1
|
Xue R, Qin C, Li L, Huang L, Tang K, Chen J, Liang H, Xu H, Qin X, Yang C, Tan Q. SRF/SLC31A1 signaling promotes cuproptosis induced by celastrol in NSCLC. Int Immunopharmacol 2025; 148:114165. [PMID: 39930648 DOI: 10.1016/j.intimp.2025.114165] [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: 10/31/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 05/08/2025]
Abstract
Faced with the highly malignant challenge of lung cancer, traditional chemotherapeutic agents, although predominantly inducing apoptosis, are severely limited in their therapeutic effect by the overexpression of anti-apoptotic proteins in lung cancer. Recently discovered copper-induced non-apoptotic cell death, known as cuproptosis, represents a novel mechanism for regulating cell death. Whether Celastrol (Cel), a potential anti-tumor drug, can counter non-small cell lung cancer (NSCLC) through inducing cuproptosis remains to be thoroughly investigated. This study demonstrates that the copper chelator tetrathiomolybdate (TTM) is more effective in rescuing Cel-induced NSCLC cell death compared to other inhibitors. RNA sequencing revealed that Cel significantly upregulates the copper transporter protein SLC31A1. In addition, Cel also promotes intracellular copper accumulation, reduces GSH levels, and exhibits features of cuproptosis, including loss of iron-sulfur cluster proteins (FDX1, SDHB, POLD1), increased HSP70, and DLAT oligomerization. Experiments also found that Cel significantly increases reactive oxygen species (ROS) levels, reduces mitochondrial membrane potential, and lowers ATP levels. It was predicted through online databases that SRF may be the transcription factor for SLC31A1, and this was validated through overexpression experiments. In vivo data demonstrated that Cel significantly inhibits tumor growth without damaging the heart, liver, or kidneys of mice. This study first reveals that celastrol disrupts intracellular copper homeostasis through the SRF/SLC31A1 pathway, promoting cuproptosis in NSCLC cells, providing support for Cel as a potential safe and effective chemotherapeutic agent.
Collapse
Affiliation(s)
- Rui Xue
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Chuling Qin
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Lanyu Li
- Guilin Medical University Laboratory Animal Center, Guilin 541001 China
| | - Lingyue Huang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Ke Tang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Jianning Chen
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Huihui Liang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Huimin Xu
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Xuanjie Qin
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Chang Yang
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China
| | - Qinyou Tan
- School of Pharmacy of Guilin Medical University, Guilin 541001 China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001 China; Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, Affiliated Hospital of Guilin Medical University, Guilin 541001 China; China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001 China.
| |
Collapse
|
2
|
Pan C, Ji Z, Wang Q, Zhang Z, Wang Z, Li C, Lu S, Ge P. Cuproptosis: Mechanisms, biological significance, and advances in disease treatment-A systematic review. CNS Neurosci Ther 2024; 30:e70039. [PMID: 39267265 PMCID: PMC11392831 DOI: 10.1111/cns.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Copper is an essential trace element for biological systems, as it plays a critical role in the activity of various enzymes and metabolic processes. However, the dysregulation of copper homeostasis is closely associated with the onset and progression of numerous diseases. In recent years, copper-induced cell death, a novel form of cellular demise, has garnered significant attention. This process is characterized by the abnormal accumulation of intracellular copper ions, leading to cellular dysfunction and eventual cell death. Copper toxicity occurs through the interaction of copper with acylated enzymes in the tricarboxylic acid (TCA) cycle. This interaction results in subsequent protein aggregation, causing proteotoxic stress and ultimately resulting in cell death. Despite the promise of these findings, the detailed mechanisms and broader implications of cuproptosis remain underexplored. Therefore, our study aimed to investigate the role of copper in cell death and autophagy, focusing on the molecular mechanisms of cuproptosis. We also aimed to discuss recent advancements in copper-related research across various diseases and tumors, providing insights for future studies and potential therapeutic applications. MAIN BODY This review delves into the biological significance of copper metabolism and the molecular mechanisms underlying copper-induced cell death. Furthermore, we discuss the role of copper toxicity in the pathogenesis of various diseases, emphasizing recent advancements in the field of oncology. Additionally, we explore the therapeutic potential of targeting copper toxicity. CONCLUSION The study highlights the need for further research to explore alternative pathways of copper-induced cell death, detailed mechanisms of cuproptosis, and biomarkers for copper poisoning. Future research should focus on exploring the molecular mechanisms of cuproptosis, developing new therapeutic strategies, and verifying their safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Chengliang Pan
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Zhilin Ji
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Qingxuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Zhao Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Zhenchuan Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Chen Li
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Shan Lu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| | - Pengfei Ge
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
3
|
Tu D, Xu Q, Luan Y, Sun J, Zuo X, Ma C. Integrative analysis of bioinformatics and machine learning to identify cuprotosis-related biomarkers and immunological characteristics in heart failure. Front Cardiovasc Med 2024; 11:1349363. [PMID: 38562184 PMCID: PMC10982316 DOI: 10.3389/fcvm.2024.1349363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Backgrounds Cuprotosis is a newly discovered programmed cell death by modulating tricarboxylic acid cycle. Emerging evidence showed that cuprotosis-related genes (CRGs) are implicated in the occurrence and progression of multiple diseases. However, the mechanism of cuprotosis in heart failure (HF) has not been investigated yet. Methods The HF microarray datasets GSE16499, GSE26887, GSE42955, GSE57338, GSE76701, and GSE79962 were downloaded from the Gene Expression Omnibus (GEO) database to identify differentially expressed CRGs between HF patients and nonfailing donors (NFDs). Four machine learning models were used to identify key CRGs features for HF diagnosis. The expression profiles of key CRGs were further validated in a merged GEO external validation dataset and human samples through quantitative reverse-transcription polymerase chain reaction (qRT-PCR). In addition, Gene Ontology (GO) function enrichment, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, and immune infiltration analysis were used to investigate potential biological functions of key CRGs. Results We discovered nine differentially expressed CRGs in heart tissues from HF patients and NFDs. With the aid of four machine learning algorithms, we identified three indicators of cuprotosis (DLAT, SLC31A1, and DLST) in HF, which showed good diagnostic properties. In addition, their differential expression between HF patients and NFDs was confirmed through qRT-PCR. Moreover, the results of enrichment analyses and immune infiltration exhibited that these diagnostic markers of CRGs were strongly correlated to energy metabolism and immune activity. Conclusions Our study discovered that cuprotosis was strongly related to the pathogenesis of HF, probably by regulating energy metabolism-associated and immune-associated signaling pathways.
