 5:37-45. [DOI: 10.5528/wjtm.v5.i1.37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/09/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Cisplatin, a DNA crosslinking agent, is widely used for the treatment of a variety of solid tumors. Numerous studies have demonstrated that sphingolipid metabolism, which acts as a target for cisplatin treatment, is a highly complex network that consists of sphingolipid signaling molecules and related catalytic enzymes. Ceramide (Cer), which is the central molecule of this network, has been established to induce apoptosis. However, another molecule, sphingosine-1-phosphate (S1P), exerts the opposite function, i.e., serves as a regulator of pro-survival. Other sphingolipid molecules, including dihydroceramide, ceramide-1-phosphate, glucosylceramide (GluCer), and sphingosine (Sph), or sphingolipid catalytic enzymes such as Sph kinase (SphK), Cer synthase (CerS), and S1P lyase, have also attracted considerable attention, particularly Cer, GluCer, SphK, CerS, and S1P lyase, which have been implicated in cisplatin resistance. This review summarizes specific molecules involved in sphingolipid metabolism and related catalytic enzymes affecting the anticancer effect of cisplatin, particularly in relation to induction of apoptosis and drug resistance.
Collapse
|
60
|
Prognostic value of glucosylceramide synthase and P-glycoprotein expression in oral cavity cancer. Int J Clin Oncol 2016; 21:883-889. [DOI: 10.1007/s10147-016-0973-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/06/2016] [Indexed: 12/16/2022]
|
61
|
The human organic cation transporter OCT1 mediates high affinity uptake of the anticancer drug daunorubicin. Sci Rep 2016; 6:20508. [PMID: 26861753 PMCID: PMC4748219 DOI: 10.1038/srep20508] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/05/2016] [Indexed: 01/11/2023] Open
Abstract
Anthracyclines such as daunorubicin are anticancer agents that are transported into cells, and exert cytotoxicity by blocking DNA metabolism. Although there is evidence for active uptake of anthracyclines into cells, the specific transporter involved in this process has not been identified. Using the high-grade serous ovarian cancer cell line TOV2223G, we show that OCT1 mediated the high affinity (Km ~ 5 μM) uptake of daunorubicin into the cells, and that micromolar amounts of choline completely abolished the drug entry. OCT1 downregulation by shRNA impaired daunorubicin uptake into the TOV2223G cells, and these cells were significantly more resistant to the drug in comparison to the control shRNA. Transfection of HEK293T cells, which accommodated the ectopic expression of OCT1, with a plasmid expressing OCT1-EYFP showed that the transporter was predominantly localized to the plasma membrane. These transfected cells exhibited an increase in the uptake of daunorubicin in comparison to control cells transfected with an empty EYFP vector. Furthermore, a variant of OCT1, OCT1-D474C-EYFP, failed to enhance daunorubicin uptake. This is the first report demonstrating that human OCT1 is involved in the high affinity transport of anthracyclines. We postulate that OCT1 defects may contribute to the resistance of cancer cells treated with anthracyclines.
Collapse
|
62
|
The Emerging Role of Extracellular Vesicle-Mediated Drug Resistance in Cancers: Implications in Advanced Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:454837. [PMID: 26587537 PMCID: PMC4637461 DOI: 10.1155/2015/454837] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 07/05/2015] [Indexed: 01/07/2023]
Abstract
Emerging evidence has shown that the extracellular vesicles (EVs) regulate various biological processes and can control cell proliferation and survival, as well as being involved in normal cell development and diseases such as cancers. In cancer treatment, development of acquired drug resistance phenotype is a serious issue. Recently it has been shown that the presence of multidrug resistance proteins such as Pgp-1 and enrichment of the lipid ceramide in EVs could have a role in mediating drug resistance. EVs could also mediate multidrug resistance through uptake of drugs in vesicles and thus limit the bioavailability of drugs to treat cancer cells. In this review, we discussed the emerging evidence of the role EVs play in mediating drug resistance in cancers and in particular the role of EVs mediating drug resistance in advanced prostate cancer. The role of EV-associated multidrug resistance proteins, miRNA, mRNA, and lipid as well as the potential interaction(s) among these factors was probed. Lastly, we provide an overview of the current available treatments for advanced prostate cancer, considering where EVs may mediate the development of resistance against these drugs.
Collapse
|
63
|
Zhang Y, Talmon G, Wang J. MicroRNA-587 antagonizes 5-FU-induced apoptosis and confers drug resistance by regulating PPP2R1B expression in colorectal cancer. Cell Death Dis 2015; 6:e1845. [PMID: 26247730 PMCID: PMC4558495 DOI: 10.1038/cddis.2015.200] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/21/2015] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
Drug resistance is one of the major hurdles for cancer treatment. However, the underlying mechanisms are still largely unknown and therapeutic options remain limited. In this study, we show that microRNA (miR)-587 confers resistance to 5-fluorouracil (5-FU)-induced apoptosis in vitro and reduces the potency of 5-FU in the inhibition of tumor growth in a mouse xenograft model in vivo. Further studies indicate that miR-587 modulates drug resistance through downregulation of expression of PPP2R1B, a regulatory subunit of the PP2A complex, which negatively regulates AKT activation. Knockdown of PPP2R1B expression increases AKT phosphorylation, which leads to elevated XIAP expression and enhanced 5-FU resistance; whereas rescue of PPP2R1B expression in miR-587-expressing cells decreases AKT phosphorylation/XIAP expression, re-sensitizing colon cancer cells to 5-FU-induced apoptosis. Moreover, a specific and potent AKT inhibitor, MK2206, reverses miR-587-conferred 5-FU resistance. Importantly, studies of colorectal cancer specimens indicate that the expression of miR-587 and PPP2R1B positively and inversely correlates with chemoresistance, respectively, in colorectal cancer. These findings indicate that the miR-587/PPP2R1B/pAKT/XIAP signaling axis has an important role in mediating response to chemotherapy in colorectal cancer. A major implication of our study is that inhibition of miR-587 or restoration of PPP2R1B expression may have significant therapeutic potential to overcome drug resistance in colorectal cancer patients and that the combined use of an AKT inhibitor with 5-FU may increase efficacy in colorectal cancer treatment.
