1
|
Luo ZH, Zhou B, Yu JY, Li H, Li Z, Ma SQ. Role of SLC31A1 in prognosis and immune infiltration in breast cancer: a novel insight. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:329-345. [PMID: 39544714 PMCID: PMC11558315 DOI: 10.62347/loyi1808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/04/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Copper, an essential metal element for humans, plays vital functions in cancer prognosis and immunity. SLC31A1, a high-affinity copper transporter, helps regulate copper homeostasis and has been implicated in tumor prognosis through mechanisms such as drug resistance, autophagy, ferroptosis, and cuproptosis. However, the role of SLC31A1 in breast cancer (BRCA) and its association with tumor immune infiltration has not been fully elucidated. This study aimed to investigate the expression pattern of SLC31A1, its clinical significance, and its effect on tumor immune infiltration in BRCA. METHODS We comprehensively analyzed multiple datasets, including Gene Expression Profiling Interaction Analysis (GEPIA), Tumor Immune Estimation Resource (TIMER), UALCAN, and Kaplan-Meier (KM) plotter, to assess the expression of SLC31A1 and its prognostic value in BRCA. Additionally, TIMER and TISIDB were used to explore the correlation between SLC31A1 expression and the extent of tumor immune infiltration. RESULTS SLC31A1 was significantly upregulated in BRCA tissues compared to adjacent non-tumor tissues. Higher SLC31A1 expression levels were associated with poorer clinical outcome. Multivariate Cox regression analysis confirmed that SLC31A1 served as an independent prognostic indicator. Furthermore, SLC31A1 expression showed significant associations with various immunomodulators, chemokines, chemokine receptors, and tumor-infiltrating lymphocytes (TILs), including CD8+ T cells, CD4+ T cells, regulatory T cells (Tregs), follicular helper T cells (Tfh), neutrophils, M2 macrophages, tumor-associated macrophages (TAMs), and monocytes. These findings suggest that SLC31A1 may regulate macrophage polarization and T cell exhaustion in BRCA, contributing to immune evasion and poor prognosis. CONCLUSION Our study underscores the importance of further research to explore the therapeutic potential of targeting SLC31A1 and to uncover its additional roles in BRCA beyond the known mechanisms of drug resistance, autophagy, ferroptosis, and cuproptosis.
Collapse
Affiliation(s)
- Zhen-Hua Luo
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Bo Zhou
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Jun-Yi Yu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - He Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
- Hunan Provincial Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, Changsha Medical UniversityChangsha 410219, Hunan, The People’s Republic of China
| | - Zan Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South UniversityChangsha 410008, Hunan, The People’s Republic of China
| | - Si-Qing Ma
- Department of Pharmacy, Hunan Chest HospitalChangsha 4100013, Hunan, The People’s Republic of China
| |
Collapse
|
2
|
Phadte P, Bishnu A, Dey P, M M, Mehrotra M, Singh P, Chakrabarty S, Majumdar R, Rekhi B, Patra M, De A, Ray P. Autophagy-mediated ID1 turnover dictates chemo-resistant fate in ovarian cancer stem cells. J Exp Clin Cancer Res 2024; 43:222. [PMID: 39123206 PMCID: PMC11316295 DOI: 10.1186/s13046-024-03147-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The mechanisms enabling dynamic shifts between drug-resistant and drug-sensitive states in cancer cells are still underexplored. This study investigated the role of targeted autophagic protein degradation in regulating ovarian cancer stem cell (CSC) fate decisions and chemo-resistance. METHODS Autophagy levels were compared between CSC-enriched side population (SP) and non-SP cells (NSP) in multiple ovarian cancer cell lines using immunoblotting, immunofluorescence, and transmission electron microscopy. The impact of autophagy modulation on CSC markers and differentiation was assessed by flow cytometry, immunoblotting and qRT-PCR. In silico modeling and co-immunoprecipitation identified ID1 interacting proteins. Pharmacological and genetic approaches along with Annexin-PI assay, ChIP assay, western blotting, qRT-PCR and ICP-MS were used to evaluate effects on cisplatin sensitivity, apoptosis, SLC31A1 expression, promoter binding, and intracellular platinum accumulation in ID1 depleted backdrop. Patient-derived tumor spheroids were analyzed for autophagy and SLC31A1 levels. RESULTS Ovarian CSCs exhibited increased basal autophagy compared to non-CSCs. Further autophagy stimulation by serum-starvation and chemical modes triggered proteolysis of the stemness regulator ID1, driving the differentiation of chemo-resistant CSCs into chemo-sensitive non-CSCs. In silico modeling predicted TCF12 as a potent ID1 interactor, which was validated by co-immunoprecipitation. ID1 depletion freed TCF12 to transactivate the cisplatin influx transporter SLC31A1, increasing intracellular cisplatin levels and cytotoxicity. Patient-derived tumor spheroids exhibited a functional association between autophagy, ID1, SLC31A1, and platinum sensitivity. CONCLUSIONS This study reveals a novel autophagy-ID1-TCF12-SLC31A1 axis where targeted autophagic degradation of ID1 enables rapid remodeling of CSCs to reverse chemo-resistance. Modulating this pathway could counter drug resistance in ovarian cancer.
Collapse
Affiliation(s)
- Pratham Phadte
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Aniketh Bishnu
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Pranay Dey
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Manikandan M
- Laboratory of Medicinal Chemistry and Cell Biology, Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Megha Mehrotra
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Prerna Singh
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Shritama Chakrabarty
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Indian Institute of Science Education and Research, Bhopal, 462066, India
| | - Rounak Majumdar
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Indian Institute of Science Education and Research, Kolkata, 741246, India
| | - Bharat Rekhi
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Department of Pathology, Tata Memorial Hospital, Mumbai, 400012, India
| | - Malay Patra
- Laboratory of Medicinal Chemistry and Cell Biology, Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Pritha Ray
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India.
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India.
| |
Collapse
|
3
|
Jiang Y, Glandorff C, Sun M. GSH and Ferroptosis: Side-by-Side Partners in the Fight against Tumors. Antioxidants (Basel) 2024; 13:697. [PMID: 38929136 PMCID: PMC11201279 DOI: 10.3390/antiox13060697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/26/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Glutathione (GSH), a prominent antioxidant in organisms, exhibits diverse biological functions and is crucial in safeguarding cells against oxidative harm and upholding a stable redox milieu. The metabolism of GSH is implicated in numerous diseases, particularly in the progression of malignant tumors. Consequently, therapeutic strategies targeting the regulation of GSH synthesis and metabolism to modulate GSH levels represent a promising avenue for future research. This study aimed to elucidate the intricate relationship between GSH metabolism and ferroptosis, highlighting how modulation of GSH metabolism can impact cellular susceptibility to ferroptosis and consequently influence the development of tumors and other diseases. The paper provides a comprehensive overview of the physiological functions of GSH, including its structural characteristics, physicochemical properties, sources, and metabolic pathways, as well as investigate the molecular mechanisms underlying GSH regulation of ferroptosis and potential therapeutic interventions. Unraveling the biological role of GSH holds promise for individuals afflicted with tumors.
Collapse
Affiliation(s)
- Yulang Jiang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Christian Glandorff
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- University Clinic of Hamburg at the HanseMerkur Center of TCM, 20251 Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
4
|
Zhong Y, Zeng W, Chen Y, Zhu X. The effect of lipid metabolism on cuproptosis-inducing cancer therapy. Biomed Pharmacother 2024; 172:116247. [PMID: 38330710 DOI: 10.1016/j.biopha.2024.116247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Cuproptosis provides a new therapeutic strategy for cancer treatment and is thought to have broad clinical application prospects. Nevertheless, some oncological clinical trials have yet to demonstrate favorable outcomes, highlighting the need for further research into the molecular mechanisms underlying cuproptosis in tumors. Cuproptosis primarily hinges on the intracellular accumulation of copper, with lipid metabolism exerting a profound influence on its course. The interaction between copper metabolism and lipid metabolism is closely related to cuproptosis. Copper imbalance can affect mitochondrial respiration and lipid metabolism changes, while lipid accumulation can promote copper uptake and absorption, and inhibit cuproptosis induced by copper. Anomalies in lipid metabolism can disrupt copper homeostasis within cells, potentially triggering cuproptosis. The interaction between cuproptosis and lipid metabolism regulates the occurrence, development, metastasis, chemotherapy drug resistance, and tumor immunity of cancer. Cuproptosis is a promising new target for cancer treatment. However, the influence of lipid metabolism and other factors should be taken into consideration. This review provides a brief overview of the characteristics of the interaction between cuproptosis and lipid metabolism in cancer and analyses potential strategies of applying cuproptosis for cancer treatment.
Collapse
Affiliation(s)
- Yue Zhong
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Wei Zeng
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yongbo Chen
- Rehabilitation College of Gannan Medical University, Ganzhou 341000, China
| | - Xiuzhi Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
5
|
Schötz S, Griepe AK, Goerisch BB, Kortam S, Vainer YS, Dimde M, Koeppe H, Wedepohl S, Quaas E, Achazi K, Schroeder A, Haag R. Esterase-Responsive Polyglycerol-Based Nanogels for Intracellular Drug Delivery in Rare Gastrointestinal Stromal Tumors. Pharmaceuticals (Basel) 2023; 16:1618. [PMID: 38004483 PMCID: PMC10675119 DOI: 10.3390/ph16111618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Rare gastrointestinal stromal tumors (GISTs) are caused by mutations in the KIT and PDGFRA genes. Avapritinib (BLU-285) is a targeted selective inhibitor for mutated KIT and PDGFRA receptors that can be used to treat these tumors. However, there are subtypes of GISTs that exhibit resistance against BLU-285 and thus require other treatment strategies. This can be addressed by employing a drug delivery system that transports a combination of drugs with distinct cell targets. In this work, we present the synthesis of esterase-responsive polyglycerol-based nanogels (NGs) to overcome drug resistance in rare GISTs. Using inverse nanoprecipitation mediated with inverse electron-demand Diels-Alder cyclizations (iEDDA) between dPG-methyl tetrazine and dPG-norbornene, multi-drug-loaded NGs were formed based on a surfactant-free encapsulation protocol. The obtained NGs displayed great stability in the presence of fetal bovine serum (FBS) and did not trigger hemolysis in red blood cells over a period of 24 h. Exposing the NGs to Candida Antarctica Lipase B (CALB) led to the degradation of the NG network, indicating the capability of targeted drug release. The bioactivity of the loaded NGs was tested in vitro on various cell lines of the GIST-T1 family, which exhibit different drug resistances. Cell internalization with comparable uptake kinetics of the NGs could be confirmed by confocal laser scanning microscopy (CLSM) and flow cytometry for all cell lines. Cell viability and live cell imaging studies revealed that the loaded NGs are capable of intracellular drug release by showing similar IC50 values to those of the free drugs. Furthermore, multi-drug-loaded NGs were capable of overcoming BLU-285 resistance in T1-α-D842V + G680R cells, demonstrating the utility of this carrier system.
Collapse
Affiliation(s)
- Sebastian Schötz
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr, 3, 14195 Berlin, Germany; (S.S.); (A.K.G.); (B.B.G.); (H.K.)
| | - Adele K. Griepe
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr, 3, 14195 Berlin, Germany; (S.S.); (A.K.G.); (B.B.G.); (H.K.)
| | - Björn B. Goerisch
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr, 3, 14195 Berlin, Germany; (S.S.); (A.K.G.); (B.B.G.); (H.K.)
| | - Sally Kortam
- School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Yael Shammai Vainer
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Technion, Haifa 32000, Israel;
| | - Mathias Dimde
- Research Center of Electron Microscopy, Freie Universität Berlin, Fabeckstr, 36A, 14195 Berlin, Germany;
| | - Hanna Koeppe
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr, 3, 14195 Berlin, Germany; (S.S.); (A.K.G.); (B.B.G.); (H.K.)
| | - Stefanie Wedepohl
- Research Building SupraFAB, Freie Universität Berlin, Altensteinstr, 23a, 14195 Berlin, Germany (E.Q.); (K.A.)
| | - Elisa Quaas
- Research Building SupraFAB, Freie Universität Berlin, Altensteinstr, 23a, 14195 Berlin, Germany (E.Q.); (K.A.)
| | - Katharina Achazi
- Research Building SupraFAB, Freie Universität Berlin, Altensteinstr, 23a, 14195 Berlin, Germany (E.Q.); (K.A.)