Collapse
Affiliation(s)
- Dingyuan Tu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Shenyang, Liaoning, China
- Department of Cardiology, The 961st Hospital of PLA Joint Logistic Support Force, Qiqihar, Heilongjiang, China
| | - Qiang Xu
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Department of Cardiology, Navy 905 Hospital, Naval Medical University, Shanghai, China
| | - Yanmin Luan
- Reproductive Medicine Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Sun
- Hospital-Acquired Infection Control Department, Yantai Ludong Hospital, Yantai, Shandong, China
| | - Xiaoli Zuo
- Department of Cardiology, The 961st Hospital of PLA Joint Logistic Support Force, Qiqihar, Heilongjiang, China
| | - Chaoqun Ma
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Shenyang, Liaoning, China
| |
Collapse
|
4
|
Liu J, Liu Y, Yang C, Liu J, Hao J. Comprehensive analysis for the immune related biomarkers of platinum-based chemotherapy in ovarian cancer. Transl Oncol 2023; 37:101762. [PMID: 37619523 PMCID: PMC10458992 DOI: 10.1016/j.tranon.2023.101762] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/26/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is one of the most lethal gynecological malignancies. This study aimed to identify biomarkers that were sensitive to platinum-based chemotherapeutic agents and can be used in immunotherapy and explore the importance of their mechanisms of action. METHODS RNA-seq profiles and clinicopathological data for OC samples were obtained from The Cancer Genome Atlas (TCGA) and cBioPortal platform, respectively. Platinum-sensitive and platinum-resistant OC samples in the TCGA cohort were selected based on the clinical information. RNA-seq data for 70 OC samples withSingle-sample gene set enrichment analysis (ssGSEA) and unsupervised clustering were used to classify OC patients from the TCGA cohort into clusters with different proportions of infiltrating immune cells. ESTIMATE analysis was used to assess the immune landscape among clusters. Differential expression, univariate Cox regression, and LASSO regression analyses were performed to construct prognostic model. Spearman correlation analysis was conducted to investigate the correlations among immune checkpoint inhibitors (ICIs) and risk score, half-maximal drug inhibitory concentration (IC50) and risk score. RESULTS Using ssGSEA and unsupervised clustering, OC samples were divided into two clusters with different immune cell infiltration. Then, 1715 differentially expressed immune-related genes (DEIRGs) were identified between two clusters, 984 differentially expressed platinum-sensitive related genes (DEPSRGs) between 149 platinum-sensitive and 63 platinum-resistant OC samples were identified, and 5384 differentially expressed genes (DEGs) between 380 OC and 194 normal samples were detected from the TCGA cohort. Six biomarkers (GMPPB, SRPK1, STC1, PRSS16, HPDL, and SPTSSB) were detected to establish a prognostic model. The OC patients in the TCGA cohort were classified into high- and low-risk groups. The receive operating characteristic (ROC) curve was plotted and demonstrated that the prognostic model performed well with the area under ROC curve (AUC) greater than 0.6. The expressions of 5 ICIs, including CD200, TNFRSF18, CD160, CD200R1, and CD274 (PD-L1), were significantly different between two risk groups, and the risk score was significant negative associated with CTLA4, TNFRSF4, TNFRSF18, and CD274. Moreover, there were significant differences in IC50 of 10 chemo drugs between two risk groups, patients in the high-risk group could be more resistant to po0tinib, dasatinib, and neratinib. CONCLUSION In summary, this study constructed a novel prognostic model based on six prognostic biomarkers, including GMPPB, SRPK1, STC1, PRSS16, HPDL, and SPTSSB, which can be utilized for predicting the prognosis of OC patients. These biomarkers were the potential therapeutic targets.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Gynecology, Benxi Central Hospital, Benxi 117000, Liaoning Province, China
| | - Yaoyao Liu
- Department of Gynecology, Benxi Central Hospital, Benxi 117000, Liaoning Province, China
| | - Chunjiao Yang
- Department of Radiotheropy, Benxi Central Hospital, Benxi 117000, Liaoning Province, China
| | - Jingjing Liu
- Department of Gynecology, Benxi Central Hospital, Benxi 117000, Liaoning Province, China
| | - Jiaxin Hao
- Department of Orthopedics, Benxi Central Hospital, Benxi 117000, Liaoning Province, China.
| |
Collapse
|
5
|
Qasem Z, Pavlin M, Ritacco I, Avivi MY, Meron S, Hirsch M, Shenberger Y, Gevorkyan-Airapetov L, Magistrato A, Ruthstein S. Disrupting Cu trafficking as a potential therapy for cancer. Front Mol Biosci 2022; 9:1011294. [PMID: 36299299 PMCID: PMC9589254 DOI: 10.3389/fmolb.2022.1011294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Copper ions play a crucial role in various cellular biological processes. However, these copper ions can also lead to toxicity when their concentration is not controlled by a sophisticated copper-trafficking system. Copper dys-homeostasis has been linked to a variety of diseases, including neurodegeneration and cancer. Therefore, manipulating Cu-trafficking to trigger selective cancer cell death may be a viable strategy with therapeutic benefit. By exploiting combined in silico and experimental strategies, we identified small peptides able to bind Atox1 and metal-binding domains 3-4 of ATP7B proteins. We found that these peptides reduced the proliferation of cancer cells owing to increased cellular copper ions concentration. These outcomes support the idea of harming copper trafficking as an opportunity for devising novel anti-cancer therapies.