Collapse
Affiliation(s)
- Yang Zhang
- 1] Eppley Institute for Research in Cancer and Allied Diseases, 985950 Nebraska Medical Center, Omaha, NE 68198, USA [2] Department of Genetics, Cell Biology and Anatomy, 985950 Nebraska Medical Center, Omaha, NE 68198, USA
| | - G Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985950 Nebraska Medical Center, Omaha, NE 68198, USA
| | - J Wang
- 1] Eppley Institute for Research in Cancer and Allied Diseases, 985950 Nebraska Medical Center, Omaha, NE 68198, USA [2] Department of Genetics, Cell Biology and Anatomy, 985950 Nebraska Medical Center, Omaha, NE 68198, USA [3] Department of Biochemistry and Molecular Biology, Fred & Pamela Buffett Cancer Center, 985950 Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
64
|
Abstract
Fenretinide, N-(4-hydroxyphenyl)retinamide, (4-HPR), a synthetic retinoid, owes its cancer-toxic effects in part to the generation of ceramide, a potent tumor-suppressing sphingolipid. As such, 4-HPR has garnered considerable interest as a chemotherapeutic. Cancer cells, however, via various metabolic routes, inactivate ceramide, and this can limit 4-HPR efficacy. As relatively little is known regarding 4-HPR-induced ceramide management in acute myelogeneous leukemia (AML), we undertook the present study to evaluate the impact of 4-HPR on ceramide production, metabolism, and cytotoxicity. In KG-1, HL-60, and HL-60/VCR (multidrug resistant) human leukemia cells, 4-HPR induced 15-, 2-, and 20-fold increases in ceramide (measured using [3H]palmitic acid), respectively. By use of specific inhibitors we show that ceramide was produced by sphingomyelinase and de novo pathways in response to 4-HPR exposure. HL-60/VCR cells metabolized ceramide to glucosylceramide (GC). 4-HPR exposure (1.25-10 μM) reduced viability in all cell lines, with approximate IC50's ranging from 1 to 8.0 μM. Reactive oxygen species (ROS) were generated in response to 4-HPR treatment, and the concomitant cytotoxicity was reversed by addition of vitamin E. 4-HPR was not cytotoxic nor did it elicit ceramide formation in K562, a chronic myeloid leukemia cell line; however, K562 cells were sensitive to a cell-deliverable form of ceramide, C6-ceramide. Treatment of Molt-3, an acute lymphoblastic leukemia cell line, with 4-HPR revealed moderate ceramide production (5-fold over control), robust conversion of ceramide to GC and sphingomyelin, and resistance to 4-HPR and C6-ceramide. In conclusion, this work demonstrates diversity within and among leukemia in 4-HPR sensitivity and ceramide generation and subsequent metabolism. As such, knowledge of these metabolic pathways can provide guidance for enhancing ceramide-driven effects of 4-HPR in treatment of leukemia.
Collapse
|
65
|
Su X, Song H, Niu F, Yang K, Kou G, Wang X, Chen H, Li W, Guo S, Li J, Li B, Feng SS, Jiang J, Yin C, Gao J. Co-delivery of doxorubicin and PEGylated C16-ceramide by nanoliposomes for enhanced therapy against multidrug resistance. Nanomedicine (Lond) 2015; 10:2033-50. [PMID: 26084553 DOI: 10.2217/nnm.15.50] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aim: To develop novel nanoliposomes (Lip-ADR-Cer) codelivering doxorubicin (ADR) and PEGylated C16 ceramide (PEG-ceramide C16) to overcome multidrug resistance. Materials & methods: The antitumor activity and mechanism of Lip-ADR-Cer were evaluated. Results & conclusion: The IC50 of Lip-ADR-Cer after 48-h treatment with the MCF-7/ADR and HL-60/ADR cancer cells, both being ADR resistant, was 2.2- and 1.4-fold effective respectively versus the general nanoliposomes with no PEG-ceramide C16 (Lip-ADR). The antitumor assay in mice bearing MCF-7/ADR or HL-60/ADR xenograft tumors confirmed the superior antitumor activity of Lip-ADR-Cer over Lip-ADR. We found that the improved therapeutic effect of Lip-ADR-Cer may be attributed to both of the cytotoxic effect of PEG-ceramide C16 and glucosylceramide synthase overexpression in multidrug resistance cells.
Collapse
Affiliation(s)
- Xiao Su
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Hao Song
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
- Centre for Stem Cell & Regenerative Medicine, Liaocheng People's Hospital, 67 Dongchang West Road, Liaocheng, Shangdong 252000, China
| | - Fangfang Niu
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Kaixuan Yang
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Geng Kou
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
| | - Xiaohang Wang
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Huaiwen Chen
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Wei Li
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
| | - Shangjing Guo
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
| | - Jun Li
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
| | - Bohua Li
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
| | - Si-shen Feng
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
- Department of Chemical & Biomolecular Engineering, National University of Singapore, Block E5, 02–11, 4 Engineering Drive 4, Singapore 117576, Singapore
- Suzhou NanoStar Biopharm Inc Ltd, BioBay, Bld B2, Unit 604, 218 Xing-Hu Street, Suzhou Industrial Park, Suzhou 215123, China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Hubei Province Tumor Hospital, Wuhan, Hubei 430079, China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jie Gao
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
- College of Pharmacy, Liaocheng University, 1 Hu'nan Road, Liaocheng, Shandong 25200, PR China
- Department of Pharmaceutical Sciences, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| |
Collapse
|
66
|
Roh JL, Kim EH, Park JY, Kim JW. Inhibition of Glucosylceramide Synthase Sensitizes Head and Neck Cancer to Cisplatin. Mol Cancer Ther 2015; 14:1907-15. [DOI: 10.1158/1535-7163.mct-15-0171] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/03/2015] [Indexed: 11/16/2022]
|
67
|
Tyler A, Johansson A, Karlsson T, Gudey SK, Brännström T, Grankvist K, Behnam-Motlagh P. Targeting glucosylceramide synthase induction of cell surface globotriaosylceramide (Gb3) in acquired cisplatin-resistance of lung cancer and malignant pleural mesothelioma cells. Exp Cell Res 2015; 336:23-32. [PMID: 26004871 DOI: 10.1016/j.yexcr.2015.05.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND Acquired resistance to cisplatin treatment is a caveat when treating patients with non-small cell lung cancer (NSCLC) and malignant pleural mesothelioma (MPM). Ceramide increases in response to chemotherapy, leading to proliferation arrest and apoptosis. However, a tumour stress activation of glucosylceramide synthase (GCS) follows to eliminate ceramide by formation of glycosphingolipids (GSLs) such as globotriaosylceramide (Gb3), the functional receptor of verotoxin-1. Ceramide elimination enhances cell proliferation and apoptosis blockade, thus stimulating tumor progression. GSLs transactivate multidrug resistance 1/P-glycoprotein (MDR1) and multidrug resistance-associated protein 1 (MRP1) expression which further prevents ceramide accumulation and stimulates drug efflux. We investigated the expression of Gb3, MDR1 and MRP1 in NSCLC and MPM cells with acquired cisplatin resistance, and if GCS activity or MDR1 pump inhibitors would reduce their expression and reverse cisplatin-resistance. METHODS Cell surface expression of Gb3, MDR1 and MRP1 and intracellular expression of MDR1 and MRP1 was analyzed by flow cytometry and confocal microscopy on P31 MPM and H1299 NSCLC cells and subline cells with acquired cisplatin resistance. The effect of GCS inhibitor PPMP and MDR1 pump inhibitor cyclosporin A for 72h on expression and cisplatin cytotoxicity was tested. RESULTS The cisplatin-resistant cells expressed increased cell surface Gb3. Cell surface Gb3 expression of resistant cells was annihilated by PPMP whereas cyclosporin A decreased Gb3 and MDR1 expression in H1299 cells. No decrease of MDR1 by PPMP was noted in using flow cytometry, whereas a decrease of MDR1 in H1299 and H1299res was indicated with confocal microscopy. No certain co-localization of Gb3 and MDR1 was noted. PPMP, but not cyclosporin A, potentiated cisplatin cytotoxicity in all cells. CONCLUSIONS Cell surface Gb3 expression is a likely tumour biomarker for acquired cisplatin resistance of NSCLC and MPM cells. Tumour cell resistance to MDR1 inhibitors of cell surface MDR1 and Gb3 could explain the aggressiveness of NSCLC and MPM. Therapy with GCS activity inhibitors or toxin targeting of the Gb3 receptor may substantially reduce acquired cisplatin drug resistance of NSCLC and MPM cells.