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Technion, Haifa 32000, Israel;
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr, 3, 14195 Berlin, Germany; (S.S.); (A.K.G.); (B.B.G.); (H.K.)
- Research Building SupraFAB, Freie Universität Berlin, Altensteinstr, 23a, 14195 Berlin, Germany (E.Q.); (K.A.)
| |
Collapse
|
6
|
Yu W, Chen Y, Putluri N, Osman A, Coarfa C, Putluri V, Kamal AHM, Asmussen JK, Katsonis P, Myers JN, Lai SY, Lu W, Stephan CC, Powell RT, Johnson FM, Skinner HD, Kazi J, Ahmed KM, Hu L, Threet A, Meyer MD, Bankson JA, Wang T, Davis J, Parker KR, Harris MA, Baek ML, Echeverria GV, Qi X, Wang J, Frederick AI, Walsh AJ, Lichtarge O, Frederick MJ, Sandulache VC. Evolution of cisplatin resistance through coordinated metabolic reprogramming of the cellular reductive state. Br J Cancer 2023; 128:2013-2024. [PMID: 37012319 PMCID: PMC10205814 DOI: 10.1038/s41416-023-02253-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Cisplatin (CDDP) is a mainstay treatment for advanced head and neck squamous cell carcinomas (HNSCC) despite a high frequency of innate and acquired resistance. We hypothesised that tumours acquire CDDP resistance through an enhanced reductive state dependent on metabolic rewiring. METHODS To validate this model and understand how an adaptive metabolic programme might be imprinted, we performed an integrated analysis of CDDP-resistant HNSCC clones from multiple genomic backgrounds by whole-exome sequencing, RNA-seq, mass spectrometry, steady state and flux metabolomics. RESULTS Inactivating KEAP1 mutations or reductions in KEAP1 RNA correlated with Nrf2 activation in CDDP-resistant cells, which functionally contributed to resistance. Proteomics identified elevation of downstream Nrf2 targets and the enrichment of enzymes involved in generation of biomass and reducing equivalents, metabolism of glucose, glutathione, NAD(P), and oxoacids. This was accompanied by biochemical and metabolic evidence of an enhanced reductive state dependent on coordinated glucose and glutamine catabolism, associated with reduced energy production and proliferation, despite normal mitochondrial structure and function. CONCLUSIONS Our analysis identified coordinated metabolic changes associated with CDDP resistance that may provide new therapeutic avenues through targeting of these convergent pathways.
Collapse
Affiliation(s)
- Wangie Yu
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Yunyun Chen
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Abdullah Osman
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Vasanta Putluri
- Advanced Technology core, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Abu H M Kamal
- Advanced Technology core, Dan Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer Kay Asmussen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen Y Lai
- Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wuhao Lu
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Clifford C Stephan
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX, USA
| | - Reid T Powell
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
- Department of Translational Medical Sciences, School of Medicine, Texas A&M University, Houston, TX, USA
| | - Faye M Johnson
- Department of Thoracic Head and Neck Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heath D Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Jawad Kazi
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Kazi Mokim Ahmed
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Linghao Hu
- Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Addison Threet
- Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, USA
| | - James A Bankson
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tony Wang
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Jack Davis
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Kirby R Parker
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Madison A Harris
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Mokryun L Baek
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Gloria V Echeverria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoli Qi
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jin Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
| | - Andy I Frederick
- School of Electrical and Computer Engineering Undergraduate Department, Cornell University, NY, USA
| | - Alex J Walsh
- Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry & Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Program in Quantitative and Computational Biosciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
- Computational and Integrative Biomedical Research Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Mitchell J Frederick
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
| | - Vlad C Sandulache
- Bobby R. Alford Department of Otolaryngology Head and Neck Surgery, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
7
|
Guan D, Zhao L, Shi X, Ma X, Chen Z. Copper in cancer: From pathogenesis to therapy. Biomed Pharmacother 2023; 163:114791. [PMID: 37105071 DOI: 10.1016/j.biopha.2023.114791] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023] Open
Abstract
One of the basic trace elements for the structure and metabolism of human tissue is copper. However, as a heavy metal, excessive intake or abnormal accumulation of copper in the body can cause inevitable damage to the organism because copper can result in direct injury to various cell components or disruption of the redox balance, eventually leading to cell death. Interestingly, a growing body of research reports that diverse cancers have raised serum and tumor copper levels. Tumor cells depend on more copper for their metabolism than normal cells, and a decrease in copper or copper overload can have a detrimental effect on tumor cells. New modalities for identifying and characterizing copper-dependent signals offer translational opportunities for tumor therapy, but their mechanisms remain unclear. Therefore, this article summarizes what we currently know about the correlation between copper and cancer and describes the characteristics of copper metabolism in tumor cells and the prospective application of copper-derived therapeutics.
Collapse
Affiliation(s)
- Defeng Guan
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China
| | - Lihui Zhao
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China
| | - Xin Shi
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China
| | - Xiaoling Ma
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China; Gansu key Laboratory of Reproductive Medicine and Embryology, Lanzhou, China.
| | - Zhou Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou, China; The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Jiang M, Yang T, Chu Y, Zhang Z, Sun H, Liang H, Yang F. Design of a novel Pt(II) complex to reverse cisplatin-induced resistance in lung cancer via a multi-mechanism. Dalton Trans 2022; 51:5257-5270. [PMID: 35285843 DOI: 10.1039/d1dt03964d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In order to develop a novel platinum (Pt) complex aiming to overcome cisplatin resistance, we synthesised a series of novel Pt complexes (C1-C6). These Pt complexes displayed potent cytotoxicity activity against resistant lung cancer cells (A549cisR) in vitro and efficiently inhibited tumour growth in vivo. The Pt complexes can target DNA, lead to DNA platination and cause cell cycle arrest in the S phase, thus impeding precise DNA synthesis. C6, in particular, induced not only apoptosis but also lethal autophagy in A549cisR cells. In addition, these novel Pt complexes reversed cisplatin-induced resistance via inhibiting the expression of P-glycoprotein and decreasing the level of glutathione in A549cisR cells. Moreover, the ERK pathway was involved in C6-induced overcoming cisplatin resistance.
Collapse
Affiliation(s)
- Ming Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China. .,School of food and biochemical engineering, Guangxi Science & Technology Normal University, Laibin, Guangxi 546199, China
| | - Tongfu Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Yong Chu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China.
| |
Collapse
|
9
|
Amino Acid Metabolism in Cancer Drug Resistance. Cells 2022; 11:cells11010140. [PMID: 35011702 PMCID: PMC8750102 DOI: 10.3390/cells11010140] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Despite the numerous investigations on resistance mechanisms, drug resistance in cancer therapies still limits favorable outcomes in cancer patients. The complexities of the inherent characteristics of tumors, such as tumor heterogeneity and the complicated interaction within the tumor microenvironment, still hinder efforts to overcome drug resistance in cancer cells, requiring innovative approaches. In this review, we describe recent studies offering evidence for the essential roles of amino acid metabolism in driving drug resistance in cancer cells. Amino acids support cancer cells in counteracting therapies by maintaining redox homeostasis, sustaining biosynthetic processes, regulating epigenetic modification, and providing metabolic intermediates for energy generation. In addition, amino acid metabolism impacts anticancer immune responses, creating an immunosuppressive or immunoeffective microenvironment. A comprehensive understanding of amino acid metabolism as it relates to therapeutic resistance mechanisms will improve anticancer therapeutic strategies.
Collapse
|
10
|
Wen MH, Xie X, Huang PS, Yang K, Chen TY. Crossroads between membrane trafficking machinery and copper homeostasis in the nerve system. Open Biol 2021; 11:210128. [PMID: 34847776 PMCID: PMC8633785 DOI: 10.1098/rsob.210128] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Imbalanced copper homeostasis and perturbation of membrane trafficking are two common symptoms that have been associated with the pathogenesis of neurodegenerative and neurodevelopmental diseases. Accumulating evidence from biophysical, cellular and in vivo studies suggest that membrane trafficking orchestrates both copper homeostasis and neural functions-however, a systematic review of how copper homeostasis and membrane trafficking interplays in neurons remains lacking. Here, we summarize current knowledge of the general trafficking itineraries for copper transporters and highlight several critical membrane trafficking regulators in maintaining copper homeostasis. We discuss how membrane trafficking regulators may alter copper transporter distribution in different membrane compartments to regulate intracellular copper homeostasis. Using Parkinson's disease and MEDNIK as examples, we further elaborate how misregulated trafficking regulators may interplay parallelly or synergistically with copper dyshomeostasis in devastating pathogenesis in neurodegenerative diseases. Finally, we explore multiple unsolved questions and highlight the existing challenges to understand how copper homeostasis is modulated through membrane trafficking.
Collapse
Affiliation(s)
- Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Xihong Xie
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Karen Yang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
11
|
Zhang QZ, Wen F, Yang HL, Cao YY, Peng RG, Wang YM, Nie L, Qin YK, Wu JJ, Zhao X, Zi D. GADD45α alleviates the CDDP resistance of SK-OV3/cddp cells via redox-mediated DNA damage. Oncol Lett 2021; 22:720. [PMID: 34429760 PMCID: PMC8371983 DOI: 10.3892/ol.2021.12981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 03/08/2021] [Indexed: 11/25/2022] Open
Abstract
Epithelial ovarian cancer has the highest mortality rate of all malignant ovarian cancer types. Great progress has been made in the treatment of ovarian cancer in recent years. However, drug resistance has led to a low level of 5-year survival rate of epithelial ovarian cancer, and the molecular mechanism of which remains unknown. The aim of the present study was to identify the role of redox status in the cisplatin (CDDP) resistance of ovarian cancer. CDDP-resistant SK-OV3 (SK-OV3/cddp) cells were prepared and their reactive oxygen species and glutathione levels were investigated. The effects of hydrogen peroxide on the CDDP sensitivity of the SK-OV3/cddp cells and their expression levels of the redox-associated protein growth arrest and DNA damage 45a (GADD45α) were also investigated. In addition, the impact of GADD45α overexpression on cell viability was evaluated in vitro and in vivo, and the levels of Ser-139 phosphorylated H2A histone family member X (γ-H2AX), which is associated with DNA damage, were detected. The results suggested that redox status affected the drug resistance of the ovarian cancer cells by increasing the expression of GADD45α. The overexpression of GADD45α reversed the CDDP resistance of the SK-OV3/cddp cells and increased the level of γ-H2AX. In conclusion, GADD45α alleviated the CDDP resistance of SK-OV3/cddp cells via the induction of redox-mediated DNA damage.