Collapse
Affiliation(s)
- Zena Qasem
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Matic Pavlin
- National Research Council of Italy (CNR)—Institute of Material (IOM) C/o International School for Advanced Studies (SISSA), Trieste, Italy
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Ljubljana, Slovenia
| | - Ida Ritacco
- National Research Council of Italy (CNR)—Institute of Material (IOM) C/o International School for Advanced Studies (SISSA), Trieste, Italy
- Department of Chemistry, University of Salerno, Salerno, Italy
| | - Matan Y. Avivi
- The Mina and Everard Goodman Faculty of Life-Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Shelly Meron
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Melanie Hirsch
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Yulia Shenberger
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Lada Gevorkyan-Airapetov
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Alessandra Magistrato
- National Research Council of Italy (CNR)—Institute of Material (IOM) C/o International School for Advanced Studies (SISSA), Trieste, Italy
- *Correspondence: Alessandra Magistrato, ; Sharon Ruthstein,
| | - Sharon Ruthstein
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
- *Correspondence: Alessandra Magistrato, ; Sharon Ruthstein,
| |
Collapse
|
6
|
de Brito RV, Mancini MW, Palumbo MDN, de Moraes LHO, Rodrigues GJ, Cervantes O, Sercarz JA, Paiva MB. The Rationale for "Laser-Induced Thermal Therapy (LITT) and Intratumoral Cisplatin" Approach for Cancer Treatment. Int J Mol Sci 2022; 23:5934. [PMID: 35682611 PMCID: PMC9180481 DOI: 10.3390/ijms23115934] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 01/27/2023] Open
Abstract
Cisplatin is one of the most widely used anticancer drugs in the treatment of various types of solid human cancers, as well as germ cell tumors, sarcomas, and lymphomas. Strong evidence from research has demonstrated higher efficacy of a combination of cisplatin and derivatives, together with hyperthermia and light, in overcoming drug resistance and improving tumoricidal efficacy. It is well known that the antioncogenic potential of CDDP is markedly enhanced by hyperthermia compared to drug treatment alone. However, more recently, accelerators of high energy particles, such as synchrotrons, have been used to produce powerful and monochromatizable radiation to induce an Auger electron cascade in cis-platinum molecules. This is the concept that makes photoactivation of cis-platinum theoretically possible. Both heat and light increase cisplatin anticancer activity via multiple mechanisms, generating DNA lesions by interacting with purine bases in DNA followed by activation of several signal transduction pathways which finally lead to apoptosis. For the past twenty-seven years, our group has developed infrared photo-thermal activation of cisplatin for cancer treatment from bench to bedside. The future development of photoactivatable prodrugs of platinum-based agents injected intratumorally will increase selectivity, lower toxicity and increase efficacy of this important class of antitumor drugs, particularly when treating tumors accessible to laser-based fiber-optic devices, as in head and neck cancer. In this article, the mechanistic rationale of combined intratumor injections of cisplatin and laser-induced thermal therapy (CDDP-LITT) and the clinical application of such minimally invasive treatment for cancer are reviewed.
Collapse
Affiliation(s)
- Renan Vieira de Brito
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
| | - Marília Wellichan Mancini
- Biophotonics Department, Institute of Research and Education in the Health Area (NUPEN), Sao Carlos 13562-030, SP, Brazil;
| | - Marcel das Neves Palumbo
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
| | - Luis Henrique Oliveira de Moraes
- Department of Physiological Sciences, Federal University of Sao Carlos (UFSCar), Sao Carlos 13565-905, SP, Brazil; (L.H.O.d.M.); (G.J.R.)
| | - Gerson Jhonatan Rodrigues
- Department of Physiological Sciences, Federal University of Sao Carlos (UFSCar), Sao Carlos 13565-905, SP, Brazil; (L.H.O.d.M.); (G.J.R.)
| | - Onivaldo Cervantes
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
| | - Joel Avram Sercarz
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Marcos Bandiera Paiva
- Department of Otolaryngology and Head and Neck Surgery, Federal University of São Paulo (UNIFESP), Sao Paulo 04023-062, SP, Brazil; (R.V.d.B.); (M.d.N.P.); (O.C.)
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
7
|
Walke G, Aupič J, Kashoua H, Janoš P, Meron S, Shenberger Y, Qasem Z, Gevorkyan-Airapetov L, Magistrato A, Ruthstein S. Dynamical interplay between the human high-affinity copper transporter hCtr1 and its cognate metal ion. Biophys J 2022; 121:1194-1204. [PMID: 35202609 PMCID: PMC9034245 DOI: 10.1016/j.bpj.2022.02.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/12/2021] [Accepted: 02/17/2022] [Indexed: 11/02/2022] Open
Abstract
Abnormal cellular copper levels have been clearly implicated in genetic diseases, cancer, and neurodegeneration. Ctr1, a high-affinity copper transporter, is a homotrimeric integral membrane protein that provides the main route for cellular copper uptake. Together with a sophisticated copper transport system, Ctr1 regulates Cu(I) metabolism in eukaryotes. Despite its pivotal role in normal cell function, the molecular mechanism of copper uptake and transport via Ctr1 remains elusive. In this study, electron paramagnetic resonance (EPR), UV-visible spectroscopy, and all-atom simulations were employed to explore Cu(I) binding to full-length human Ctr1 (hCtr1), thereby elucidating how metal binding at multiple distinct sites affects the hCtr1 conformational dynamics. We demonstrate that each hCtr1 monomer binds up to five Cu(I) ions and that progressive Cu(I) binding triggers a marked structural rearrangement in the hCtr1 C-terminal region. The observed Cu(I)-induced conformational remodeling suggests that the C-terminal region may play a dual role, serving both as a channel gate and as a shuttle mediating the delivery of copper ions from the extracellular hCtr1 selectivity filter to intracellular metallochaperones. Our findings thus contribute to a more complete understanding of the mechanism of hCtr1-mediated Cu(I) uptake and provide a conceptual basis for developing mechanism-based therapeutics for treating pathological conditions linked to de-regulated copper metabolism.