Collapse
Affiliation(s)
- Andreas Tyler
- Department of Medical Biosciences, Umeå University, S-901 85 Umea, Sweden.
| | - Anders Johansson
- Department of Odontology, Umeå University, S-901 85 Umea, Sweden
| | - Terese Karlsson
- Department of Radiation Sciences, Oncology, S-901 85 Umea, Sweden
| | - Shyam Kumar Gudey
- Department of Medical Biosciences, Umeå University, S-901 85 Umea, Sweden
| | - Thomas Brännström
- Department of Medical Biosciences, Umeå University, S-901 85 Umea, Sweden
| | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, S-901 85 Umea, Sweden
| | | |
Collapse
|
68
|
Morad SAF, Cabot MC. Tamoxifen regulation of sphingolipid metabolism--Therapeutic implications. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1134-45. [PMID: 25964209 DOI: 10.1016/j.bbalip.2015.05.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/23/2015] [Accepted: 05/04/2015] [Indexed: 12/25/2022]
Abstract
Tamoxifen, a triphenylethylene antiestrogen and one of the first-line endocrine therapies used to treat estrogen receptor-positive breast cancer, has a number of interesting, off-target effects, and among these is the inhibition of sphingolipid metabolism. More specifically, tamoxifen inhibits ceramide glycosylation, and enzymatic step that can adventitiously support the influential tumor-suppressor properties of ceramide, the aliphatic backbone of sphingolipids. Additionally, tamoxifen and metabolites N-desmethyltamoxifen and 4-hydroxytamoxifen, have been shown to inhibit ceramide hydrolysis by the enzyme acid ceramidase. This particular intervention slows ceramide destruction and thereby depresses formation of sphingosine 1-phosphate, a mitogenic sphingolipid with cancer growth-promoting properties. As ceramide-centric therapies are becoming appealing clinical interventions in the treatment of cancer, agents like tamoxifen that can retard the generation of mitogenic sphingolipids and buffer ceramide clearance via inhibition of glycosylation, take on new importance. In this review, we present an abridged, lay introduction to sphingolipid metabolism, briefly chronicle tamoxifen's history in the clinic, examine studies that demonstrate the impact of triphenylethylenes on sphingolipid metabolism in cancer cells, and canvass works relevant to the use of tamoxifen as adjuvant to drive ceramide-centric therapies in cancer treatment. The objective is to inform the readership of what could be a novel, off-label indication of tamoxifen and structurally-related triphenylethylenes, an indication divorced from estrogen receptor status and one with application in drug resistance.
Collapse
Affiliation(s)
- Samy A F Morad
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, Greenville, NC 27834, USA; East Carolina Diabetes and Obesity Institute, 115 Heart Drive, Greenville, NC 27834, USA; Department of Pharmacology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, Greenville, NC 27834, USA; East Carolina Diabetes and Obesity Institute, 115 Heart Drive, Greenville, NC 27834, USA.
| |
Collapse
|
69
|
Abstract
Parthenolide (PTL) has shown great promise as a novel anti-leukemia agent as it selectively eliminates acute myeloid leukemia (AML) blast cells and leukemia stem cells (LSCs) while sparing normal hematopoietic cells. This success has not yet translated to the clinical setting because PTL is rapidly cleared from blood due to its hydrophobicity. To increase the aqueous solubility of PTL, we previously developed micelles formed from predominantly hydrophobic amphiphilic diblock copolymers of poly(styrene-alt-maleic anhydride)-b-poly(styrene) (e.g., PSMA100-b-PS258) that exhibit robust PTL loading (75%efficiency, 11% w/w capacity) and release PTL over 24 h. Here, PTL-loaded PSMA-b-PS micelles were thoroughly characterized in vitro for PTL delivery to MV4-11 AML cells. Additionally, the mechanisms governing micelle-mediated cytotoxicity were examined in comparison to free PTL. PSMA-b-PS micelles were taken up by MV4-11 cells as evidenced by transmission electron microscopy and flow cytometry. Specifically, MV4-11 cells relied on clathrin-mediated endocytosis, rather than caveolae-mediated endocytosis and macropinocytosis. In addition, PTL-loaded PSMA-b-PS micelles exhibited a dose-dependent cytotoxicity towards AML cells and were capable of reducing cell viability by 75% at 10 μM PTL, while unloaded micelles were nontoxic. At 10 μM PTL, the cytotoxicity of PTL-loaded micelles increased gradually over 24 h while free PTL achieved maximal cytotoxicity between 2 and 4 h, demonstrating micelle-mediated delivery of PTL to AML cells and stability of the drug-loaded micelle even in the presence of cells. Both free PTL and PTL-loaded micelles induced NF-κB inhibition at 10 μM PTL doses, demonstrating some mechanistic similarities in cytotoxicity. However, free PTL relied more heavily on exofacial free thiol interactions to induce cytotoxicity than PTL-loaded micelles; free PTL cytotoxicity was reduced by over twofold when cell surface free thiols were depleted, where PTL-loaded micelle doses were unaffected by cell surface thiol modulation. The physical properties, stability, and efficacy of PTL-loaded PSMA-b-PS micelles support further development of a leukemia therapeutic with greater bioavailability and the potential to eliminate LSCs.
Collapse
|
70
|
Pickens CA, Sordillo LM, Comstock SS, Harris WS, Hortos K, Kovan B, Fenton JI. Plasma phospholipids, non-esterified plasma polyunsaturated fatty acids and oxylipids are associated with BMI. Prostaglandins Leukot Essent Fatty Acids 2015; 95:31-40. [PMID: 25559239 PMCID: PMC4361296 DOI: 10.1016/j.plefa.2014.12.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 02/06/2023]
Abstract
The obese lipid profile is associated with increased free fatty acids and triacylglycerides. Currently, little is known about the plasma lipid species associated with obesity. In this study, we compared plasma lipid fatty acid (FA) profiles as a function of BMI. Profiling phospholipid (PL) FAs and their respective oxylipids could predict which obese individuals are more likely to suffer from diseases associated with chronic inflammation or oxidative stress. We investigated the relationship between BMI and plasma PL (PPL) FA composition in 126 men using a quantitative gas chromatography analysis. BMI was inversely associated with both PPL nervonic and linoleic acid (LA) but was positively associated with both dihomo-γ-linolenic and palmitoleic acid. Compared to lean individuals, obese participants were more likely to have ω-6 FAs, except arachidonic acid and LA, incorporated into PPLs. Obese participants were less likely to have EPA and DHA incorporated into PPLs compared to lean participants. Non-esterified plasma PUFA and oxylipid analysis showed ω-6 oxylipids were more abundant in the obese plasma pool. These ω-6 oxylipids are associated with increased angiogenesis (i.e. epoxyeicosatrienoates), reactive oxygen species (i.e. 9-hydroxyeicosatetraenoate), and inflammation resolution (i.e. Lipoxin A4). In summary, BMI is directly associated with specific PPL FA and increased ω-6 oxylipids.