Collapse
Affiliation(s)
- Qi-Zhu Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Fang Wen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China.,Department of Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550004, P.R. China
| | - Han-Lin Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yan-Yan Cao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ren Guo Peng
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yuan-Mei Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Lei Nie
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yuan-Kun Qin
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Jin-Jian Wu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Xing Zhao
- National and Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guiyang, Guizhou 550004, P.R. China
| | - Dan Zi
- Department of Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550004, P.R. China.,National and Guizhou Joint Engineering Laboratory for Cell Engineering and Biomedicine Technique, Center for Tissue Engineering and Stem Cell Research, Guizhou Province Key Laboratory of Regenerative Medicine, Guiyang, Guizhou 550004, P.R. China.,Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
12
|
Maung MT, Carlson A, Olea-Flores M, Elkhadragy L, Schachtschneider KM, Navarro-Tito N, Padilla-Benavides T. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J 2021; 35:e21810. [PMID: 34390520 DOI: 10.1096/fj.202100273rr] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/23/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022]
Abstract
Copper (Cu) is an essential micronutrient required for the activity of redox-active enzymes involved in critical metabolic reactions, signaling pathways, and biological functions. Transporters and chaperones control Cu ion levels and bioavailability to ensure proper subcellular and systemic Cu distribution. Intensive research has focused on understanding how mammalian cells maintain Cu homeostasis, and how molecular signals coordinate Cu acquisition and storage within organs. In humans, mutations of genes that regulate Cu homeostasis or facilitate interactions with Cu ions lead to numerous pathologic conditions. Malfunctions of the Cu+ -transporting ATPases ATP7A and ATP7B cause Menkes disease and Wilson disease, respectively. Additionally, defects in the mitochondrial and cellular distributions and homeostasis of Cu lead to severe neurodegenerative conditions, mitochondrial myopathies, and metabolic diseases. Cu has a dual nature in carcinogenesis as a promotor of tumor growth and an inducer of redox stress in cancer cells. Cu also plays role in cancer treatment as a component of drugs and a regulator of drug sensitivity and uptake. In this review, we provide an overview of the current knowledge of Cu metabolism and transport and its relation to various human pathologies.
Collapse
Affiliation(s)
- May T Maung
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Alyssa Carlson
- Department of Molecular Biology and Biochemistry, Wesleyan University, Middletown, CT, USA
| | - Monserrat Olea-Flores
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | - Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Napoleon Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Guerrero, Mexico
| | | |
Collapse
|
13
|
Kuo MT, Huang YF, Chou CY, Chen HHW. Targeting the Copper Transport System to Improve Treatment Efficacies of Platinum-Containing Drugs in Cancer Chemotherapy. Pharmaceuticals (Basel) 2021; 14:ph14060549. [PMID: 34201235 PMCID: PMC8227247 DOI: 10.3390/ph14060549] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022] Open
Abstract
The platinum (Pt)-containing antitumor drugs including cisplatin (cis-diamminedichloroplatinum II, cDDP), carboplatin, and oxaliplatin, have been the mainstay of cancer chemotherapy. These drugs are effective in treating many human malignancies. The major cell-killing target of Pt drugs is DNA. Recent findings underscored the important roles of Pt drug transport system in cancer therapy. While many mechanisms have been proposed for Pt-drug transport, the high-affinity copper transporter (hCtr1), Cu chaperone (Atox1), and Cu exporters (ATP7A and ATP7B) are also involved in cDDP transport, highlighting Cu homeostasis regulation in Pt-based cancer therapy. It was demonstrated that by reducing cellular Cu bioavailable levels by Cu chelators, hCtr1 is transcriptionally upregulated by transcription factor Sp1, which binds the promoters of Sp1 and hCtr1. In contrast, elevated Cu poisons Sp1, resulting in suppression of hCtr1 and Sp1, constituting the Cu-Sp1-hCtr1 mutually regulatory loop. Clinical investigations using copper chelator (trientine) in carboplatin treatment have been conducted for overcoming Pt drug resistance due in part to defective transport. While results are encouraging, future development may include targeting multiple steps in Cu transport system for improving the efficacies of Pt-based cancer chemotherapy. The focus of this review is to delineate the mechanistic interrelationships between Cu homeostasis regulation and antitumor efficacy of Pt drugs.
Collapse
Affiliation(s)
- Macus Tien Kuo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yu-Fang Huang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Cheng-Yang Chou
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
- Correspondence: (C.-Y.C.); (H.H.W.C.)
| | - Helen H. W. Chen
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (C.-Y.C.); (H.H.W.C.)
| |
Collapse
|
14
|
Tchounwou PB, Dasari S, Noubissi FK, Ray P, Kumar S. Advances in Our Understanding of the Molecular Mechanisms of Action of Cisplatin in Cancer Therapy. J Exp Pharmacol 2021; 13:303-328. [PMID: 33776489 PMCID: PMC7987268 DOI: 10.2147/jep.s267383] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 02/23/2021] [Indexed: 12/15/2022] Open
Abstract
Cisplatin and other platinum-based chemotherapeutic drugs have been used extensively for the treatment of human cancers such as bladder, blood, breast, cervical, esophageal, head and neck, lung, ovarian, testicular cancers, and sarcoma. Cisplatin is commonly administered intravenously as a first-line chemotherapy for patients suffering from various malignancies. Upon absorption into the cancer cell, cisplatin interacts with cellular macromolecules and exerts its cytotoxic effects through a series of biochemical mechanisms by binding to Deoxyribonucleic acid (DNA) and forming intra-strand DNA adducts leading to the inhibition of DNA synthesis and cell growth. Its primary molecular mechanism of action has been associated with the induction of both intrinsic and extrinsic pathways of apoptosis resulting from the production of reactive oxygen species through lipid peroxidation, activation of various signal transduction pathways, induction of p53 signaling and cell cycle arrest, upregulation of pro-apoptotic genes/proteins, and down-regulation of proto-oncogenes and anti-apoptotic genes/proteins. Despite great clinical outcomes, many studies have reported substantial side effects associated with cisplatin monotherapy, while others have shown substantial drug resistance in some cancer patients. Hence, new formulations and several combinational therapies with other drugs have been tested for the purpose of improving the clinical utility of cisplatin. Therefore, this review provides a comprehensive understanding of its molecular mechanisms of action in cancer therapy and discusses the therapeutic approaches to overcome cisplatin resistance and side effects.
Collapse
Affiliation(s)
- Paul B Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH-RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS, USA
| | - Shaloam Dasari
- Cellomics and Toxicogenomics Research Laboratory, NIH-RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS, USA
| | - Felicite K Noubissi
- Cellomics and Toxicogenomics Research Laboratory, NIH-RCMI Center for Health Disparities Research, Jackson State University, Jackson, MS, USA
| | - Paresh Ray
- Department of Chemistry and Biochemistry, College of Science, Engineering and Technology, Jackson State University, Jackson, MS, USA
| | - Sanjay Kumar
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| |
Collapse
|
15
|
Tsai TL, Lai YH, HW Chen H, Su WC. Overcoming Radiation Resistance by Iron-Platinum Metal Alloy Nanoparticles in Human Copper Transport 1-Overexpressing Cancer Cells via Mitochondrial Disturbance. Int J Nanomedicine 2021; 16:2071-2085. [PMID: 33727814 PMCID: PMC7955785 DOI: 10.2147/ijn.s283147] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Radiation therapy remains an important treatment modality in cancer therapy, however, resistance is a major problem for treatment failure. Elevated expression of glutathione is known to associate with radiation resistance. We used glutathione overexpressing small cell lung cancer cell lines, SR3A-13 and SR3A-14, established by transfection with γ-glutamylcysteine synthetase (γ-GCS) cDNA, as a model for investigating strategies of overcoming radiation resistance. These radiation-resistant cells exhibit upregulated human copper transporter 1 (hCtr1), which also transports cisplatin. This study was initiated to investigate the effect and the underlying mechanism of iron-platinum nanoparticles (FePt NPs) on radiation sensitization in cancer cells. MATERIALS AND METHODS Uptakes of FePt NPs in these cells were studied by plasma optical emission spectrometry and transmission electron microscopy. Effects of the combination of FePt NPs and ionizing radiation were investigated by colony formation assay and animal experiment. Intracellular reactive oxygen species (ROS) were assessed by using fluorescent probes and imaged by a fluorescence-activated-cell-sorting caliber flow cytometer. Oxygen consumption rate (OCR) in mitochondria after FePt NP and IR treatment was investigated by a Seahorse XF24 cell energy metabolism analyzer. RESULTS These hCtr1-overexpressing cells exhibited elevated resistance to IR and the resistance could be overcome by FePt NPs via enhanced uptake of FePt NPs. Overexpression of hCtr1 was responsible for the increased uptake/transport of FePt NPs as demonstrated by using hCtr1-transfected parental SR3A (SR3A-hCtr1-WT) cells. Increased ROS and drastic mitochondrial damages with substantial reduction of oxygen consumption rate were observed in FePt NPs and IR-treated cells, indicating that structural and functional insults of mitochondria are the lethal mechanism of FePt NPs. Furthermore, FePt NPs also increased the efficacy of radiotherapy in mice bearing SR3A-hCtr1-WT-xenograft tumors. CONCLUSION These results suggest that FePt NPs can potentially be a novel strategy to improve radiotherapeutic efficacy in hCtr1-overexpressing cancer cells via enhanced uptake and mitochondria targeting.
Collapse
Affiliation(s)
- Tsung-Lin Tsai
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yu-Hsuan Lai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Helen HW Chen
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wu-Chou Su
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
16
|
Abstract
Therapeutic nanomaterials serve as an important platform for drug delivery under image guidance. Despite significant growth and broad applications, their design specifics remain a subject of continued interest primarily due to multifunctional factors involved, ranging from nanomaterial properties, imaging modalities, and therapeutic agents to activation strategies. This review article summarizes key findings on their design characteristics with a particular interest in strategies developed for therapeutic activation (release). First, their activation can be controlled using either an endogenous factor including low pH and glutathione or an external stimulation by light, ultrasound, or electromagnetic field. The former is passively controlled from a spatiotemporal aspect compared to the latter, which is otherwise actively controlled through drug linker photolysis, nanomaterial disassembly, or gate opening. Second, light stimulation serves a most notable strategy due to its essential role in controlled drug release, photothermal activation (hyperthermia), and photodynamic production of reactive oxygen species (ROS). Third, some of those activation strategies that rely on ultrasound, photothermal, photoacoustic, magnetic field, or X-ray radiation are dually functional due to their role in imaging modalities. In summary, this review article presents recent advances and new insights that pertain to nanotherapeutic delivery systems. It also addresses their technical limitations associated with tissue penetration (light), spatial resolution (ultrasound, hyperthermia), and occurrence of cellular resistance (ROS).
Collapse
|
17
|
Bartnicka JJ, Al-Salemee F, Firth G, Blower PJ. L-Cysteine-mediated modulation of copper trafficking in prostate cancer cells: an in vitro and in vivo investigation with 64Cu and 64Cu-PET. Metallomics 2020; 12:1508-1520. [PMID: 32959856 DOI: 10.1039/d0mt00161a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Copper imbalance is implicated in many diseases, including cancer. Copper in blood is mainly transported by carrier proteins but a small fraction is bound to low molecular weight species, possibly amino acids. Their roles in cellular copper delivery are unknown. Our aim was to test whether accumulation of 64Cu into cancer-derived cells can be influenced by copper-binding serum amino acids. In vitro cellular accumulation of 64Cu was measured in Hank's Balanced Salt Solution in the presence of 100 μM l-histidine, l-methionine, l-cysteine and l-threonine. l-Cysteine markedly increased 64Cu accumulation and retention in DU145, PC3 and SK-OV-3 cells, while some other cell lines did not show an effect. This effect was not due to 64Cu delivery in the form of a 64Cu-cysteine complex, nor to reduction of 64Cu(ii) to 64Cu(i) by l-cysteine. Pre-incubation of cells with l-cysteine increased 64Cu accumulation, even if l-cysteine was removed from HBSS before 64Cu was added. The effect of l-cysteine on 64Cu accumulation was not mediated by increased glutathione synthesis. Despite the demonstrable in vitro effect, pre-injection of l-cysteine precursor N-acetyl-cysteine (NAC) in vivo did not enhance 64Cu delivery to DU145 xenografts in mice. Instead, it decreased 64Cu accumulation in the DU145 tumour and in brain, as assessed by PET imaging. We conclude that 64Cu is not delivered to DU145 cancer cells in vitro as a complex with amino acids but its cellular accumulation is enhanced by l-cysteine or NAC influx to cells. The latter effect was not demonstrable in vivo in the DU145 xenograft.