Collapse
Affiliation(s)
- Gulshan Walke
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Jana Aupič
- Department National Research Council of Italy (CNR) - Institute of Material (IOM) c/o International School for Advanced Studies (SISSA), Trieste, Italy
| | - Hadeel Kashoua
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Pavel Janoš
- Department National Research Council of Italy (CNR) - Institute of Material (IOM) c/o International School for Advanced Studies (SISSA), Trieste, Italy
| | - Shelly Meron
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Yulia Shenberger
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Zena Qasem
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Lada Gevorkyan-Airapetov
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Alessandra Magistrato
- Department National Research Council of Italy (CNR) - Institute of Material (IOM) c/o International School for Advanced Studies (SISSA), Trieste, Italy.
| | - Sharon Ruthstein
- Department of Chemistry and the Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
8
|
Magrì A, Tabbì G, Naletova I, Attanasio F, Arena G, Rizzarelli E. A Deeper Insight in Metal Binding to the hCtr1 N-terminus Fragment: Affinity, Speciation and Binding Mode of Binuclear Cu 2+ and Mononuclear Ag + Complex Species. Int J Mol Sci 2022; 23:ijms23062929. [PMID: 35328348 PMCID: PMC8953729 DOI: 10.3390/ijms23062929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 01/27/2023] Open
Abstract
Ctr1 regulates copper uptake and its intracellular distribution. The first 14 amino acid sequence of the Ctr1 ectodomain Ctr1(1-14) encompasses the characteristic Amino Terminal Cu2+ and Ni2+ binding motif (ATCUN) as well as the bis-His binding motif (His5 and His6). We report a combined thermodynamic and spectroscopic (UV-vis, CD, EPR) study dealing with the formation of Cu2+ homobinuclear complexes with Ctr1(1-14), the percentage of which is not negligible even in the presence of a small Cu2+ excess and clearly prevails at a M/L ratio of 1.9. Ascorbate fails to reduce Cu2+ when bound to the ATCUN motif, while it reduces Cu2+ when bound to the His5-His6 motif involved in the formation of binuclear species. The histidine diade characterizes the second binding site and is thought to be responsible for ascorbate oxidation. Binding constants and speciation of Ag+ complexes with Ctr1(1-14), which are assumed to mimic Cu+ interaction with N-terminus of Ctr1(1-14), were also determined. A preliminary immunoblot assay evidences that the anti-Ctr1 extracellular antibody recognizes Ctr1(1-14) in a different way from the longer Ctr1(1-25) that encompasses a second His and Met rich domain.
Collapse
Affiliation(s)
- Antonio Magrì
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy; (A.M.); (G.T.); (I.N.)
| | - Giovanni Tabbì
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy; (A.M.); (G.T.); (I.N.)
| | - Irina Naletova
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy; (A.M.); (G.T.); (I.N.)
- Consorzio Interuniversitario per la Ricerca dei Metalli nei Sistemi Biologici, Via Ulpiani 27, 70126 Bari, Italy
| | - Francesco Attanasio
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy; (A.M.); (G.T.); (I.N.)
- Correspondence: (F.A.); (E.R.); Tel.: +39-095-7385070 (E.R.)
| | - Giuseppe Arena
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research, CNR, S.S. Catania, Via P. Gaifami 18, 95126 Catania, Italy; (A.M.); (G.T.); (I.N.)
- Consorzio Interuniversitario per la Ricerca dei Metalli nei Sistemi Biologici, Via Ulpiani 27, 70126 Bari, Italy
- Department of Chemical Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy;
- Correspondence: (F.A.); (E.R.); Tel.: +39-095-7385070 (E.R.)
| |
Collapse
|
9
|
Copper(II) import and reduction are dependent on His-Met clusters in the extracellular amino terminus of human copper transporter-1. J Biol Chem 2022; 298:101631. [PMID: 35090891 PMCID: PMC8867124 DOI: 10.1016/j.jbc.2022.101631] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/22/2022] Open
Abstract
Copper(I) is an essential metal for all life forms. Though Cu(II) is the most abundant and stable state, its reduction to Cu(I) via an unclear mechanism is prerequisite for its bioutilization. In eukaryotes, the copper transporter-1 (CTR1) is the primary high-affinity copper importer, although its mechanism and role in Cu(II) reduction remain uncharacterized. Here we show that extracellular amino-terminus of human CTR1 contains two methionine-histidine clusters and neighboring aspartates that distinctly bind Cu(I) and Cu(II) preceding its import. We determined that hCTR1 localizes at the basolateral membrane of polarized MDCK-II cells and that its endocytosis to Common-Recycling-Endosomes is regulated by reduction of Cu(II) to Cu(I) and subsequent Cu(I) coordination by the methionine cluster. We demonstrate the transient binding of both Cu(II) and Cu(I) during the reduction process is facilitated by aspartates that also act as another crucial determinant of hCTR1 endocytosis. Mutating the first Methionine cluster (7Met-Gly-Met9) and Asp13 abrogated copper uptake and endocytosis upon copper treatment. This phenotype could be reverted by treating the cells with reduced and nonreoxidizable Cu(I). We show that histidine clusters, on other hand, bind Cu(II) and are crucial for hCTR1 functioning at limiting copper. Finally, we show that two N-terminal His-Met-Asp clusters exhibit functional complementarity, as the second cluster is sufficient to preserve copper-induced CTR1 endocytosis upon complete deletion of the first cluster. We propose a novel and detailed mechanism by which the two His-Met-Asp residues of hCTR1 amino-terminus not only bind copper, but also maintain its reduced state, crucial for intracellular uptake.