Collapse
Affiliation(s)
- C Austin Pickens
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Lorraine M Sordillo
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - Sarah S Comstock
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - William S Harris
- Sanford School of Medicine, The University of South Dakota, Sioux Falls, SD, USA
| | - Kari Hortos
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Bruce Kovan
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA; Tri-County Gastroenterology Professional Corporation, Clinton Tri-County Gastroenterology Professional Corporation, MI, USA
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
71
|
Comprehensive lipid profiling of plasma in patients with benign breast tumor and breast cancer reveals novel biomarkers. Anal Bioanal Chem 2015; 407:5065-77. [DOI: 10.1007/s00216-015-8484-x] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/07/2015] [Accepted: 01/12/2015] [Indexed: 01/31/2023]
|
72
|
Zhang Y, Geng L, Talmon G, Wang J. MicroRNA-520g confers drug resistance by regulating p21 expression in colorectal cancer. J Biol Chem 2015; 290:6215-25. [PMID: 25616665 DOI: 10.1074/jbc.m114.620252] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Development of drug resistance is one of the major causes of colorectal cancer recurrence, yet mechanistic understanding and therapeutic options remain limited. Here, we show that expression of microRNA (miR)-520g is correlated with drug resistance of colon cancer cells. Ectopic expression of miR-520g conferred resistance to 5-fluorouracil (5-FU)- or oxaliplatin-induced apoptosis in vitro and reduced the effectiveness of 5-FU in the inhibition of tumor growth in a mouse xenograft model in vivo. Further studies indicated that miR-520g mediated drug resistance through down-regulation of p21 expression. Moreover, p53 suppressed miR-520g expression, and deletion of p53 up-regulated miR-520g expression. Inhibition of miR-520g in p53(-/-) cells increased their sensitivity to 5-FU treatment. Importantly, studies of patient samples indicated that expression of miR-520g correlated with chemoresistance in colorectal cancer. These findings indicate that the p53/miR-520g/p21 signaling axis plays an important role in the response of colorectal cancer to chemotherapy. A major implication of our studies is that inhibition of miR-520g or restoration of p21 expression may have considerable therapeutic potential to overcome drug resistance in colorectal cancer patients, especially in those with mutant p53.
Collapse
Affiliation(s)
- Yang Zhang
- From the Eppley Institute for Research in Cancer and Allied Diseases, Department of Genetics, Cell Biology, and Anatomy
| | - Liying Geng
- From the Eppley Institute for Research in Cancer and Allied Diseases
| | - Geoffrey Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198
| | - Jing Wang
- From the Eppley Institute for Research in Cancer and Allied Diseases, Department of Genetics, Cell Biology, and Anatomy, Department of Biochemistry and Molecular Biology and Fred & Pamela Buffett Cancer Center, and
| |
Collapse
|
73
|
Zhu T, Xu L, Xu X, Wang Z, Zhu J, Xie Q, Zhang B, Wang Y, Ju L, He Y, Ye X, Zhou D, Li Y. Analysis of breast cancer-associated glycosphingolipids using electrospray ionization-linear ion trap quadrupole mass spectrometry. Carbohydr Res 2015; 402:189-99. [DOI: 10.1016/j.carres.2014.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 09/27/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
|
74
|
Eid SY, El-Readi MZ, Fatani SH, Mohamed Nour Eldin EE, Wink M. Natural Products Modulate the Multifactorial Multidrug Resistance of Cancer. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/pp.2015.63017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
75
|
Abdi J, Chen G, Chang H. Drug resistance in multiple myeloma: latest findings and new concepts on molecular mechanisms. Oncotarget 2014; 4:2186-207. [PMID: 24327604 PMCID: PMC3926819 DOI: 10.18632/oncotarget.1497] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In the era of new and mostly effective therapeutic protocols, multiple myeloma still tends to be a hard-to-treat hematologic cancer. This hallmark of the disease is in fact a sequel to drug resistant phenotypes persisting initially or emerging in the course of treatment. Furthermore, the heterogeneous nature of multiple myeloma makes treating patients with the same drug challenging because finding a drugable oncogenic process common to all patients is not yet feasible, while our current knowledge of genetic/epigenetic basis of multiple myeloma pathogenesis is outstanding. Nonetheless, bone marrow microenvironment components are well known as playing critical roles in myeloma tumor cell survival and environment-mediated drug resistance happening most possibly in all myeloma patients. Generally speaking, however; real mechanisms underlying drug resistance in multiple myeloma are not completely understood. The present review will discuss the latest findings and concepts in this regard. It reviews the association of important chromosomal translocations, oncogenes (e.g. TP53) mutations and deranged signaling pathways (e.g. NFκB) with drug response in clinical and experimental investigations. It will also highlight how bone marrow microenvironment signals (Wnt, Notch) and myeloma cancer stem cells could contribute to drug resistance in multiple myeloma.
Collapse
Affiliation(s)
- Jahangir Abdi
- Dept. of Laboratory Medicine & Pathobiology, University of Toronto, Ontario, Canada
| | | | | |
Collapse
|
76
|
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: 29] [Impact Index Per Article: 2.6] [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
|
77
|
Kozovska Z, Gabrisova V, Kucerova L. Colon cancer: Cancer stem cells markers, drug resistance and treatment. Biomed Pharmacother 2014; 68:911-6. [DOI: 10.1016/j.biopha.2014.10.019] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/15/2014] [Indexed: 12/14/2022] Open
|
78
|
Tyrosine kinase inhibitors as reversal agents for ABC transporter mediated drug resistance. Molecules 2014; 19:13848-77. [PMID: 25191874 PMCID: PMC6271846 DOI: 10.3390/molecules190913848] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 08/25/2014] [Accepted: 08/29/2014] [Indexed: 01/27/2023] Open
Abstract
Tyrosine kinases (TKs) play an important role in pathways that regulate cancer cell proliferation, apoptosis, angiogenesis and metastasis. Aberrant activity of TKs has been implicated in several types of cancers. In recent years, tyrosine kinase inhibitors (TKIs) have been developed to interfere with the activity of deregulated kinases. These TKIs are remarkably effective in the treatment of various human cancers including head and neck, gastric, prostate and breast cancer and several types of leukemia. However, these TKIs are transported out of the cell by ATP-binding cassette (ABC) transporters, resulting in development of a characteristic drug resistance phenotype in cancer patients. Interestingly, some of these TKIs also inhibit the ABC transporter mediated multi drug resistance (MDR) thereby; enhancing the efficacy of conventional chemotherapeutic drugs. This review discusses the clinically relevant TKIs and their interaction with ABC drug transporters in modulating MDR.
Collapse
|
79
|
Mahdavian E, Palyok P, Adelmund S, Williams-Hart T, Furmanski BD, Kim YJ, Gu Y, Barzegar M, Wu Y, Bhinge KN, Kolluru GK, Quick Q, Liu YY, Kevil CG, Salvatore BA, Huang S, Clifford JL. Biological activities of fusarochromanone: a potent anti-cancer agent. BMC Res Notes 2014; 7:601. [PMID: 25187308 PMCID: PMC4168212 DOI: 10.1186/1756-0500-7-601] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 08/15/2014] [Indexed: 11/10/2022] Open
Abstract
Background Fusarochromanone (FC101) is a small molecule fungal metabolite with a host of interesting biological functions, including very potent anti-angiogenic and direct anti-cancer activity. Results Herein, we report that FC101 exhibits very potent in-vitro growth inhibitory effects (IC50 ranging from 10nM-2.5 μM) against HaCat (pre-malignant skin), P9-WT (malignant skin), MCF-7 (low malignant breast), MDA-231 (malignant breast), SV-HUC (premalignant bladder), UM-UC14 (malignant bladder), and PC3 (malignant prostate) in a time-course and dose-dependent manner, with the UM-UC14 cells being the most sensitive. FC101 induces apoptosis and an increase in proportion of cells in the sub-G1 phase in both HaCat and P9-WT cell lines as evidenced by cell cycle profile analysis. In a mouse xenograft SCC tumor model, FC101 was well tolerated, non-toxic, and achieved a 30% reduction in tumor size at a dose of 8 mg/kg/day. FC101 is also a potent anti-angiogenenic agent. At nanomolar doses, FC101 inhibits the vascular endothelial growth factor-A (VEGF-A)-mediated proliferation of endothelial cells. Conclusions Our data presented here indicates that FC101 is an excellent lead candidate for a small molecule anti-cancer agent that simultaneously affects angiogenesis signaling, cancer signal transduction, and apoptosis. Further understanding of the underlying FC101’s molecular mechanism may lead to the design of novel targeted and selective therapeutics, both of which are pursued targets in cancer drug discovery.