Collapse
Affiliation(s)
- Joanna J Bartnicka
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| | - Fahad Al-Salemee
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| | - George Firth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
18
|
Choi SK. Photoactivation Strategies for Therapeutic Release in Nanodelivery Systems. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences University of Michigan Medical School Ann Arbor MI 48109 USA
- Department of Internal Medicine University of Michigan Medical School Ann Arbor MI 48109 USA
| |
Collapse
|
19
|
Chen A, Jiang P, Zeb F, Wu X, Xu C, Chen L, Feng Q. EGCG regulates CTR1 expression through its pro-oxidative property in non-small-cell lung cancer cells. J Cell Physiol 2020; 235:7970-7981. [PMID: 31943177 DOI: 10.1002/jcp.29451] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/03/2020] [Indexed: 12/21/2022]
Abstract
Copper transporter 1 (CTR1) plays an important role in increasing cisplatin intake. Our previous studies showed that CTR1 expression was upregulated by (-)-epigallocatechin-3-gallate (EGCG), a green tea polyphenol, therefore enhanced cisplatin sensitivity in ovary cancer and non-small-cell lung cancer (NSCLC) cells. In the current study in the non-small-cell lung cancer cells, we uncovered a potential mechanism of EGCG-induced CTR1 through its pro-oxidative property. We found that EGCG increased reactive oxygen species (ROS) generation, while in the presence of ROS scavenger N-acetyl-cysteine (NAC), ROS production was eliminated. Changes of CTR1 expression were consistent with the ROS level. Simultaneously, EGCG downregulated ERK1/2 while upregulated lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) through ROS to induce CTR1 expression. Besides, in a nude mouse xenografts model, EGCG treatment raised ROS level, expression of CTR1 and NEAT1 in tumor tissue. Also, ERK1/2 and p-ERK1/2 were suppressed as well. Taken together, these results suggested a novel mechanism that EGCG mediated ROS to regulate CTR1 expression through the ERK1/2/NEAT1 signaling pathway, which provided more possibilities for EGCG as a natural agent in adjuvant therapy of lung cancer.
Collapse
Affiliation(s)
- Aochang Chen
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pan Jiang
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Falak Zeb
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoyue Wu
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuyue Xu
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lijun Chen
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Feng
- Key Laboratory of Toxicology, Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
20
|
Benyettou F, Prakasam T, Ramdas Nair A, Witzel II, Alhashimi M, Skorjanc T, Olsen JC, Sadler KC, Trabolsi A. Potent and selective in vitro and in vivo antiproliferative effects of metal-organic trefoil knots. Chem Sci 2019; 10:5884-5892. [PMID: 31360392 PMCID: PMC6582759 DOI: 10.1039/c9sc01218d] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023] Open
Abstract
A set of metal-organic trefoil knots (M-TKs) generated by metal-templated self-assembly of a simple pair of chelating ligands were well tolerated in vitro by non-cancer cells but were significantly more potent than cisplatin in both human cancer cells--including those resistant to cisplatin--and in zebrafish embryos. In cultured cells, M-TKs generated reactive oxygen species that triggered apoptosis via the mitochondrial pathway without directly disrupting the cell-membrane or damaging nuclear DNA. The cytotoxicity and wide scope for structural variation of M-TKs indicate the potential of synthetic metal-organic knots as a new field of chemical space for pharmaceutical design and development.
Collapse
Affiliation(s)
- Farah Benyettou
- Program in Chemistry , New York University Abu Dhabi , UAE .
| | | | | | | | - Marwa Alhashimi
- Program in Chemistry , New York University Abu Dhabi , UAE .
| | - Tina Skorjanc
- Program in Chemistry , New York University Abu Dhabi , UAE .
| | - John-Carl Olsen
- Department of Chemistry , University of Rochester , Rochester , New York , USA
| | | | - Ali Trabolsi
- Program in Chemistry , New York University Abu Dhabi , UAE .
| |
Collapse
|
21
|
Lai YH, Kuo C, Kuo MT, Chen HHW. Modulating Chemosensitivity of Tumors to Platinum-Based Antitumor Drugs by Transcriptional Regulation of Copper Homeostasis. Int J Mol Sci 2018; 19:ijms19051486. [PMID: 29772714 PMCID: PMC5983780 DOI: 10.3390/ijms19051486] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/10/2018] [Accepted: 05/12/2018] [Indexed: 12/21/2022] Open
Abstract
Platinum (Pt)-based antitumor agents have been effective in treating many human malignancies. Drug importing, intracellular shuffling, and exporting—carried out by the high-affinity copper (Cu) transporter (hCtr1), Cu chaperone (Ato x1), and Cu exporters (ATP7A and ATP7B), respectively—cumulatively contribute to the chemosensitivity of Pt drugs including cisplatin and carboplatin, but not oxaliplatin. This entire system can also handle Pt drugs via interactions between Pt and the thiol-containing amino acid residues in these proteins; the interactions are strongly influenced by cellular redox regulators such as glutathione. hCtr1 expression is induced by acute Cu deprivation, and the induction is regulated by the transcription factor specific protein 1 (Sp1) which by itself is also regulated by Cu concentration variations. Copper displaces zinc (Zn) coordination at the zinc finger (ZF) domains of Sp1 and inactivates its DNA binding, whereas Cu deprivation enhances Sp1-DNA interactions and increases Sp1 expression, which in turn upregulates hCtr1. Because of the shared transport system, chemosensitivity of Pt drugs can be modulated by targeting Cu transporters. A Cu-lowering agent (trientine) in combination with a Pt drug (carboplatin) has been used in clinical studies for overcoming Pt-resistance. Future research should aim at further developing effective Pt drug retention strategies for improving the treatment efficacy.
Collapse
Affiliation(s)
- Yu-Hsuan Lai
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Chin Kuo
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
| | - Macus Tien Kuo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Helen H W Chen
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan.
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
22
|
Banerjee A, Banerjee K, Sinha A, Das S, Majumder S, Majumdar S, Choudhuri SK. A zinc Schiff base complex inhibits cancer progression both in vivo and in vitro by inducing apoptosis. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2017; 56:383-392. [PMID: 29145169 DOI: 10.1016/j.etap.2017.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 11/03/2017] [Accepted: 11/04/2017] [Indexed: 05/25/2023]
Abstract
Cancer chemotherapy suffers from selectivity and undesired toxicity of the drugs. Since zinc is a biocompatible tracer element and cysteine derivatives are used in cancer chemoprevention, we intend to develop a complex of zinc and cysteine-derivatives as potent, non-toxic anticancer agents. Herein, we synthesized and characterized cysteine based ligand, 2-[(2-Hydroxy-3-methoxy-benzylidene)-amino]-3-mercapto-propionic acid and its Zn-complex, which are found to be non-toxic towards normal human PBMC. Data also revealed that only Zn-complex exhibited remarkable apoptosis in drug-sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 cancer cells as assessed by MTT, Cell cycle and AnnexinV binding assay. Moreover, Zn-complex altered ROS and GSH level of the respective cell lines. Finally, treatment of Zn-complex in Swiss albino mice did not show any systemic toxicity in preliminary trials in normal mice and remarkably increased the life-span of EAC bearing mice. In conclusion, the synthesized Zn-complex may be developed for efficacious, multidrug resistance reversal, non-toxic chemotherapeutic agents in future.
Collapse
Affiliation(s)
- Arpita Banerjee
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India; Department of Chemistry, Rammohan College, 102/1, Raja Rammohan Sarani, College Street, Kolkata, West Bengal 700009, India
| | - Kaushik Banerjee
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Abhinaba Sinha
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Satyajit Das
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India
| | - Saikat Majumder
- Division of Molecular Medicine, Bose Institute, P 1/12, C. I. T. Road, Scheme - VIIM, Kolkata, 700054, West Bengal, India
| | - Subrata Majumdar
- Division of Molecular Medicine, Bose Institute, P 1/12, C. I. T. Road, Scheme - VIIM, Kolkata, 700054, West Bengal, India
| | - Soumitra Kumar Choudhuri
- In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, Kolkata, 700 026, West Bengal, India.
| |
Collapse
|
23
|
Chen F, Qin X, Xu G, Gou S, Jin X. Reversal of cisplatin resistance in human gastric cancer cells by a wogonin-conjugated Pt(IV) prodrug via attenuating Casein Kinase 2-mediated Nuclear Factor-κB pathways. Biochem Pharmacol 2017; 135:50-68. [PMID: 28288821 DOI: 10.1016/j.bcp.2017.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/07/2017] [Indexed: 01/02/2023]
Abstract
Pt(IV) prodrugs, with two additional coordination sites in contrast to Pt(II) drugs, have been actively studied nowadays, for they can perform well in enhancing the accumulation and retention of the corresponding Pt(II) drugs in cancer cells. Our designed Pt(II) drug, DN604, was recently found to exhibit significant anticancer activity and low toxicity, while, wogonin, a naturally O-methylated flavones, has been widely investigated for its tumor therapeutic potential. Thus, two Pt(IV)-based prodrugs were derived by addition of a wogonin unit to the axial position of DN604 and its analogue DN603 via a linker group. In vitro cytotoxicity assay indicated that the resulting compound 8 not only inherited the genotoxicity of DN604 on gastric cancer cells, but also obtained the COX inhibitory property arising from wogonin. Further studies revealed that compound 8 caused the accumulation of ROS production and decreased the mitochondrial membrane potential (ΔΨm). The CK2α kinase activity assay, ChIP and luciferase assays showed that CK2 plays an important role in the blockade of compound 8 on activated NF-κB survival pathways, which were established for sensitivity of cancer cells to platinum drugs. Similarly in vivo, in nude mice with SGC-7901/cDDP xenografts, compound 8 improved the effectiveness of DN604 via reversing tumor resistance and maintaining low toxicity. Overall, compound 8 is a promising Pt(IV) prodrug, which could be used to promote the anticancer activity of its counterpart Pt(II) species and reverse drug resistance via attenuating CK2-mediated NF-κB pathways during platinum-based chemotherapies.
Collapse
Affiliation(s)
- Feihong Chen
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Xiaodong Qin
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Gang Xu
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China.
| | - Xiufeng Jin
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| |
Collapse
|
24
|
Li Q, Zhan M, Chen W, Zhao B, Yang K, Yang J, Yi J, Huang Q, Mohan M, Hou Z, Wang J. Phenylethyl isothiocyanate reverses cisplatin resistance in biliary tract cancer cells via glutathionylation-dependent degradation of Mcl-1. Oncotarget 2016; 7:10271-82. [PMID: 26848531 PMCID: PMC4891119 DOI: 10.18632/oncotarget.7171] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 01/21/2016] [Indexed: 02/05/2023] Open
Abstract
Biliary tract cancer (BTC) is a highly malignant cancer. BTC exhibits a low response rate to cisplatin (CDDP) treatment, and therefore, an understanding of the mechanism of CDDP resistance is urgently needed. Here, we show that BTC cells develop CDDP resistance due, in part, to upregulation of myeloid cell leukemia 1 (Mcl-1). Phenylethyl isothiocyanate (PEITC), a natural compound found in watercress, could enhance the efficacy of CDDP by degrading Mcl-1. PEITC-CDDP co-treatment also increased the rate of apoptosis of cancer stem-like side population (SP) cells and inhibited xenograft tumor growth without obvious toxic effects. In vitro, PEITC decreased reduced glutathione (GSH), which resulted in decreased GSH/oxidized glutathione (GSSG) ratio and increased glutathionylation of Mcl-1, leading to rapid proteasomal degradation of Mcl-1. Furthermore, we identified Cys16 and Cys286 as Mcl-1 glutathionylation sites, and mutating them resulted in PEITC-mediated degradation resistant Mcl-1 protein. In conclusion, we demonstrate for the first time that CDDP resistance is partially associated with Mcl-1 in BTC cells and we identify a novel mechanism that PEITC can enhance CDDP-induced apoptosis via glutathionylation-dependent degradation of Mcl-1. Hence, our results provide support that dietary intake of watercress may help reverse CDDP resistance in BTC patients.