Collapse
|
10
|
Yu H, Wang H, Qie A, Wang J, Liu Y, Gu G, Yang J, Zhang H, Pan W, Tian Z, Wang C. FGF13 enhances resistance to platinum drugs by regulating hCTR1 and ATP7A via a microtubule-stabilizing effect. Cancer Sci 2021; 112:4655-4668. [PMID: 34533854 PMCID: PMC8586689 DOI: 10.1111/cas.15137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 12/25/2022] Open
Abstract
Platinum‐based regimens are the most widely used chemotherapy regimens, but cancer cells often develop resistance, which impedes therapy outcome for patients. Previous studies have shown that fibroblast growth factor 13 (FGF13) is associated with resistance to platinum drugs in HeLa cells. However, the mechanism and universality of this effect have not been clarified. Here, we found that FGF13 was associated with poor platinum‐based chemotherapy outcomes in a variety of cancers, such as lung, endometrial, and cervical cancers, through bioinformatics analysis. We then found that FGF13 simultaneously regulates the expression and distribution of hCTR1 and ATP7A in cancer cells, causes reduced platinum influx, and promotes platinum sequestration and efflux upon cisplatin exposure. We subsequently observed that FGF13‐mediated platinum resistance requires the microtubule‐stabilizing effect of FGF13. Only overexpression of FGF13 with the ‐SMIYRQQQ‐ tubulin‐binding domain could induce the platinum resistance effect. This phenomenon was also observed in SK‐MES‐1 cells, KLE cells, and 5637 cells. Our research reveals the mechanism of FGF13‐induced platinum drug resistance and suggests that FGF13 can be a sensibilization target and prognostic biomarker for chemotherapy.
Collapse
Affiliation(s)
- Hang Yu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China.,Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China.,College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Handong Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Anran Qie
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China.,Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaqi Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Yang
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hanqiu Zhang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Wensen Pan
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ziqiang Tian
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
11
|
Mandal T, Kar S, Maji S, Sen S, Gupta A. Structural and Functional Diversity Among the Members of CTR, the Membrane Copper Transporter Family. J Membr Biol 2020; 253:459-468. [PMID: 32975619 DOI: 10.1007/s00232-020-00139-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022]
Abstract
Copper is crucial for carrying out normal physiological functions in all higher life forms. Copper Transporter 1 (CTR1) is the high-affinity copper importer found in all eukaryotic organisms. The copper transporter family primarily comprises ~ six members (CTR1-6) and the related members share high sequence homology with CTR. However, with the exception of CTR1, not all six CTRs are present in every organism. Despite having a simple trimeric channel structure, CTR1 and other members exhibit some unique regulatory properties. In the present review, we attempt to understand the diversity and similarity of regulation and functioning of the members of this copper transporter family.
Collapse
Affiliation(s)
- Taniya Mandal
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Sumanta Kar
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Saptarshi Maji
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Samarpita Sen
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Arnab Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.
| |
Collapse
|
12
|
Ghaffari R, Richburg JH. Mice with a Sertoli cell-specific knockout of the Ctr1 gene exhibit a reduced sensitivity to cisplatin-induced testicular germ cell apoptosis. Toxicol Res (Camb) 2019; 8:972-978. [PMID: 32665841 PMCID: PMC7344760 DOI: 10.1039/c9tx00142e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
Exposure to the chemotherapeutic agent cis-diamminedichloroplatinum(ii) (cDDP) is well known to instigate acute and prolonged testicular injury in male patients.
Exposure to the chemotherapeutic agent cis-diamminedichloroplatinum(ii) (cDDP) is well known to instigate acute and prolonged testicular injury in male patients. Many investigators have hypothesized that cDDP-induced dysfunction of Sertoli cells (SCs) may, in part, account for the cDDP-induced lasting testicular injury. Nevertheless, the relative contribution of cDDP-induced SC injury versus direct effects on germ cells (GCs) to the pathogenesis of GC loss remains to be elucidated. The expression of the copper transporter 1 (CTR1) protein in cells directly corresponds with cDDP uptake and its cellular toxicity. Therefore, to discern the role of SCs in the pathogenic mechanism, mice were developed with a SC-specific disruption of the Ctr1 gene (SCΔCtr1) as a strategy to prevent their exposure to cDDP. Adult mice at postnatal day (PND) 60 were treated with 5 mg kg–1 cDDP and then testis collected at 48 hours. A two-fold increase in GC-apoptosis occurred in the testis of cDDP-treated wildtype (WT) mice as compared to saline-treated WT mice. In contrast, cDDP-treated SCΔCtr1 mice exhibited only a half-fold increase in GC-apoptosis as compared to the saline-treated SCΔCtr1 mice. This reduced incidence of GC apoptosis in the SCΔCtr1 mice corresponded to a significantly lower level of platinum within the testis. Taken together, these findings reveal that the uptake of cDDP by CTR1 in SCs accounts for the accumulation of cDDP in the testis and plays a pivotal role in the pathogenic sequence of events leading to the loss of germ cells via apoptosis.
Collapse
Affiliation(s)
- Rashin Ghaffari
- Institute of Cellular and Molecular Biology , College of Natural Sciences , The University of Texas at Austin , Austin , TX , USA .