Collapse
Affiliation(s)
- Elahe Mahdavian
- Department of Chemistry and Physics, LSU-Shreveport, One University Place, Shreveport, LA 71115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Foo J, Michor F. Evolution of acquired resistance to anti-cancer therapy. J Theor Biol 2014; 355:10-20. [PMID: 24681298 PMCID: PMC4058397 DOI: 10.1016/j.jtbi.2014.02.025] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/19/2014] [Accepted: 02/20/2014] [Indexed: 12/21/2022]
Abstract
Acquired drug resistance is a major limitation for the successful treatment of cancer. Resistance can emerge due to a variety of reasons including host environmental factors as well as genetic or epigenetic alterations in the cancer cells. Evolutionary theory has contributed to the understanding of the dynamics of resistance mutations in a cancer cell population, the risk of resistance pre-existing before the initiation of therapy, the composition of drug cocktails necessary to prevent the emergence of resistance, and optimum drug administration schedules for patient populations at risk of evolving acquired resistance. Here we review recent advances towards elucidating the evolutionary dynamics of acquired drug resistance and outline how evolutionary thinking can contribute to outstanding questions in the field.
Collapse
Affiliation(s)
- Jasmine Foo
- School of Mathematics, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biostatistics, Harvard School of Public Health, Boston, MA 02215, USA.
| |
Collapse
|
81
|
Gutiérrez-Iglesias G, Hurtado Y, Palma-Lara I, López-Marure R. Resistance to the antiproliferative effect induced by a short-chain ceramide is associated with an increase of glucosylceramide synthase, P-glycoprotein, and multidrug-resistance gene-1 in cervical cancer cells. Cancer Chemother Pharmacol 2014; 74:809-17. [DOI: 10.1007/s00280-014-2552-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/25/2014] [Indexed: 11/30/2022]
|
82
|
Panchuk R, Skorokhyd N, Chumak V, Lehka L, Omelyanchik S, Gurinovich V, Moiseenok A, Heffeter P, Berger W, Stoika R. Specific antioxidant compounds differentially modulate cytotoxic activity of doxorubicin and cisplatin: in vitro and in vivo study. Croat Med J 2014; 55:206-17. [PMID: 24891279 PMCID: PMC4049213 DOI: 10.3325/cmj.2014.55.206] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim To use the antioxidant compounds (sodium selenite, selenomethionine, D-pantethine) for modulation of cytotoxic effect of doxorubicin and cisplatin toward wild type and drug-resistant mutants of several human tumor cells. Similar treatments were applied in vivo toward adult male Wistar rats. Methods Human tumor cells of different lines (HCT-116, Jurkat and HL-60) with various mechanisms of drug-resistance were treated with doxorubicin or cisplatin, alone or in combination with sodium selenite, selenomethionine, or D-pantethine. Cell viability, induction of apoptosis, and production of O2- radicals were measured. Activity of redox potential modulating enzymes was measured in the liver and blood plasma of adult male Wistar rats subjected to similar treatments. Results All antioxidants used in physiologically harmless concentration inhibited cytotoxic action of doxorubicin toward tumor cells sensitive to chemotherapy treatment by 15%-30%, and slightly enhanced cytotoxic effect of this medicine toward drug-resistant malignant cells. At the same time, there was no significant effect of these antioxidants on cisplatin action. Such effects were accompanied by a complete inhibition of production of superoxide radicals induced by doxorubicin. The results of in vivo study in adult male Wistar rats were in agreement with the results of in vitro study of human tumor cells. Conclusion Protective effect of specific antioxidant agents during cytotoxic action of doxorubicin was demonstrated in vitro in drug-sensitive human tumor cells and in adult male Wistar rats, while there was no protective effect in drug-resistant sub-lines of these tumor cells during action of doxorubicin and cisplatin.
Collapse
Affiliation(s)
- Rostyslav Panchuk
- Rostyslav Panchuk, Institute of Cell Biology, National Academy of Sciences of Ukraine, Drahomanov Street 14/16, 79005, Lviv, Ukraine,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Chiu WH, Chen HHW, Chang JY, Luo SJ, Li CL, Chen CL, Su WC, Lin CF. Inhibiting glucosylceramide synthase facilitates the radiosensitizing effects of vinorelbine in lung adenocarcinoma cells. Cancer Lett 2014; 349:144-51. [DOI: 10.1016/j.canlet.2014.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/11/2014] [Accepted: 04/07/2014] [Indexed: 10/25/2022]
|
84
|
Greer AH, Yong T, Fennell K, Moustafa YW, Fowler M, Galiano F, Ng SW, Berkowitz RS, Cardelli J, Meyers S, Davis JN. Knockdown of core binding factorβ alters sphingolipid metabolism. J Cell Physiol 2014; 228:2350-64. [PMID: 23813439 DOI: 10.1002/jcp.24406] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 05/10/2013] [Indexed: 12/13/2022]
Abstract
Core binding factor (CBF) is a heterodimeric transcription factor containing one of three DNA-binding proteins of the Runt-related transcription factor family (RUNX1-3) and the non-DNA-binding protein, CBFβ. RUNX1 and CBFβ are the most common targets of chromosomal rearrangements in leukemia. CBF has been implicated in other cancer types; for example RUNX1 and RUNX2 are implicated in cancers of epithelial origin, including prostate, breast, and ovarian cancers. In these tumors, CBF is involved in maintaining the malignant phenotype and, when highly over-expressed, contributes to metastatic growth in bone. Herein, lentiviral delivery of CBFβ-specific shRNAs was used to achieve a 95% reduction of CBFβ in an ovarian cancer cell line. This drastic reduction in CBFβ expression resulted in growth inhibition that was not associated with a cell cycle block or an increase in apoptosis. However, CBFβ silencing resulted in increased autophagy and production of reactive oxygen species (ROS). Since sphingolipid and ceramide metabolism regulates non-apoptotic cell death, autophagy, and ROS production, fumonsin B1 (FB1), an inhibitor of ceramide synthase, was used to alter ceramide production in the CBFβ-silenced cells. FB1 treatment inhibited the CBFβ-dependent increase in autophagy and provided a modest increase in cell survival. To document alterations to sphingolipids in the CBFβ-silenced cells, ceramide, and lactosylceramide levels were directly examined by mass spectrometry. Substantial increases in ceramide species and decreases in lactosylceramides were identified. Altogether, this report provides evidence that CBF transcriptional pathways control cellular survival, at least in part, through sphingolipid metabolism.