Collapse
Affiliation(s)
- Qiwei Li
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Zhan
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Chen
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Benpeng Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Kai Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jie Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jing Yi
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qihong Huang
- The Wistar Institute, University of Pennsylvania and Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Man Mohan
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhaoyuan Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Institutes of Medical Sciences, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Zhang Q, Shi J, Yuan F, Wang H, Fu W, Pan J, Huang Y, Yu J, Yang J, Chen Z. Higher expression of XPF is a critical factor in intrinsic chemotherapy resistance of human renal cell carcinoma. Int J Cancer 2016; 139:2827-2837. [PMID: 27542841 DOI: 10.1002/ijc.30396] [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: 01/31/2016] [Revised: 07/26/2016] [Accepted: 08/08/2016] [Indexed: 02/03/2023]
Abstract
Human renal cancer is extremely resistant to chemotherapy and radiation therapy. This clinical characteristic reduces the efficacy of chemotherapeutic agents in the treatment of recurrence or metastasis following surgical resection. Understanding the mechanism of chemotherapy resistance in renal cell carcinoma remains a significant challenge. In this study, we have shown that varied level of XPF expression was organ-tissue specific by comparing human renal cancer, bladder cancer, testicular cancer and their normal tissue counterparts, respectively. The expression of XPF was significantly higher in renal cancer than in bladder cancer and testicular cancer and correlated with the clinical characteristic of their chemotherapeutics sensitivity. These novel findings proposed that the intrinsic chemoresistance of human renal cell carcinomas might be derived from the high level of XPF expression. In a panel of five cancer cell lines, decreasing cisplatin sensitivity correlated with increasing levels of XPF expression. Knockdown of XPF expression not only increased sensitivity of renal carcinoma cells to cisplatin treatment by affecting the DNA damage response, including DNA repair, cell cycle regulation and apoptosis, but also increased senescence of renal cancer cell. Furthermore, experiment in vivo confirmed that silenced XPF significantly increased the sensitivity and survival following treatment with cisplatin in xenograft mice bearing renal cell tumor. These findings firstly uncover a partial mechanism of intrinsic chemoresistance in renal cancer and may provide a new approach to break through the obstacle of intrinsic chemoresistance by targeting the XPF protein with a potential new inhibitor.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Cell Biology, The Third Military Medical University, Chongqing, China
| | - Jiazhong Shi
- Department of Cell Biology, The Third Military Medical University, Chongqing, China
| | - Fang Yuan
- Department of Urology, Chongqing Oncology Hospital, Chongqing, China
| | - Huanhuan Wang
- Department of Cell Biology, The Third Military Medical University, Chongqing, China
| | - Weihua Fu
- Urology Institute of PLA, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Jinhong Pan
- Urology Institute of PLA, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Yaqin Huang
- Department of Cell Biology, The Third Military Medical University, Chongqing, China
| | - Jin Yu
- Department of Cell Biology, The Third Military Medical University, Chongqing, China
| | - Jin Yang
- Department of Cell Biology, The Third Military Medical University, Chongqing, China.
| | - Zhiwen Chen
- Urology Institute of PLA, Southwest Hospital, The Third Military Medical University, Chongqing, China. .,Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
26
|
Disulfiram anti-cancer efficacy without copper overload is enhanced by extracellular H2O2 generation: antagonism by tetrathiomolybdate. Oncotarget 2016; 6:29771-81. [PMID: 26356671 PMCID: PMC4745761 DOI: 10.18632/oncotarget.4833] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 07/27/2015] [Indexed: 12/24/2022] Open
Abstract
Highlights Background Cu/Zn superoxide dismutases (SODs) like the extracellular SOD3 and cytoplasmic SOD1 regulate cell proliferation by generating hydrogen peroxide (H2O2). This pro-oxidant inactivates essential cysteine residues in protein tyrosine phosphatases (PTP) helping receptor tyrosine kinase activation by growth factor signaling, and further promoting downstream MEK/ERK linked cell proliferation. Disulfiram (DSF), currently in clinical cancer trials is activated by copper chelation, being potentially capable of diminishing the copper dependent activation of MEK1/2 and SOD1/SOD3 and promoting reactive oxygen species (ROS) toxicity. However, copper (Cu) overload may occur when co-administered with DSF, resulting in toxicity and mutagenicity against normal tissue, through generation of the hydroxyl radical (•OH) by the Fenton reaction. Purpose To investigate: a) whether sub-toxic DSF efficacy can be increased without Cu overload against human melanoma cells with unequal BRAF(V600E) mutant status and Her2-overexpressing SKBR3 breast cancer cells, by increasing H2O2from exogenous SOD; b) to compare the anti-tumor efficacy of DSF with that of another clinically used copper chelator, tetrathiomolybdate (TTM) Results a) without copper supplementation, exogenous SOD potentiated sub-toxic DSF toxicity antagonized by sub-toxic TTM or by the anti-oxidant N-acetylcysteine; b) exogenous glucose oxidase, another H2O2 generator resembled exogenous SOD in potentiating sub-toxic DSF. Conclusions potentiation of sub-lethal DSF toxicity by extracellular H2O2 against the human tumor cell lines investigated, only requires basal Cu and increased ROS production, being unrelated to non-specific or TTM copper chelator sequestration. Significance These findings emphasize the relevance of extracellular H2O2 as a novel mechanism to improve disulfiram anticancer effects minimizing copper toxicity.
Collapse
|
27
|
Song W, Tang Z, Shen N, Yu H, Jia Y, Zhang D, Jiang J, He C, Tian H, Chen X. Combining disulfiram and poly(l-glutamic acid)-cisplatin conjugates for combating cisplatin resistance. J Control Release 2016; 231:94-102. [PMID: 26928530 DOI: 10.1016/j.jconrel.2016.02.039] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/08/2016] [Accepted: 02/24/2016] [Indexed: 12/31/2022]
Abstract
A poly(l-glutamic acid) graft polyethylene glycol-cisplatin complex (PGA-CisPt) performs well in reducing the toxicity of free cisplatin and greatly enhances the accumulation and retention of cisplatin in solid tumors. However, there is a lack of effective treatment options for cisplatin-resistant tumors. A major reason for this is the dense PEG shell, which ensures that the PGA-CisPt maintains a long retention time in the blood that may result in it bypassing the tumor cells or failing to be endocytosed within the tumor microenvironment. Consequently, the cisplatin from PGA-CisPt is released to the extracellular space in the presence of cisplatin-resistant tumor cells and the resistant problem to free cisplatin still valid. Therefore, we devised a strategy to combat the resistance of cisplatin in the tumor microenvironment using nanoparticles-loaded disulfiram (NPs-DSF) as a modulator. In vitro, cisplatin, in combination with DSF, had a synergistic effect and decreased cell survival rate of cisplatin-resistant A549DDP cells. This effect was also observed when combining PGA-CisPt with NPs-DSF. Similarly, in Balb/C nude mice with A549DDP xenografts, NPs-DSF improved PGA-CisPt effectiveness in inhibiting tumor growth while maintaining low toxicity. Our data demonstrate that DSF reduces intracellular glutathione (GSH) levels, inhibits NFκB activity, and modulates the expression of apoptosis-related proteins Bcl-2 and Bax, thereby improves the effectiveness of cisplatin in resistant cell lines. Here, we provide a promising method for overcoming cisplatin resistance in tumors, while maintaining the in vivo benefits of the PGA-CisPt complex.
Collapse
Affiliation(s)
- Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Yanjie Jia
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Jian Jiang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; University of Chinese Academy of Sciences, Beijing 100039, PR China
| | - Chaoliang He
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| |
Collapse
|
28
|
The Essential Role of H19 Contributing to Cisplatin Resistance by Regulating Glutathione Metabolism in High-Grade Serous Ovarian Cancer. Sci Rep 2016; 6:26093. [PMID: 27193186 PMCID: PMC4872133 DOI: 10.1038/srep26093] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022] Open
Abstract
Primary and acquired drug resistance is one of the main obstacles encountered in high-grade serous ovarian cancer (HGSC) chemotherapy. Cisplatin induces DNA damage through cross-linking and long integrated non-coding RNAs (lincRNAs) play an important role in chemical induced DNA-damage response, which suggests that lincRNAs may be also associated with cisplatin resistance. However, the mechanism of long integrated non-coding RNAs (lincRNAs) acting on cisplatin resistance is not well understood. Here, we showed that expression of lin-RECK-3, H19, LUCAT1, LINC00961, and linc-CARS2-2 was enhanced in cisplatin-resistant A2780-DR cells, while transcriptome sequencing showed decreased Linc-TNFRSF19-1 and LINC00515 expression. Additionally, we verified that different H19 expression levels in HGSC tissues showed strong correlation with cancer recurrence. H19 knockdown in A2780-DR cells resulted in recovery of cisplatin sensitivity in vitro and in vivo. Quantitative proteomics analysis indicated that six NRF2-targeted proteins, including NQO1, GSR, G6PD, GCLC, GCLM and GSTP1 involved in the glutathione metabolism pathway, were reduced in H19-knockdown cells. Furthermore, H19-knockdown cells were markedly more sensitive to hydrogen-peroxide treatment and exhibited lower glutathione levels. Our results reveal a previously unknown link between H19 and glutathione metabolism in the regulation of cancer-drug resistance.
Collapse
|
29
|
Grabner S, Modec B, Bukovec N, Bukovec P, Čemažar M, Kranjc S, Serša G, Sčančar J. Cytotoxic trans-platinum(II) complex with 3-hydroxymethylpyridine: Synthesis, X-ray structure and biological activity evaluation. J Inorg Biochem 2016; 161:40-51. [PMID: 27189143 DOI: 10.1016/j.jinorgbio.2016.04.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/21/2016] [Accepted: 04/25/2016] [Indexed: 01/07/2023]
Abstract
To assess the potential cytostatic properties of Pt(II) complexes with 3-hydroxymethylpyridine (3-hmpy) as the only carrier ligand, novel cis-[PtCl2(3-hmpy)2] (1) and trans-[PtCl2(3-hmpy)2] (2) have been prepared. Elemental analysis, FTIR spectroscopy, multinuclear NMR spectroscopy and X-ray crystallography were used to determine their structures. Based on the results obtained with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay and clonogenic assay on T24 human bladder carcinoma cells (T24), the most potent compound 2 was further tested for cytotoxicity in human ovarian carcinoma cell lines - cisplatin sensitive (IGROV 1) and its resistant subclone (IGROV 1/RDDP). The cytotoxicity of compound 2 in IGROV 1/RDDP is comparable to cisplatin. Furthermore, compound 2 induced severe conformational changes in plasmid DNA, which resulted in a delayed onset of apoptosis in T24 cells, and higher amounts of Pt in tumours and serum compared to cisplatin. In addition, in vivo antitumour effectiveness was comparable to that of cisplatin with a smaller reduction of animals' body weight, thus demonstrating that it is a promising transplatin analogue which deserves further studies.