| | - John H Richburg
- Institute of Cellular and Molecular Biology , College of Natural Sciences , The University of Texas at Austin , Austin , TX , USA . .,The Center for Molecular Carcinogenesis and Toxicology , Division of Pharmacology & Toxicology , College of Pharmacy , The University of Texas at Austin , Austin , TX , USA
| |
Collapse
|
13
|
Zeng X, Baba T, Hamanishi J, Matsumura N, Kharma B, Mise Y, Abiko K, Yamaguchi K, Horikawa N, Hunstman DG, Mulati K, Kitamura S, Taki M, Murakami R, Hosoe Y, Mandai M. Phosphorylation of STAT1 serine 727 enhances platinum resistance in uterine serous carcinoma. Int J Cancer 2019; 145:1635-1647. [PMID: 31228268 DOI: 10.1002/ijc.32501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/12/2019] [Accepted: 05/24/2019] [Indexed: 11/07/2022]
Abstract
Uterine serous carcinoma (USC) is a highly aggressive histological subtype of endometrial cancers harboring highly metastatic and chemoresistant features. Our previous study showed that STAT1 is highly expressed in USC and acts as a key molecule that is positively correlated with tumor progression, but it remains unclear whether STAT1 is relevant to the malicious chemorefractory nature of USC. In the present study, we investigated the regulatory role of STAT1 toward platinum-cytotoxicity in USC. STAT1 suppression sensitized USC cells to increase cisplatin-mediated apoptosis (p < 0.001). Furthermore, phosphorylation of STAT1 was prominently observed on serine-727 (pSTAT1-Ser727), but not on tyrosine-701, in the nucleus of USC cells treated with cisplatin. Mechanistically, the inhibition of pSTAT1-Ser727 by dominant-negative plasmid elevated cisplatin-mediated apoptosis by increasing intracellular accumulation of cisplatin through upregulation of CTR1 expression. TBB has an inhibitory effect on casein kinase 2 (CK2), which phosphorylate STAT1 at serine residues. Sequential treatment with TBB and cisplatin on USC cells greatly reduced nuclear pSTAT1-Ser727, enhanced intracellular accumulation of cisplatin, and subsequently increased apoptosis. Tumor load was significantly reduced by combination therapy of TBB and cisplatin in in vivo xenograft models (p < 0.001). Our results collectively suggest that pSTAT1-Ser727 may play a key role in platinum resistance as well as tumor progression in USC. Thus, targeting the STAT1 pathway via CK2 inhibitor can be a novel method for attenuating the chemorefractory nature of USC.
Collapse
Affiliation(s)
- Xiang Zeng
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Budiman Kharma
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuka Mise
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Horikawa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - David G Hunstman
- Department of Pathology and Laboratory Medicine, University of British Columbia, British Columbia Cancer Agency, Vancouver, BC, Canada.,Genetic Pathology Evaluation Centre, Vancouver General Hospital, Vancouver, BC, Canada
| | - Kumuluzi Mulati
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sachiko Kitamura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuko Hosoe
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Pavlin M, Qasem Z, Sameach H, Gevorkyan-Airapetov L, Ritacco I, Ruthstein S, Magistrato A. Unraveling the Impact of Cysteine-to-Serine Mutations on the Structural and Functional Properties of Cu(I)-Binding Proteins. Int J Mol Sci 2019; 20:E3462. [PMID: 31337158 PMCID: PMC6679193 DOI: 10.3390/ijms20143462] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 02/03/2023] Open
Abstract
Appropriate maintenance of Cu(I) homeostasis is an essential requirement for proper cell function because its misregulation induces the onset of major human diseases and mortality. For this reason, several research efforts have been devoted to dissecting the inner working mechanism of Cu(I)-binding proteins and transporters. A commonly adopted strategy relies on mutations of cysteine residues, for which Cu(I) has an exquisite complementarity, to serines. Nevertheless, in spite of the similarity between these two amino acids, the structural and functional impact of serine mutations on Cu(I)-binding biomolecules remains unclear. Here, we applied various biochemical and biophysical methods, together with all-atom simulations, to investigate the effect of these mutations on the stability, structure, and aggregation propensity of Cu(I)-binding proteins, as well as their interaction with specific partner proteins. Among Cu(I)-binding biomolecules, we focused on the eukaryotic Atox1-ATP7B system, and the prokaryotic CueR metalloregulator. Our results reveal that proteins containing cysteine-to-serine mutations can still bind Cu(I) ions; however, this alters their stability and aggregation propensity. These results contribute to deciphering the critical biological principles underlying the regulatory mechanism of the in-cell Cu(I) concentration, and provide a basis for interpreting future studies that will take advantage of cysteine-to-serine mutations in Cu(I)-binding systems.
Collapse
Affiliation(s)
- Matic Pavlin
- CNR-IOM at SISSA, via Bonomea 265, 34135 Trieste, Italy
| | - Zena Qasem
- Department of Chemistry, Faculty of Exact Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Hila Sameach
- Department of Chemistry, Faculty of Exact Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Lada Gevorkyan-Airapetov
- Department of Chemistry, Faculty of Exact Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel
| | - Ida Ritacco
- CNR-IOM at SISSA, via Bonomea 265, 34135 Trieste, Italy
| | - Sharon Ruthstein
- Department of Chemistry, Faculty of Exact Sciences, Bar Ilan University, Ramat-Gan 5290002, Israel.
| | | |
Collapse
|
15
|
Claus S, Jezierska S, Van Bogaert INA. Protein‐facilitated transport of hydrophobic molecules across the yeast plasma membrane. FEBS Lett 2019; 593:1508-1527. [DOI: 10.1002/1873-3468.13469] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Silke Claus
- Biochemical and Microbial Technology Universiteit Gent Belgium
| | | | - Inge N. A. Van Bogaert
- Lab. of Industrial Microbiology and Biocatalysis Faculty of Bioscience Engineering Ghent University Belgium
| |
Collapse
|
16
|
Magistrato A, Pavlin M, Qasem Z, Ruthstein S. Copper trafficking in eukaryotic systems: current knowledge from experimental and computational efforts. Curr Opin Struct Biol 2019; 58:26-33. [PMID: 31176065 PMCID: PMC6863429 DOI: 10.1016/j.sbi.2019.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/16/2019] [Accepted: 05/02/2019] [Indexed: 01/16/2023]
Abstract
The main copper transporter, Ctr1, can transfer Cu(I) in the cell, through two different intracellular domains. Conformational flexibility of the copper metallochaperone Atox1 controls copper transfer mechanism in the cell. Each metal binding domain in ATP7B has a specific role.