Collapse
Affiliation(s)
- Adam H Greer
- Department of Biochemistry and Molecular Biology and Feist-Weiller Cancer Center, LSUHSC School of Medicine in Shreveport, Shreveport, Louisiana
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Kathawala RJ, Chen JJ, Zhang YK, Wang YJ, Patel A, Wang DS, Talele TT, Ashby CR, Chen ZS. Masitinib antagonizes ATP-binding cassette subfamily G member 2-mediated multidrug resistance. Int J Oncol 2014; 44:1634-42. [PMID: 24626598 PMCID: PMC4027943 DOI: 10.3892/ijo.2014.2341] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/06/2014] [Indexed: 12/15/2022] Open
Abstract
In this in vitro study, we determined whether masitinib could reverse multidrug resistance (MDR) in cells overexpressing the ATP binding cassette subfamily G member 2 (ABCG2) transporter. Masitinib (1.25 and 2.5 μM) significantly decreases the resistance to mitoxantrone (MX), SN38 and doxorubicin in HEK293 and H460 cells overexpressing the ABCG2 transporter. In addition, masitinib (2.5 μM) significantly increased the intracellular accumulation of [3H]-MX, a substrate for ABCG2, by inhibiting the function of ABCG2 and significantly decreased the efflux of [3H]-MX. However, masitinib (2.5 μM) did not significantly alter the expression of the ABCG2 protein. In addition, a docking model suggested that masitinib binds within the transmembrane region of a homology-modeled human ABCG2 transporter. Overall, our in vitro findings suggest that masitinib reverses MDR to various anti-neoplastic drugs in HEK293 and H460 cells overexpressing ABCG2 by inhibiting their transport activity as opposed to altering their levels of expression.
Collapse
Affiliation(s)
- Rishil J Kathawala
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Jun-Jiang Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Yun-Kai Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Yi-Jun Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Atish Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - De-Shen Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Tanaji T Talele
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| |
Collapse
|
86
|
Giussani P, Tringali C, Riboni L, Viani P, Venerando B. Sphingolipids: key regulators of apoptosis and pivotal players in cancer drug resistance. Int J Mol Sci 2014; 15:4356-92. [PMID: 24625663 PMCID: PMC3975402 DOI: 10.3390/ijms15034356] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/07/2014] [Accepted: 02/21/2014] [Indexed: 12/17/2022] Open
Abstract
Drug resistance elicited by cancer cells still constitutes a huge problem that frequently impairs the efficacy of both conventional and novel molecular therapies. Chemotherapy usually acts to induce apoptosis in cancer cells; therefore, the investigation of apoptosis control and of the mechanisms used by cancer cells to evade apoptosis could be translated in an improvement of therapies. Among many tools acquired by cancer cells to this end, the de-regulated synthesis and metabolism of sphingolipids have been well documented. Sphingolipids are known to play many structural and signalling roles in cells, as they are involved in the control of growth, survival, adhesion, and motility. In particular, in order to increase survival, cancer cells: (a) counteract the accumulation of ceramide that is endowed with pro-apoptotic potential and is induced by many drugs; (b) increase the synthesis of sphingosine-1-phosphate and glucosylceramide that are pro-survivals signals; (c) modify the synthesis and the metabolism of complex glycosphingolipids, particularly increasing the levels of modified species of gangliosides such as 9-O acetylated GD3 (αNeu5Ac(2-8)αNeu5Ac(2-3)βGal(1-4)βGlc(1-1)Cer) or N-glycolyl GM3 (αNeu5Ac (2-3)βGal(1-4)βGlc(1-1)Cer) and de-N-acetyl GM3 (NeuNH(2)βGal(1-4)βGlc(1-1)Cer) endowed with anti-apoptotic roles and of globoside Gb3 related to a higher expression of the multidrug resistance gene MDR1. In light of this evidence, the employment of chemical or genetic approaches specifically targeting sphingolipid dysregulations appears a promising tool for the improvement of current chemotherapy efficacy.
Collapse
Affiliation(s)
- Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan 20090), Italy.
| | - Cristina Tringali
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan 20090), Italy.
| | - Laura Riboni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan 20090), Italy.
| | - Paola Viani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan 20090), Italy.
| | - Bruno Venerando
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Segrate (Milan 20090), Italy.
| |
Collapse
|
87
|
Dahdouh F, Raane M, Thévenod F, Lee WK. Nickel-induced cell death and survival pathways in cultured renal proximal tubule cells: roles of reactive oxygen species, ceramide and ABCB1. Arch Toxicol 2014; 88:881-92. [DOI: 10.1007/s00204-014-1194-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/09/2014] [Indexed: 12/17/2022]
|
88
|
Owczarek TB, Suchanski J, Pula B, Kmiecik AM, Chadalski M, Jethon A, Dziegiel P, Ugorski M. Galactosylceramide affects tumorigenic and metastatic properties of breast cancer cells as an anti-apoptotic molecule. PLoS One 2013; 8:e84191. [PMID: 24391908 PMCID: PMC3877204 DOI: 10.1371/journal.pone.0084191] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/13/2013] [Indexed: 11/17/2022] Open
Abstract
It was recently proposed that UDP-galactose:ceramide galactosyltransferase (UGT8), enzyme responsible for synthesis of galactosylceramide (GalCer), is a significant index of tumor aggressiveness and a potential marker for the prognostic evaluation of lung metastases in breast cancer. To further reveal the role of UGT8 and GalCer in breast cancer progression, tumorigenicity and metastatic potential of control MDA-MB-231 cells (MDA/LUC) and MDA-MB-231 cells (MDA/LUC-shUGT8) with highly decreased expression of UGT8 and GalCer after stable expression of shRNA directed against UGT8 mRNA was studied in vivo in athymic nu/nu mice. Control MDA/LUC cells formed tumors and metastatic colonies much more efficiently in comparison to MDA/LUC-shUGT8 cells with suppressed synthesis of GalCer after their, respectively, orthotopic and intracardiac transplantation. These findings indicate that UGT8 and GalCer have a profound effect on tumorigenic and metastatic properties of breast cancer cells. In accordance with this finding, immunohistochemical staining of tumor specimens revealed that high expression of UGT8 accompanied by accumulation of GalCer in MDA-MB-231 cells is associated with a much higher proliferative index and a lower number of apoptotic cells in comparison to the MDA/LUC-shUGT8 cells. In addition, it was found that expression of UGT8 in MDA-MB-231 cells increased their resistance to apoptosis induced by doxorubicin in vitro. Therefore, these data suggest that accumulation of GalCer in tumor cells inhibits apoptosis, which would facilitates metastatic cells to survive in the hostile microenvironment of tumor in target organ.
Collapse
Affiliation(s)
- Tomasz B Owczarek
- Laboratory of Glycobiology and Cell Interactions, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland ; Department of Biochemistry, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Environmental and Life Sciences, Wroclaw, Poland
| | - Jarosław Suchanski
- Department of Biochemistry, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Environmental and Life Sciences, Wroclaw, Poland
| | - Bartosz Pula
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Alicja M Kmiecik
- Laboratory of Glycobiology and Cell Interactions, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marek Chadalski
- Laboratory of Glycobiology and Cell Interactions, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Aleksandra Jethon
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Piotr Dziegiel
- Department of Histology and Embryology, Wroclaw Medical University, Wroclaw, Poland ; Department of Physiotherapy, Wroclaw University School of Physical Education, Wroclaw, Poland
| | - Maciej Ugorski
- Laboratory of Glycobiology and Cell Interactions, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland ; Department of Biochemistry, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Environmental and Life Sciences, Wroclaw, Poland
| |
Collapse
|
89
|
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: 6.2] [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
|
90
|
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: 365] [Impact Index Per Article: 30.4] [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
|
91
|
Gupta V, Liu YY. New Insights on Glucosylceramide Synthase in Cancer Drug Resistance and Myelosuppression. BIOCHEMISTRY & PHARMACOLOGY : OPEN ACCESS 2013; 2. [PMID: 25401049 PMCID: PMC4229685 DOI: 10.4172/2167-0501.1000120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Vineet Gupta
- Department of Basic Pharmaceutical Sciences, University of Louisiana at Monroe, LA 71209, USA
| | - Yong-Yu Liu
- Department of Basic Pharmaceutical Sciences, University of Louisiana at Monroe, LA 71209, USA
| |
Collapse
|
92
|
García-Barros M, Coant N, Truman JP, Snider AJ, Hannun YA. Sphingolipids in colon cancer. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:773-82. [PMID: 24060581 DOI: 10.1016/j.bbalip.2013.09.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/12/2013] [Accepted: 09/16/2013] [Indexed: 01/28/2023]
Abstract
Colorectal cancer is one of the major causes of death in the western world. Despite increasing knowledge of the molecular signaling pathways implicated in colon cancer, therapeutic outcomes are still only moderately successful. Sphingolipids, a family of N-acyl linked lipids, have not only structural functions but are also implicated in important biological functions. Ceramide, sphingosine and sphingosine-1-phosphate are the most important bioactive lipids, and they regulate several key cellular functions. Accumulating evidence suggests that many cancers present alterations in sphingolipids and their metabolizing enzymes. The aim of this review is to discuss the emerging roles of sphingolipids, both endogenous and dietary, in colon cancer and the interaction of sphingolipids with WNT/β-catenin pathway, one of the most important signaling cascades that regulate development and homeostasis in intestine. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Mónica García-Barros
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 101 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA.