Collapse
Affiliation(s)
- Sabina Grabner
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia.
| | - Barbara Modec
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Nataša Bukovec
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Peter Bukovec
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, SI-1000 Ljubljana, Slovenia
| | - Maja Čemažar
- Institute of Oncology Ljubljana, Zaloška 2, SI-1000 Ljubljana, Slovenia
| | - Simona Kranjc
- Institute of Oncology Ljubljana, Zaloška 2, SI-1000 Ljubljana, Slovenia
| | - Gregor Serša
- Institute of Oncology Ljubljana, Zaloška 2, SI-1000 Ljubljana, Slovenia
| | - Janez Sčančar
- Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
30
|
Muggia FM, Bonetti A, Hoeschele JD, Rozencweig M, Howell SB. Platinum Antitumor Complexes: 50 Years Since Barnett Rosenberg's Discovery. J Clin Oncol 2015; 33:4219-26. [PMID: 26503202 DOI: 10.1200/jco.2015.60.7481] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Franco M Muggia
- Franco M. Muggia, New York University School of Medicine, New York, NY; Andrea Bonetti, Mater Salutis Hospitaļ Legnago, Italy; James D. Hoeschele, Eastern Michigan University, Ypsilanti, MI; Marcel Rozencweig, Innate Pharma, Marseille, France; and Stephen B. Howell, University of California San Diego (UCSD) and Moores UCSD Cancer Center, La Jolla, CA.
| | - Andrea Bonetti
- Franco M. Muggia, New York University School of Medicine, New York, NY; Andrea Bonetti, Mater Salutis Hospitaļ Legnago, Italy; James D. Hoeschele, Eastern Michigan University, Ypsilanti, MI; Marcel Rozencweig, Innate Pharma, Marseille, France; and Stephen B. Howell, University of California San Diego (UCSD) and Moores UCSD Cancer Center, La Jolla, CA
| | - James D Hoeschele
- Franco M. Muggia, New York University School of Medicine, New York, NY; Andrea Bonetti, Mater Salutis Hospitaļ Legnago, Italy; James D. Hoeschele, Eastern Michigan University, Ypsilanti, MI; Marcel Rozencweig, Innate Pharma, Marseille, France; and Stephen B. Howell, University of California San Diego (UCSD) and Moores UCSD Cancer Center, La Jolla, CA
| | - Marcel Rozencweig
- Franco M. Muggia, New York University School of Medicine, New York, NY; Andrea Bonetti, Mater Salutis Hospitaļ Legnago, Italy; James D. Hoeschele, Eastern Michigan University, Ypsilanti, MI; Marcel Rozencweig, Innate Pharma, Marseille, France; and Stephen B. Howell, University of California San Diego (UCSD) and Moores UCSD Cancer Center, La Jolla, CA
| | - Stephen B Howell
- Franco M. Muggia, New York University School of Medicine, New York, NY; Andrea Bonetti, Mater Salutis Hospitaļ Legnago, Italy; James D. Hoeschele, Eastern Michigan University, Ypsilanti, MI; Marcel Rozencweig, Innate Pharma, Marseille, France; and Stephen B. Howell, University of California San Diego (UCSD) and Moores UCSD Cancer Center, La Jolla, CA
| |
Collapse
|
31
|
Glutathione selectively modulates the binding of platinum drugs to human copper chaperone Cox17. Biochem J 2015; 472:217-23. [PMID: 26399480 DOI: 10.1042/bj20150634] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 09/22/2015] [Indexed: 12/31/2022]
Abstract
The copper chaperone Cox17 (cytochrome c oxidase copper chaperone) has been shown to facilitate the delivery of cisplatin to mitochondria, which contributes to the overall cytotoxicity of the drug [Zhao et al. (2014) Chem. Commun. 50: , 2667-2669]. Kinetic data indicate that Cox17 has reactivity similar to glutathione (GSH), the most abundant thiol-rich molecule in the cytoplasm. In the present study, we found that GSH significantly modulates the reaction of platinum complexes with Cox17. GSH enhances the reactivity of three anti-cancer drugs (cisplatin, carboplatin and oxaliplatin) to Cox17, but suppresses the reaction of transplatin. Surprisingly, the pre-formed cisplatin-GSH adducts are highly reactive to Cox17; over 90% platinum transfers from GSH to Cox17. On the other hand, transplatin-GSH adducts are inert to Cox17. These different effects are consistent with the drug activity of these platinum complexes. In addition, GSH attenuates the protein aggregation of Cox17 induced by platination. These results indicate that the platinum-protein interactions could be substantially influenced by the cellular environment.
Collapse
|
32
|
Denoyer D, Masaldan S, La Fontaine S, Cater MA. Targeting copper in cancer therapy: 'Copper That Cancer'. Metallomics 2015; 7:1459-76. [PMID: 26313539 DOI: 10.1039/c5mt00149h] [Citation(s) in RCA: 545] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Copper is an essential micronutrient involved in fundamental life processes that are conserved throughout all forms of life. The ability of copper to catalyze oxidation-reduction (redox) reactions, which can inadvertently lead to the production of reactive oxygen species (ROS), necessitates the tight homeostatic regulation of copper within the body. Many cancer types exhibit increased intratumoral copper and/or altered systemic copper distribution. The realization that copper serves as a limiting factor for multiple aspects of tumor progression, including growth, angiogenesis and metastasis, has prompted the development of copper-specific chelators as therapies to inhibit these processes. Another therapeutic approach utilizes specific ionophores that deliver copper to cells to increase intracellular copper levels. The therapeutic window between normal and cancerous cells when intracellular copper is forcibly increased, is the premise for the development of copper-ionophores endowed with anticancer properties. Also under investigation is the use of copper to replace platinum in coordination complexes currently used as mainstream chemotherapies. In comparison to platinum-based drugs, these promising copper coordination complexes may be more potent anticancer agents, with reduced toxicity toward normal cells and they may potentially circumvent the chemoresistance associated with recurrent platinum treatment. In addition, cancerous cells can adapt their copper homeostatic mechanisms to acquire resistance to conventional platinum-based drugs and certain copper coordination complexes can re-sensitize cancer cells to these drugs. This review will outline the biological importance of copper and copper homeostasis in mammalian cells, followed by a discussion of our current understanding of copper dysregulation in cancer, and the recent therapeutic advances using copper coordination complexes as anticancer agents.
Collapse
Affiliation(s)
- Delphine Denoyer
- Centre for Cellular and Molecular Biology, School of Life and Environmental Sciences, Deakin University, Burwood, Victoria, Australia.
| | | | | | | |
Collapse
|
33
|
Chen HHW, Chen WC, Liang ZD, Tsai WB, Long Y, Aiba I, Fu S, Broaddus R, Liu J, Feun LG, Savaraj N, Kuo MT. Targeting drug transport mechanisms for improving platinum-based cancer chemotherapy. Expert Opin Ther Targets 2015; 19:1307-17. [PMID: 26004625 DOI: 10.1517/14728222.2015.1043269] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Platinum (Pt)-based antitumor agents remain important chemotherapeutic agents for treating many human malignancies. Elevated expression of the human high-affinity copper transporter 1 (hCtr1), resulting in enhanced Pt drug transport into cells, has been shown to be associated with improved treatment efficacy. Thus, targeting hCtr1 upregulation is an attractive strategy for improving the treatment efficacy of Pt-based cancer chemotherapy. AREA COVERED Regulation of hCtr1 expression by cellular copper homeostasis is discussed. Association of elevated hCtr1 expression with intrinsic sensitivity of ovarian cancer to Pt drugs is presented. Mechanism of copper-lowering agents in enhancing hCtr1-mediated cis-diamminedichloroplatinum (II) (cisplatin, cDDP) transport is reviewed. Applications of copper chelation strategy in overcoming cDDP resistance through enhanced hCtr1 expression are evaluated. EXPERT OPINION While both transcriptional and post-translational mechanisms of hCtr1 regulation by cellular copper bioavailability have been proposed, detailed molecular insights into hCtr1 regulation by copper homeostasis remain needed. Recent clinical study using a copper-lowering agent in enhancing hCtr1-mediated drug transport has achieved incremental improvement in overcoming Pt drug resistance. Further improvements in identifying predictive measures in the subpopulation of patients that can benefit from the treatment are needed.
Collapse
Affiliation(s)
- Helen H W Chen
- a 1 National Cheng Kung University, National Cheng Kung University Hospital, College of Medicine, Department of Radiation Oncology , Tainan, Taiwan
| | - Wen-Chung Chen
- b 2 National Cheng Kung University, National Cheng Kung University Hospital, College of Medicine, Department of Pathology , Tainan, Taiwan
| | - Zhang-Dong Liang
- c 3 The University of Texas MD Anderson Cancer Center, Department of Translational Molecular Pathology , Houston, TX 77030, USA
| | - Wen-Bin Tsai
- c 3 The University of Texas MD Anderson Cancer Center, Department of Translational Molecular Pathology , Houston, TX 77030, USA
| | - Yan Long
- d 4 The University of Texas MD Anderson Cancer Center, Department of Translational Molecular Pathology , Houston, TX 77030, USA
| | - Isamu Aiba
- e 5 The University of Texas MD Anderson Cancer Center, Department of Translational Molecular Pathology , Houston, TX 77030, USA
| | - Siqing Fu
- f 6 The University of Texas MD Anderson Cancer Center, Departments of Investigative Cancer Therapeutics , Houston, TX, USA
| | - Russell Broaddus
- g 7 The University of Texas MD Anderson Cancer Center, Departments of Pathology , Houston, TX, USA
| | - Jinsong Liu
- g 7 The University of Texas MD Anderson Cancer Center, Departments of Pathology , Houston, TX, USA
| | - Lynn G Feun
- h 8 University of Miami, Sylvester Comprehensive Cancer Center , 1475 NW 12th Avenue, Miami, FL 33136, USA
| | - Niramol Savaraj
- h 8 University of Miami, Sylvester Comprehensive Cancer Center , 1475 NW 12th Avenue, Miami, FL 33136, USA
| | - Macus Tien Kuo
- i 9 The University of Texas MD Anderson Cancer Center, Department of Translational Molecular Pathology , Unit 2951, LSP 9.4206, 2130 W. Holcombe Blvd, Houston, TX 77030, USA +1 713 834 6038 ; +1 713 834 6085 ;
| |
Collapse
|
34
|
Pedraz-Cuesta E, Christensen S, Jensen AA, Jensen NF, Bunch L, Romer MU, Brünner N, Stenvang J, Pedersen SF. The glutamate transport inhibitor DL-Threo-β-Benzyloxyaspartic acid (DL-TBOA) differentially affects SN38- and oxaliplatin-induced death of drug-resistant colorectal cancer cells. BMC Cancer 2015; 15:411. [PMID: 25981639 PMCID: PMC4445981 DOI: 10.1186/s12885-015-1405-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/29/2015] [Indexed: 11/10/2022] Open
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer death globally and new biomarkers and treatments are severely needed. Methods Here, we employed HCT116 and LoVo human CRC cells made resistant to either SN38 or oxaliplatin, to investigate whether altered expression of the high affinity glutamate transporters Solute Carrier (SLC)-1A1 and -1A3 (EAAT3, EAAT1) is associated with the resistant phenotypes. Analyses included real-time quantitative PCR, immunoblotting and immunofluorescence analyses, radioactive tracer flux measurements, and biochemical analyses of cell viability and glutathione content. Results were evaluated using one- and two-way ANOVA and Students two-tailed t-test, as relevant. Results In SN38-resistant HCT116 and LoVo cells, SLC1A1 expression was down-regulated ~60 % and up-regulated ~4-fold, respectively, at both mRNA and protein level, whereas SLC1A3 protein was undetectable. The changes in SLC1A1 expression were accompanied by parallel changes in DL-Threo-β-Benzyloxyaspartic acid (TBOA)-sensitive, UCPH101-insensitive [3H]-D-Aspartate uptake, consistent with increased activity of SLC1A1 (or other family members), yet not of SLC1A3. DL-TBOA co-treatment concentration-dependently augmented loss of cell viability induced by SN38, while strongly counteracting that induced by oxaliplatin, in both HCT116 and LoVo cells. This reflected neither altered expression of the oxaliplatin transporter Cu2+-transporter-1 (CTR1), nor changes in cellular reduced glutathione (GSH), although HCT116 cell resistance per se correlated with increased cellular GSH. DL-TBOA did not significantly alter cellular levels of p21, cleaved PARP-1, or phospho-Retinoblastoma protein, yet altered SLC1A1 subcellular localization, and reduced chemotherapy-induced p53 induction. Conclusions SLC1A1 expression and glutamate transporter activity are altered in SN38-resistant CRC cells. Importantly, the non-selective glutamate transporter inhibitor DL-TBOA reduces chemotherapy-induced p53 induction and augments CRC cell death induced by SN38, while attenuating that induced by oxaliplatin. These findings may point to novel treatment options in treatment-resistant CRC. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1405-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena Pedraz-Cuesta
- Department of Biology, Faculty of Science, University of Copenhagen, 13, Universitetsparken, DK-2100, Copenhagen, Denmark.
| | - Sandra Christensen
- Department of Biology, Faculty of Science, University of Copenhagen, 13, Universitetsparken, DK-2100, Copenhagen, Denmark.