Copper plays a vital role in fundamental cellular functions, and its concentration in the cell must be tightly regulated, as dysfunction of copper homeostasis is linked to severe neurological diseases and cancer. This review provides a compendium of current knowledge regarding the mechanism of copper transfer from the blood system to the Golgi apparatus; this mechanism involves the copper transporter hCtr1, the metallochaperone Atox1, and the ATPases ATP7A/B. We discuss key insights regarding the structural and functional properties of the hCtr1-Atox1-ATP7B cycle, obtained from diverse studies relying on distinct yet complementary biophysical, biochemical, and computational methods. We further address the mechanistic aspects of the cycle that continue to remain elusive. These knowledge gaps must be filled in order to be able to harness our understanding of copper transfer to develop therapeutic approaches with the capacity to modulate copper metabolism.
Collapse
Affiliation(s)
- Alessandra Magistrato
- National Research Council of Italy-IOM c/o International School for Advanced Studies (SISSA), via Bonomea 165, 34135, Trieste, Italy.
| | - Matic Pavlin
- National Research Council of Italy-IOM c/o International School for Advanced Studies (SISSA), via Bonomea 165, 34135, Trieste, Italy
| | - Zena Qasem
- The Chemistry Department, Faculty of Exact Sciences, Bar-Ilan University, 529002, Israel
| | - Sharon Ruthstein
- The Chemistry Department, Faculty of Exact Sciences, Bar-Ilan University, 529002, Israel.
| |
Collapse
|
17
|
Role of epigenetic mechanisms in cisplatin-induced toxicity. Crit Rev Oncol Hematol 2019; 137:131-142. [PMID: 31014509 DOI: 10.1016/j.critrevonc.2019.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/13/2019] [Accepted: 03/13/2019] [Indexed: 12/16/2022] Open
Abstract
Cisplatin (CDDP) is a highly effective antineoplastic agent, widely used in the treatment of various malignant tumors. However, its major problems are side effects associated to toxicity. Considerable inter-individual differences have been reported for CDDP-induced toxicity due to genetic and epigenetic factors. Genetic causes are well described; however, epigenetic modifications are not fully addressed. In the last few years, many evidences were found linking microRNA to the development of CDDP-mediated toxicity, particularly nephrotoxicity. In this review, we described how genetic and epigenetic modifications can be important determinants for the development of toxicity in patients treated with CDDP, and how these alterations may be interesting biomarkers for monitoring toxicity induced by CDDP. Considering the validation in different studies, we suggest that miR-34a, -146b, -378a, -192, and -193 represent an attractive study group to evaluate potential biomarkers to detect CDDP-related nephrotoxicity.
Collapse
|
18
|
Lv X, Song J, Xue K, Li Z, Li M, Zahid D, Cao H, Wang L, Song W, Ma T, Gu J, Li W. Core fucosylation of copper transporter 1 plays a crucial role in cisplatin-resistance of epithelial ovarian cancer by regulating drug uptake. Mol Carcinog 2019; 58:794-807. [PMID: 30614075 DOI: 10.1002/mc.22971] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/13/2018] [Accepted: 12/27/2018] [Indexed: 12/13/2022]
Abstract
Core fucosylation catalyzed by core fucosyltransferase (Fut8) contributes to the progressions of epithelial ovarian cancer (EOC). Copper transporter 1 (CTR1), which contains one N-glycan on Asn15 , mediates cellular transport of cisplatin (cDDP), and plays an important role in the process of cDDP-resistance in EOC. In the present study, we found that the core fucosylation level elevated significantly in the sera of cDDP-treated EOC patients. The in vitro assays also indicate that core fucosylation of CTR1 was significantly upregulated in cDDP-resistant A2780CP cells compared to the cDDP-sensitive A2780S cells. Intriguingly, the hyper core fucosylation suppressed the CTR1-cDDP interactions and cDDP-uptake into A2780CP cells. Conversely, contrast to the Fut8+/+ mouse ovarian epithelial cells, the Fut8-deleted (Fut8-/- ) cells obviously showed higher cDDP-uptake. Furthermore, the recovered core fucosylation induced the suppression of cDDP-uptake in Fut8-restored ovarian epithelial cells. In addition, the core fucosylation could regulate the phosphorylation of cDDP-resistance-associated molecules, such as AKT, ERK, JNK, and mTOR. Our findings suggest that the core fucosylation of CTR1 plays an important role in the cellular cDDP-uptake and thus provide new strategies for improving the outcome of cDDP based chemotherapy of EOC.
Collapse
Affiliation(s)
- Xiaoxue Lv
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Jiazhe Song
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Kai Xue
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Zhi Li
- Clinical Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Ming Li
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Danishi Zahid
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Hongyu Cao
- College of Life Science and Technology, Dalian University, Liaoning, China
| | - Lu Wang
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Wanli Song
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Tonghui Ma
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Jianguo Gu
- Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Wenzhe Li
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| |
Collapse
|
19
|
Yang Y, Zhu Y, Hu H, Cheng L, Liu M, Ma G, Yuan S, Cui P, Liu Y. Cuprous binding promotes interaction of copper transport protein hCTR1 with cell membranes. Chem Commun (Camb) 2019; 55:11107-11110. [DOI: 10.1039/c9cc04859f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cu(i) binding promotes the interaction of hCTR1 with cell membranes, which could initiate the cellular uptake of copper ions.