| | - Nicolas Coant
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 101 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA.
| | - Jean-Philip Truman
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 101 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA.
| | - Ashley J Snider
- VAMC Northport, 79 Middleville Road, Northport, NY, USA, Health Science Center, Stony Brook University, Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 101 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA.
| |
Collapse
|
93
|
Metabolism, physiological role, and clinical implications of sphingolipids in gastrointestinal tract. BIOMED RESEARCH INTERNATIONAL 2013; 2013:908907. [PMID: 24083248 PMCID: PMC3780527 DOI: 10.1155/2013/908907] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/30/2013] [Accepted: 08/02/2013] [Indexed: 01/21/2023]
Abstract
Sphingolipids in digestive system are responsible for numerous important physiological and pathological processes. In the membrane of gut epithelial cells, sphingolipids provide structural integrity, regulate absorption of some nutrients, and act as receptors for many microbial antigens and their toxins. Moreover, bioactive sphingolipids such as ceramide or sphingosine-1-phosphate regulate cellular growth, differentiation, and programmed cell death-apoptosis. Although it is well established that sphingolipids have clinical implications in gastrointestinal tumorigenesis or inflammation, further studies are needed to fully explore the role of sphingolipids in neoplastic and inflammatory diseases in gastrointestinal tract. Pharmacological agents which regulate metabolism of sphingolipids can be potentially used in the management of colorectal cancer or inflammatory bowel diseases. The aim of this work is to critically the review physiological and pathological roles of sphingolipids in the gastrointestinal tract.
Collapse
|
94
|
Alfonso P, Navascués J, Navarro S, Medina P, Bolado-Carrancio A, Andreu V, Irún P, Rodríguez-Rey JC, Pocoví M, España F, Giraldo P. Characterization of Variants in the Glucosylceramide Synthase Gene and their Association with Type 1 Gaucher Disease Severity. Hum Mutat 2013; 34:1396-403. [DOI: 10.1002/humu.22381] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 07/08/2013] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Silvia Navarro
- Centro de Investigación; Hospital Universitario La Fe; Valencia; Spain
| | - Pilar Medina
- Centro de Investigación; Hospital Universitario La Fe; Valencia; Spain
| | - Alfonso Bolado-Carrancio
- Departamento de Biología Molecular; Universidad de Cantabria e Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV); Santander; Spain
| | | | | | - José Carlos Rodríguez-Rey
- Departamento de Biología Molecular; Universidad de Cantabria e Instituto de Formación e Investigación Marqués de Valdecilla (IFIMAV); Santander; Spain
| | | | - Francisco España
- Centro de Investigación; Hospital Universitario La Fe; Valencia; Spain
| | | |
Collapse
|
95
|
The impact of sphingosine kinase-1 in head and neck cancer. Biomolecules 2013; 3:481-513. [PMID: 24970177 PMCID: PMC4030949 DOI: 10.3390/biom3030481] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/02/2013] [Accepted: 08/03/2013] [Indexed: 12/15/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has a high reoccurrence rate and an extremely low survival rate. There is limited availability of effective therapies to reduce the rate of recurrence, resulting in high morbidity and mortality of advanced cases. Late presentation, delay in detection of lesions, and a high rate of metastasis make HNSCC a devastating disease. This review offers insight into the role of sphingosine kinase-1 (SphK1), a key enzyme in sphingolipid metabolism, in HNSCC. Sphingolipids not only play a structural role in cellular membranes, but also modulate cell signal transduction pathways to influence biological outcomes such as senescence, differentiation, apoptosis, migration, proliferation, and angiogenesis. SphK1 is a critical regulator of the delicate balance between proliferation and apoptosis. The highest expression of SphK1 is found in the advanced stage of disease, and there is a positive correlation between SphK1 expression and recurrent tumors. On the other hand, silencing SphK1 reduces HNSCC tumor growth and sensitizes tumors to radiation-induced death. Thus, SphK1 plays an important and influential role in determining HNSCC proliferation and metastasis. We discuss roles of SphK1 and other sphingolipids in HNSCC development and therapeutic strategies against HNSCC.
Collapse
|
96
|
Limtrakul P, Khantamat O, Pintha K. Inhibition of P-Glycoprotein Function and Expression by Kaempferol and Quercetin. J Chemother 2013; 17:86-95. [PMID: 15828450 DOI: 10.1179/joc.2005.17.1.86] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The 170 kDa plasma membrane P-glycoprotein (Pgp) causes the efflux of chemotherapeutic drugs from cells and is believed to be an important mechanism in multidrug resistance (MDR) in human cancer. This study demonstrates that some putative flavonoids, i.e., flavonols (quercetin and kaempferol) and isoflavones (genistein and daidzein) markedly increase the sensitivity of the multidrug-resistant human cervical carcinoma KB-V1 cells (high Pgp expression) to vinblastine and paclitaxel dose-dependently, and also decrease the relative resistance of these anti-cancer-drugs in KB-V1 cells. None of the flavonoids had a significant effect on vinblastine and paclitaxel cytotoxicity in wildtype drug-sensitive KB-3-1 cells (lacking Pgp). These flavonoids also caused an increase in intracellular accumulation, and reduced the efflux of Rh123 and 3[H]vinblastine in KB-V1 cells, but not in KB-3-1 cells. The flavonols increased the inhibitory effectiveness of Pgp activity in MDR KB-V1 cells more than isoflavones. Only treatment with flavonols up to 48 h was able to significantly decrease the Pgp expression in a dose-dependent manner in KB-V1 cells. These findings provide evidence that flavonols reduced Pgp expression and function resulting in the inhibition of Pgp activity, but isoflavones modulated intracellular drug levels by inhibiting Pgp function with no effect on Pgp expression. Among the flavonoids tested, flavonols, particularly kaempferol, exhibit the most potent MDR reversing property in KB-V1 cells.