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, 13, Universitetsparken, DK-2100, Copenhagen, Denmark.
| | - Niels Frank Jensen
- Faculty of Health and Medical Sciences, Institute of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, 13, Universitetsparken, DK-2100, Copenhagen, Denmark.
| | - Maria Unni Romer
- Faculty of Health and Medical Sciences, Institute of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark. .,Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | - Nils Brünner
- Faculty of Health and Medical Sciences, Institute of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Jan Stenvang
- Faculty of Health and Medical Sciences, Institute of Veterinary Disease Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, 13, Universitetsparken, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
35
|
Wong PT, Choi SK. Mechanisms of Drug Release in Nanotherapeutic Delivery Systems. Chem Rev 2015; 115:3388-432. [DOI: 10.1021/cr5004634] [Citation(s) in RCA: 349] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pamela T. Wong
- Michigan
Nanotechnology Institute
for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Seok Ki Choi
- Michigan
Nanotechnology Institute
for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
36
|
Spreckelmeyer S, Orvig C, Casini A. Cellular transport mechanisms of cytotoxic metallodrugs: an overview beyond cisplatin. Molecules 2014; 19:15584-610. [PMID: 25268716 PMCID: PMC6271550 DOI: 10.3390/molecules191015584] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/17/2014] [Accepted: 09/22/2014] [Indexed: 12/21/2022] Open
Abstract
The field of medicinal inorganic chemistry has grown consistently during the past 50 years; however, metal-containing coordination compounds represent only a minor proportion of drugs currently on the market, indicating that research in this area has not yet been thoroughly realized. Although platinum-based drugs as cancer chemotherapeutic agents have been widely studied, exact knowledge of the mechanisms governing their accumulation in cells is still lacking. However, evidence suggests active uptake and efflux mechanisms are involved; this may be involved also in other experimental metal coordination and organometallic compounds with promising antitumor activities in vitro and in vivo, such as ruthenium and gold compounds. Such knowledge would be necessary to elucidate the balance between activity and toxicity profiles of metal compounds. In this review, we present an overview of the information available on the cellular accumulation of Pt compounds from in vitro, in vivo and clinical studies, as well as a summary of reports on the possible accumulation mechanisms for different families of experimental anticancer metal complexes (e.g., Ru Au and Ir). Finally, we discuss the need for rationalization of the investigational approaches available to study metallodrug cellular transport.
Collapse
Affiliation(s)
- Sarah Spreckelmeyer
- Dept. Pharmacokinetics, Toxicology and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T1Z1, Canada
| | - Angela Casini
- Dept. Pharmacokinetics, Toxicology and Targeting, Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, The Netherlands.
| |
Collapse
|
37
|
Scheiber IF, Mercer JF, Dringen R. Metabolism and functions of copper in brain. Prog Neurobiol 2014; 116:33-57. [DOI: 10.1016/j.pneurobio.2014.01.002] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 01/08/2014] [Accepted: 01/08/2014] [Indexed: 12/15/2022]
|
38
|
Schweigel-Röntgen M. The families of zinc (SLC30 and SLC39) and copper (SLC31) transporters. CURRENT TOPICS IN MEMBRANES 2014; 73:321-55. [PMID: 24745988 DOI: 10.1016/b978-0-12-800223-0.00009-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The solute carriers families 30 (SLC30; ZnT), 39 (SLC39; ZIP), and 31 (SLC31; CTR) are involved in the essential maintenance of cellular zinc (Zn²⁺) and copper (Cu²⁺) homeostasis, respectively. ZnTs mediate Zn²⁺ extrusion from cells (SLC30A1) or transport Zn²⁺ into organelles and secretory vesicles/granules (SLC30A2-SLC30A8). SLC39 family members are predominantly localized to the cell membrane where they perform Zn²⁺ uptake and increase the availability of cytosolic Zn²⁺. SLC39A1 is ubiquitously expressed, whereas other ZIP transporters (e.g., SLC39A2 and SLC39A3) show a more tissue-restricted expression consistent with organ-specific functions of these proteins. The members A1 (CTR1) and A2 (CTR2) of the SLC31 family of solute carriers belong to a network of proteins that acts to regulate the intracellular Cu²⁺ concentration within a certain range. SLC31A1 is predominantly localized to the plasma membrane, whereas SLC31A2 is mainly found in intracellular membranes of the late endosome and lysosome. The specific function of SLC31A2 is not known. SLC31A1 is ubiquitously expressed and has been characterized as a high-affinity importer of reduced copper (Cu⁺). Cu²⁺ transport function of CTR proteins is associated with oligomerization; SLC31A1 trimerizes and thereby forms a channel-like structure enabling Cu²⁺ translocation across the cell membrane. The molecular characteristics and structural details (e.g., membrane topology, conserved Zn²⁺, and Cu²⁺ binding sites) and mechanisms of translational and posttranslational regulation of expression and/or activity have been described for SLC30 and SLC39 family members, and for SLC31A1. For SLC31A1, data on tissue-specific functions (e.g., in the intestine, heart, and liver) are also available. A link between SLC31A1, immune function, and disorders such as Alzheimer's disease or cancer makes the protein a candidate therapeutic target. In secretory tissues (e.g., the mammary gland and pancreas), Zn²⁺ transporters of SLC families 30 and 39 are involved in specific functions such as insulin synthesis and secretion, metallation of digestive proenzymes, and transfer of nutrients into milk. Defective or dysregulated Zn²⁺ metabolism in these organs is associated with disorders such as diabetes and cancer, and impaired Zn²⁺ secretion into milk.
Collapse
Affiliation(s)
- Monika Schweigel-Röntgen
- Institute for Muscle Biology & Growth, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany.
| |
Collapse
|
39
|
Fu S, Hou MM, Wheler J, Hong D, Naing A, Tsimberidou A, Janku F, Zinner R, Piha-Paul S, Falchook G, Kuo MT, Kurzrock R. Exploratory study of carboplatin plus the copper-lowering agent trientine in patients with advanced malignancies. Invest New Drugs 2013; 32:465-72. [PMID: 24306314 DOI: 10.1007/s10637-013-0051-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 11/15/2013] [Indexed: 01/03/2023]
Abstract
PURPOSE Preclinical data showed that trientine, a copper-lowering agent, re-sensitized cancer cells to carboplatin through enhanced human copper transporter 1 (hCtr1) -mediated platinum uptake. EXPERIMENTAL DESIGN We studied carboplatin and trientine in patients (n = 55; 45 who had failed platinum) with advanced malignancies (Phase I, modified 3 + 3 design). RESULTS The most common cancers were head and neck (n = 13), non-small cell lung (n = 10) and epithelial ovarian (n = 8). The median number of prior regimens was four. No dose-limiting toxicity or treatment-related deaths were observed at doses up to carboplatin AUC 6 given with trientine. Eight patients achieved stable disease (SD) ≥ 6 months (six platinum failures) and one patient with platinum-resistant ovarian cancer, partial response (PR) (total SD ≥ 6 months/PR = 9, 16.4 %). The mean nadir serum copper level in the nine patients with SD ≥ 6 months/PR was 0.55 μg/mL (95 % CI, 0.34-0.75) versus 1.22 μg/mL (95 % CI, 1.02-1.42) (p < 0.001) in 38 tested patients with progression. In patients who maintained their ceruloplasmin (major copper-carrying protein) levels at 5-15 mg/dL (n = 9), the median progression-free and overall survivals were 9.2 and 15.2 months versus 1.9 (p = 0.001) and 5.7 months (p = 0.033) in patients who did not (n = 38), respectively. CONCLUSIONS The combination of a copper-lowering agent with carboplatin was well tolerated and associated with antitumor activity, especially in patients in whom copper and/or ceruloplasmin levels were lowered. Further investigation of this strategy for reversing platinum resistance is warranted.
Collapse
Affiliation(s)
- Siqing Fu
- Department of Investigational Cancer Therapeutics, Unit 0455, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Liang ZD, Long Y, Chen HHW, Savaraj N, Kuo MT. Regulation of the high-affinity copper transporter (hCtr1) expression by cisplatin and heavy metals. J Biol Inorg Chem 2013; 19:17-27. [PMID: 24132751 PMCID: PMC3889686 DOI: 10.1007/s00775-013-1051-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 10/03/2013] [Indexed: 01/11/2023]
Abstract
Platinum-based antitumor agents have been the mainstay in cancer chemotherapy for many human malignancies. Drug resistance is an important obstacle to achieving the maximal therapeutic efficacy of these drugs. Understanding how platinum drugs enter cells is of great importance in improving therapeutic efficacy. It has been demonstrated that human high-affinity copper transporter 1 (hCtr1) is involved in transporting cisplatin into cells to elicit cytotoxic effects, although other mechanisms may exist. In this communication, we demonstrate that cisplatin transcriptionally induces the expression of hCtr1 in time- and concentration-dependent manners. Cisplatin functions as a competitor for hCtr1-mediated copper transport, resulting in reduced cellular copper levels and leading to upregulated expression of Sp1, which is a positive regulator for hCtr1 expression. Thus, regulation of hCtr1 expression by cisplatin is an integral part of the copper homeostasis regulation system. We also demonstrate that Ag(I) and Zn(II), which are known to suppress hCtr1-mediated copper transport, can also induce hCtr1/Sp1 expression. In contrast, Cd(II), another inhibitor of copper transport, downregulates hCtr1 expression by suppressing Sp1 expression. Collectively, our results demonstrate diverse mechanisms of regulating copper metabolism by these heavy metals.
Collapse
Affiliation(s)
- Zheng Dong Liang
- Department of Translational Molecular Pathology (Route 2951), The University of Texas MD Anderson Cancer Center, 2130 W. Holcombe Blvd., Houston, TX 77030 USA
| | - Yan Long
- Department of Translational Molecular Pathology (Route 2951), The University of Texas MD Anderson Cancer Center, 2130 W. Holcombe Blvd., Houston, TX 77030 USA
| | - Helen H. W. Chen
- Department of Radiation Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Niramol Savaraj
- Hematology-Oncology Section, VA Medical Center, Miami, FL 33125 USA
| | - Macus Tien Kuo
- Department of Translational Molecular Pathology (Route 2951), The University of Texas MD Anderson Cancer Center, 2130 W. Holcombe Blvd., Houston, TX 77030 USA
| |
Collapse
|
41
|
Shoeib T, Sharp BL. Monomeric cisplatin complexes with glutathione: Coordination modes and binding affinities. Inorganica Chim Acta 2013. [DOI: 10.1016/j.ica.2013.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Maryon EB, Molloy SA, Kaplan JH. Cellular glutathione plays a key role in copper uptake mediated by human copper transporter 1. Am J Physiol Cell Physiol 2013; 304:C768-79. [PMID: 23426973 DOI: 10.1152/ajpcell.00417.2012] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Copper is an essential micronutrient. Following entry via the human copper transporter 1 (hCTR1), copper is delivered to several copper chaperones, which subsequently transfer the metal to specific targets via protein:protein interactions. It is has been assumed, but not demonstrated, that chaperones acquire copper directly from hCTR1. However, some reports have pointed to an intermediary role for glutathione (GSH), an abundant copper-binding tri-peptide. To address the issue of how transported copper is acquired by the copper chaperones in vivo, we measured the initial rate of (64)Cu uptake in cells in which the cellular levels of copper chaperones or GSH were substantially depleted or elevated. Knockdown or overexpression of copper chaperones ATOX1, CCS, or both had no effect on the initial rate of (64)Cu entry into HEK293 cells having endogenous or overexpressed hCTR1. In contrast, depleting cellular GSH using L-buthionine-sulfoximine (BSO) caused a 50% decrease in the initial rate of (64)Cu entry in HEK293 cells and other cell types. This decrease was reversed by washout of BSO or GSH replenishment with a permeable ester. BSO treatment under our experimental conditions had no significant effects on the viability, ATP levels, or metal content of the cells. Attenuated (64)Cu uptake in BSO was not due to oxidation of the cysteine in the putative metal-binding motif (HCH) at the intracellular hCTR1 COOH terminus, because a mutant lacking this motif was fully active, and (64)Cu uptake was still reduced by BSO treatment. Our data suggest that GSH plays an important role in copper handling at the entry step.