Collapse
Affiliation(s)
- P Limtrakul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | | | | |
Collapse
|
97
|
Dubey A, Min JW, Koo HJ, Kim H, Cook TR, Kang SC, Stang PJ, Chi KW. Anticancer potency and multidrug-resistant studies of self-assembled arene-ruthenium metallarectangles. Chemistry 2013; 19:11622-8. [PMID: 23852626 DOI: 10.1002/chem.201300870] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/22/2013] [Indexed: 12/31/2022]
Abstract
A suite of three tetraruthenium metallacycles have been obtained from [2+2] self-assemblies between N,N'-Di-(4-pyridyl)-1,4,5,8-naphthalenetetracarbo-xydiimide (4) and one of the three dinuclear arene ruthenium clips, (η(6)-p-iPrC6H4Me)2Ru2(OO∩OO)][OTf]2 (OO∩OO = oxalate 1, 2,5-dioxydo-1,4-benzoquinonato (dobq) 2, 5,8-dihydroxy-1,4-naphthaquinonato (donq) 3; OTf = triflate). All complexes were isolated in good yield (>85 %) as triflate salts and were fully characterized by using (1)H NMR and UV/Vis spectroscopies, and high-resolution electrospray mass spectrometry. A single crystal of the metallarectangle 5 was suitable for X-ray diffraction structural characterization. The biological activities of the metallacycles were determined by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assays, establishing their in vitro anticancer properties. Our results show that for the AGC (gastric cancer) cell lines, the cytotoxicity of (donq)-containing SCC 7 exceeds that of cisplatin, which was used as a control. For HCT15 (colon cancer) cell lines, the cytotoxicity is comparable to both cisplatin and doxorubicin. An in vivo hollow fiber model was used to show growth-inhibitory activity against HCT15 and image-based cytometry experiments indicated that 7 induced apoptosis as the mode of cell death. Complex 7 also showed significant antitumor activity for multidrug-resistant HCT15/CLO2 cell lines, for which doxorubicin was ineffective.
Collapse
Affiliation(s)
- Abhishek Dubey
- Department of Chemistry, University of Ulsan, Ulsan 680-749, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Abstract
Non-surgical therapies for human malignancies must negotiate complex cell signaling pathways to impede cancer cell growth, ideally promoting death of cancer cells while sparing healthy tissue. For most of the past half century, medical approaches for treating cancer have relied primarily on cytotoxic chemotherapeutics that interfere with DNA replication and cell division, susceptibilities of rapidly dividing cancer cells. As a consequence, these therapies exert considerable cell stress, promoting the generation of ceramide through de novo synthesis and recycling of complex glycosphingolipids and sphingomyelin into apoptotic ceramide. Radiotherapy of cancer exerts similar geno- and cytotoxic cell stresses, and generation of ceramide following ionizing radiation therapy is a well-described feature of radiation-induced cell death. Emerging evidence now describes sphingolipids as mediators of death in response to newer targeted therapies, cementing ceramide generation as a common mechanism of cell death in response to cancer therapy. Many studies have now shown that dysregulation of ceramide accumulation-whether by reduced generation or accelerated metabolism-is a common mechanism of resistance to standard cancer therapies. The aims of this chapter will be to discuss described mechanisms of cancer resistance to therapy related to dysregulation of sphingolipid metabolism and to explore clinical and preclinical approaches to interdict sphingolipid metabolism to improve outcomes of standard cancer therapies.
Collapse
|
99
|
Liu YY, Hill RA, Li YT. Ceramide glycosylation catalyzed by glucosylceramide synthase and cancer drug resistance. Adv Cancer Res 2013; 117:59-89. [PMID: 23290777 DOI: 10.1016/b978-0-12-394274-6.00003-0] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glucosylceramide synthase (GCS), converting ceramide to glucosylceramide, catalyzes the first reaction of ceramide glycosylation in sphingolipid metabolism. This glycosylation by GCS is a critical step regulating the modulation of cellular activities by controlling ceramide and glycosphingolipids (GSLs). An increase of ceramide in response to stresses, such as chemotherapy, drives cells to proliferation arrest and apoptosis or autophagy; however, ceramide glycosylation promptly eliminates ceramide and consequently, these induced processes, thus protecting cancer cells. Further, persistently enhanced ceramide glycosylation can increase GSLs, participating in selecting cancer cells to drug resistance. GCS is overexpressed in diverse drug-resistant cancer cells and in tumors of breast, colon, and leukemia that display poor response to chemotherapy. As ceramide glycosylation by GCS is a rate-limiting step in GSL synthesis, inhibition of GCS sensitizes cancer cells to anticancer drugs and eradicates cancer stem cells. Mechanistic studies indicate that uncoupling ceramide glycosylation can modulate gene expression, decreasing MDR1 through the cSrc/β-catenin pathway and restoring p53 expression via RNA splicing. These studies not only expand our knowledge in understanding how ceramide glycosylation affects cancer cells but also provide novel therapeutic approaches for targeting refractory tumors.
Collapse
Affiliation(s)
- Yong-Yu Liu
- Department of Basic Pharmaceutical Sciences, University of Louisiana at Monroe, Monroe, LA, USA.
| | | | | |
Collapse
|
100
|
Gramatzki D, Herrmann C, Happold C, Becker KA, Gulbins E, Weller M, Tabatabai G. Glioma cell death induced by irradiation or alkylating agent chemotherapy is independent of the intrinsic ceramide pathway. PLoS One 2013; 8:e63527. [PMID: 23667632 PMCID: PMC3646759 DOI: 10.1371/journal.pone.0063527] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 04/07/2013] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND/AIMS Resistance to genotoxic therapy is a characteristic feature of glioma cells. Acid sphingomyelinase (ASM) hydrolyzes sphingomyelin to ceramide and glucosylceramide synthase (GCS) catalyzes ceramide metabolism. Increased ceramide levels have been suggested to enhance chemotherapy-induced death of cancer cells. METHODS Microarray and clinical data for ASM and GCS in astrocytomas WHO grade II-IV were acquired from the Rembrandt database. Moreover, the glioblastoma database of the Cancer Genome Atlas network (TCGA) was used for survival data of glioblastoma patients. For in vitro studies, increases in ceramide levels were achieved either by ASM overexpression or by the GCS inhibitor DL-threo-1-phenyl-2-palmitoylamino-3-morpholino-1-propanol (PPMP) in human glioma cell lines. Combinations of alkylating chemotherapy or irradiation and ASM overexpression, PPMP or exogenous ceramide were applied in parental cells. The anti-glioma effects were investigated by assessing proliferation, metabolic activity, viability and clonogenicity. Finally, viability and clonogenicity were assessed in temozolomide (TMZ)-resistant cells upon treatment with PPMP, exogenous ceramide, alkylating chemotherapy, irradiation or their combinations. RESULTS Interrogations from the Rembrandt and TCGA database showed a better survival of glioblastoma patients with low expression of ASM or GCS. ASM overexpression or PPMP treatment alone led to ceramide accumulation but did not enhance the anti-glioma activity of alkylating chemotherapy or irradiation. PPMP or exogenous ceramide induced acute cytotoxicity in glioblastoma cells. Combined treatments with chemotherapy or irradiation led to additive, but not synergistic effects. Finally, no synergy was found when TMZ-resistant cells were treated with exogenous ceramide or PPMP alone or in combination with TMZ or irradiation. CONCLUSION Modulation of intrinsic glioma cell ceramide levels by ASM overexpression or GCS inhibition does not enhance the anti-glioma activity of alkylating chemotherapy or irradiation.
Collapse
Affiliation(s)
- Dorothee Gramatzki
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Caroline Herrmann
- Department of Preclinical Imaging and Radiopharmacy, University Hospital Tuebingen, Tuebingen, Germany
| | - Caroline Happold
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ghazaleh Tabatabai
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|