Collapse
Affiliation(s)
- Edward B Maryon
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60607, USA
| | | | | |
Collapse
|
43
|
Wu WJ, Zhang Y, Zeng ZL, Li XB, Hu KS, Luo HY, Yang J, Huang P, Xu RH. β-phenylethyl isothiocyanate reverses platinum resistance by a GSH-dependent mechanism in cancer cells with epithelial-mesenchymal transition phenotype. Biochem Pharmacol 2012; 85:486-96. [PMID: 23219523 DOI: 10.1016/j.bcp.2012.11.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 11/20/2012] [Accepted: 11/26/2012] [Indexed: 12/18/2022]
Abstract
Platinum (Pt)-based chemotherapy is an important regimen in the clinical treatment of cancer, but development of drug resistance presents a major challenge. One key mechanism involved in resistance to Pt drugs is the decrease of intracellular Pt due to the drug efflux through the glutathione (GSH)-mediated export, and this is particularly significant in cancer cells with stem-cell like properties. In the present study, we showed that two Pt-resistant human cancer cell lines exhibited stem-cell like EMT properties, had high cellular GSH and accumulated significantly less cellular Pt compared to their parental cells, and failed to undergo apoptosis when exposed to Pt at the drug concentrations toxic to the parental cells. Importantly, we found that the natural compound β-phenylethyl isothiocyanate (PEITC) was able to effectively abolish this drug resistant mechanism by effective depletion of cellular GSH, leading to a significant increase in cellular Pt as well as DNA-bound Pt. A combination of PEITC and Pt showed a striking synergistic anticancer activity both in vitro and in vivo, as evidenced by an increase in drug-induced apoptosis, a loss of colony formation capacity, and significant suppression of tumor growth in mice. Taken together, our study shows a promising therapeutic strategy to overcome drug resistance to platinum-based chemotherapy and may potentially have broad implications in clinical treatment of cancer.
Collapse
Affiliation(s)
- Wen-jing Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Wexselblatt E, Gibson D. What do we know about the reduction of Pt(IV) pro-drugs? J Inorg Biochem 2012; 117:220-9. [DOI: 10.1016/j.jinorgbio.2012.06.013] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 06/23/2012] [Accepted: 06/25/2012] [Indexed: 10/28/2022]
|
46
|
Liang ZD, Long Y, Tsai WB, Fu S, Kurzrock R, Gagea-Iurascu M, Zhang F, Chen HH, Hennessy BT, Mills GB, Savaraj N, Kuo MT. Mechanistic basis for overcoming platinum resistance using copper chelating agents. Mol Cancer Ther 2012; 11:2483-94. [PMID: 22914438 PMCID: PMC3496003 DOI: 10.1158/1535-7163.mct-12-0580] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Platinum-based antitumor agents are widely used in cancer chemotherapy. Drug resistance is a major obstacle to the successful use of these agents because once drug resistance develops, other effective treatment options are limited. Recently, we conducted a clinical trial using a copper-lowering agent to overcome platinum drug resistance in ovarian cancer patients and the preliminary results are encouraging. In supporting this clinical study, using three pairs of cisplatin (cDDP)-resistant cell lines and two ovarian cancer cell lines derived from patients who had failed in platinum-based chemotherapy, we showed that cDDP resistance associated with reduced expression of the high-affinity copper transporter (hCtr1), which is also a cDDP transporter, can be preferentially resensitized by copper-lowering agents because of enhanced hCtr1 expression, as compared with their drug-sensitive counterparts. Such a preferential induction of hCtr1 expression in cDDP-resistant variants by copper chelation can be explained by the mammalian copper homeostasis regulatory mechanism. Enhanced cell-killing efficacy by a copper-lowering agent was also observed in animal xenografts bearing cDDP-resistant cells. Finally, by analyzing a public gene expression dataset, we found that ovarian cancer patients with elevated levels of hCtr1 in their tumors, but not ATP7A and ATP7B, had more favorable outcomes after platinum drug treatment than those expressing low hCtr1 levels. This study reveals the mechanistic basis for using copper chelation to overcome cDDP resistance in clinical investigations.
Collapse
Affiliation(s)
- Zheng D. Liang
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yan Long
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wen-Bin Tsai
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigative Cancer Therapeutics (Phase I Clinical Trial Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Razelle Kurzrock
- Department of Investigative Cancer Therapeutics (Phase I Clinical Trial Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mihai Gagea-Iurascu
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fan Zhang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Helen H.W. Chen
- Department of Radiation Oncology, Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Gordon B. Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Niramol Savaraj
- Hematology-Oncology Section, Veteran Affairs Medical Center, Miami, Florida
| | - Macus Tien Kuo
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
47
|
Drayton RM, Catto JWF. Molecular mechanisms of cisplatin resistance in bladder cancer. Expert Rev Anticancer Ther 2012; 12:271-81. [PMID: 22316374 DOI: 10.1586/era.11.201] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metastatic disease is the most common mechanism of death in patients with advanced bladder cancer. As for most solid tumors, chemotherapy remains the only realistic option for palliating or curing metastatic disease. However, bladder cancer is characterized by chemoresistance. Only modest response rates are obtained using multiagent regimens including cisplatin. These low response rates and the toxicity of these regimens limit their use to patients at highest risk. Here, we review the molecular mechanisms of cisplatin resistance. These include methods to reduce cisplatin bioavailability within a cell, and defects in the machinery that produces cell death following cisplatin-induced DNA damage. While overcoming these mechanisms is a potential therapeutic approach that can increase response rates, in the short term this knowledge could be used to predict response in individual tumors.
Collapse
Affiliation(s)
- Ross M Drayton
- Institute for Cancer Studies and Academic Urology Unit, University of Sheffield, Sheffield, S10 2RX, UK.
| | | |
Collapse
|
48
|
Kuo MT, Fu S, Savaraj N, Chen HHW. Role of the human high-affinity copper transporter in copper homeostasis regulation and cisplatin sensitivity in cancer chemotherapy. Cancer Res 2012; 72:4616-21. [PMID: 22962276 PMCID: PMC3445735 DOI: 10.1158/0008-5472.can-12-0888] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The high-affinity copper transporter (Ctr1; SCLC31A1) plays an important role in regulating copper homeostasis because copper is an essential micronutrient and copper deficiency is detrimental to many important cellular functions, but excess copper is toxic. Recent research has revealed that human copper homeostasis is tightly controlled by interregulatory circuitry involving copper, Sp1, and human (hCtr1). This circuitry uses Sp1 transcription factor as a copper sensor in modulating hCtr1 expression, which in turn controls cellular copper and Sp1 levels in a 3-way mutual regulatory loop. Posttranslational regulation of hCtr1 expression by copper stresses has also been described in the literature. Because hCtr1 can also transport platinum drugs, this finding underscores the important role of hCtr1 in platinum-drug sensitivity in cancer chemotherapy. Consistent with this notion is the finding that elevated hCtr1 expression was associated with favorable treatment outcomes in cisplatin-based cancer chemotherapy. Moreover, cultured cell studies showed that elevated hCtr1 expression can be induced by depleting cellular copper levels, resulting in enhanced cisplatin uptake and its cell-killing activity. A phase I clinical trial using a combination of trientine (a copper chelator) and carboplatin has been carried out with encouraging results. This review discusses new insights into the role of hCtr1 in regulating copper homeostasis and explains how modulating cellular copper availability could influence treatment efficacy in platinum-based cancer chemotherapy through hCtr1 regulation.
Collapse
Affiliation(s)
- Macus Tien Kuo
- Department of Molecular Pathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas 77054, USA.
| | | | | | | |
Collapse
|
49
|
Fu S, Naing A, Fu C, Kuo MT, Kurzrock R. Overcoming platinum resistance through the use of a copper-lowering agent. Mol Cancer Ther 2012; 11:1221-5. [PMID: 22491798 DOI: 10.1158/1535-7163.mct-11-0864] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Low levels of human copper transporter 1 (hCtr1) mRNA are associated with a shorter progression-free survival after platinum-based therapy. Pretreatment with a copper-lowering agent such as trientine enhanced hCtr1-mediated platinum uptake. Therefore, we conducted a pilot study (NCT01178112) of carboplatin and trientine with the goal of resensitizing patients with advanced cancer to platinum chemotherapy. This case report reviews the outcomes of 5 patients with platinum-resistant high-grade epithelial ovarian cancer enrolled on the study to date. Overall, they tolerated treatment well. Severe adverse events that occurred in 2 patients were myelosuppression, notably anemia requiring transfusion. Dose-limiting toxicity was not observed within the first 28 days (cycle 1). After 2 cycles of therapy, partial remission was achieved in 1 patient (10+ months), stable disease in 3 patients (2, 3.5+, and 5 months, respectively), and 1 patient had progressive disease. These cases provide preliminary clinical evidence that the role of decreasing copper levels in reversing platinum resistance merits additional clinical investigation. Evaluation of this novel strategy is warranted in larger studies to assess the efficacy of this approach for treating platinum-resistant advanced epithelial ovarian cancer in patients with high copper levels.
Collapse
Affiliation(s)
- Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | |
Collapse
|
50
|
Wangpaichitr M, Theodoropoulos G, Wu C, You M, Feun LG, Kuo MT, Savaraj N. The relationship of thioredoxin-1 and cisplatin resistance: its impact on ROS and oxidative metabolism in lung cancer cells. Mol Cancer Ther 2012; 11:604-15. [PMID: 22248473 PMCID: PMC3326609 DOI: 10.1158/1535-7163.mct-11-0599] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Elimination of cisplatin-resistant lung cancer cells remains a major obstacle. We have shown that cisplatin-resistant tumors have higher reactive oxygen species (ROS) levels and can be exploited for targeted therapy. Here, we show that increased secretion of the antioxidant thioredoxin-1 (TRX1) resulted in lowered intracellular TRX1 and contributed to higher ROS in cisplatin-resistant tumors in vivo and in vitro. By reconstituting TRX1 protein in cisplatin-resistant cells, we increased sensitivity to cisplatin but decreased sensitivity to elesclomol (ROS inducer). Conversely, decreased TRX1 protein in parental cells reduced the sensitivity to cisplatin but increased sensitivity to elesclomol. Cisplatin-resistant cells had increased endogenous oxygen consumption and mitochondrial activity but decreased lactic acid production. They also exhibited higher levels of argininosuccinate synthetase (ASS) and fumarase mRNA, which contributed to oxidative metabolism (OXMET) when compared with parental cells. Restoring intracellular TRX1 protein in cisplatin-resistant cells resulted in lowering ASS and fumarase mRNAs, which in turn sensitized them to arginine deprivation. Interestingly, cisplatin-resistant cells also had significantly higher basal levels of acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS). Overexpressing TRX1 lowered ACC and FAS proteins expressions in cisplatin-resistant cells. Chemical inhibition and short interfering RNA of ACC resulted in significant cell death in cisplatin-resistant compared with parental cells. Conversely, TRX1 overexpressed cisplatin-resistant cells resisted 5-(tetradecyloxy)-2-furoic acid (TOFA)-induced death. Collectively, lowering TRX1 expression through increased secretion leads cisplatin-resistant cells to higher ROS production and increased dependency on OXMET. These changes raise an intriguing therapeutic potential for future therapy in cisplatin-resistant lung cancer.
Collapse
Affiliation(s)
| | | | - Chunjing Wu
- Department of Veterans Affairs, South Florida VA Foundation, Miami, FL
| | - Min You
- Department of Medicine, Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL
| | - Lynn G. Feun
- Department of Medicine, Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL
| | - Macus T. Kuo
- Department of Molecular Pathology, MD Anderson Cancer Center, Houston, TX
| | - Niramol Savaraj
- Department of Veterans Affairs, South Florida VA Foundation, Miami, FL
- Department of Medicine, Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|