Collapse
Affiliation(s)
- Yang Yang
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| | - Yang Zhu
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| | - Hongze Hu
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| | - Lanjun Cheng
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| | - Manman Liu
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| | - Guolin Ma
- Institute of Biosciences and Technology
- College of Medicine
- Texas A&M University
- Houston
- USA
| | - Siming Yuan
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| | - Peixin Cui
- Key Laboratory of Soil Environment and Pollution Remediation
- Institute of Soil Science
- the Chinese Academy of Sciences
- Nanjing 210008
- China
| | - Yangzhong Liu
- CAS High Magnetic Field Laboratory
- Department of Chemistry
- University of Science and Technology of China
- Hefei Anhui
- China
| |
Collapse
|
20
|
Wu X, Yuan S, Wang E, Tong Y, Ma G, Wei K, Liu Y. Platinum transfer from hCTR1 to Atox1 is dependent on the type of platinum complex. Metallomics 2018; 9:546-555. [PMID: 28383086 DOI: 10.1039/c6mt00303f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In spite of their wide application, the cellular uptake of platinum based anticancer drugs is still unclear. The copper transport protein, hCTR1, is proposed to facilitate the cellular uptake of cisplatin, whereas organic cation transport (OCT) is more important for oxaliplatin. It has been reported that both N-terminal and C-terminal metal binding motifs of hCTR1 are highly reactive to cisplatin, which is the initial step of protein assisted cellular uptake of cisplatin. It is still unknown how the platinum drugs in hCTR1 transfer to cytoplasmic media, and whether various platinum complexes possess different activities in this process. Herein, we investigated the reaction of the platinated C-terminal metal binding motif of hCTR1 (C8) with the down-stream protein Atox1. Results show that Atox1 is highly reactive to the platinated C8 adducts of cisplatin and transplatin, whereas the oxaliplatin/C8 adduct is much less reactive. The platinum transfer from C8 to Atox1 occurs in the reaction, which results in the protein unfolding of Atox1. These results demonstrated that the platinated intracellular-domain of hCTR1 is reactive to Atox1, and the reactivity is dependent on the ligand and the coordination structure of platinum complexes. The different reactivity is consistent with the hypothesis that hCTR1 is more significant in the transport of cisplatin than that of oxaliplatin.
Collapse
Affiliation(s)
- Xuelei Wu
- CAS Key Laboratory of Soft Matter Chemistry, CAS High Magnetic Field Laboratory, Department of Chemistry, University of Science and Technology of China, Hefei, Anhui 230026, China.
| | | | | | | | | | | | | |
Collapse
|
21
|
Interactions of cisplatin and the copper transporter CTR1 in human colon cancer cells. J Biol Inorg Chem 2017; 22:765-774. [PMID: 28516214 DOI: 10.1007/s00775-017-1467-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023]
Abstract
There is much interest in understanding the mechanisms by which platinum-based anticancer agents enter cells, and the copper transporter CTR1 has been the focus of many recent studies. While there is a clinical correlation between CTR1 levels and platinum efficacy, cellular studies have provided conflicting evidence relating to the relationship between cisplatin and CTR1. We report here our studies of the relationship between cisplatin and copper homeostasis in human colon cancer cells. While the accumulation of copper and platinum do not appear to compete with each other, we did observe that cisplatin perturbs CTR1 distribution within 10 min, a far shorter incubation time than commonly employed in cellular studies of cisplatin. Furthermore, on these short time-scales, cisplatin caused an increase in the cytoplasmic labile copper pool. While the predominant focus of studies to date has been on CTR1, these studies highlight the importance of investigating the interaction of cisplatin with other copper proteins.
Collapse
|
22
|
In vivo effect of copper status on cisplatin-induced nephrotoxicity. Biometals 2016; 29:841-9. [DOI: 10.1007/s10534-016-9955-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/26/2016] [Indexed: 10/21/2022]
|
23
|
Öhrvik H, Logeman B, Turk B, Reinheckel T, Thiele DJ. Cathepsin Protease Controls Copper and Cisplatin Accumulation via Cleavage of the Ctr1 Metal-binding Ectodomain. J Biol Chem 2016; 291:13905-13916. [PMID: 27143361 DOI: 10.1074/jbc.m116.731281] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Indexed: 11/06/2022] Open
Abstract
Copper is an essential metal ion for embryonic development, iron acquisition, cardiac function, neuropeptide biogenesis, and other critical physiological processes. Ctr1 is a high affinity Cu(+) transporter on the plasma membrane and endosomes that exists as a full-length protein and a truncated form of Ctr1 lacking the methionine- and histidine-rich metal-binding ectodomain, and it exhibits reduced Cu(+) transport activity. Here, we identify the cathepsin L/B endolysosomal proteases functioning in a direct and rate-limiting step in the Ctr1 ectodomain cleavage. Cells and mice lacking cathepsin L accumulate full-length Ctr1 and hyper-accumulate copper. As Ctr1 also transports the chemotherapeutic drug cisplatin via direct binding to the ectodomain, we demonstrate that the combination of cisplatin with a cathepsin L/B inhibitor enhances cisplatin uptake and cell killing. These studies identify a new processing event and the key protease that cleaves the Ctr1 metal-binding ectodomain, which functions to regulate cellular Cu(+) and cisplatin acquisition.
Collapse
Affiliation(s)
- Helena Öhrvik
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710.
| | - Brandon Logeman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, SI-1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Medical Faculty, Freiburg 79104 Germany; BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg 79104 Germany
| | - Dennis J Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710; Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina 27710.
| |
Collapse
|
24
|
Miranda JL, Moura LC, San Gil RA, Cruz MT, Silva AC, Barbosa ÁA. Experimental and theoretical studies on the complexes between cisplatin and guanidinoacetic acid. Polyhedron 2015. [DOI: 10.1016/j.poly.2015.09.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
25
|
Dong Z, Wang Y, Wang C, Xu H, Guan L, Li Z, Li F. Self-Assembly of the Second Transmembrane Domain of hCtr1 in Micelles and Interaction with Silver Ion. J Phys Chem B 2015; 119:8302-12. [PMID: 26061257 DOI: 10.1021/acs.jpcb.5b03744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human copper transporter 1 (hCtr1) transports copper and silver by a homotrimer. The protein contains three transmembrane domains in which the second transmembrane domain (TMD2) is a key component lining the central pore of the trimer. The MXXXM motif in the C-terminal end of TMD2 plays a significant role in the function of hCtr1. In this study, we characterized the structure and assembly of isolated TMD2 of hCtr1 in sodium dodecyl sulfate (SDS) micelles and the interaction of the micelle-bound peptide with silver ion using nuclear magnetic resonance, circular dichroism, isothermal titration calorimetry and electrophoresis techniques. We detected the formation of a trimer of the isolated hCtr1-TMD2 in SDS micelles and the binding of the trimer to Ag(I) by a chemical stoichiometry of 3:2 of peptide:Ag(I). We showed that either an intensive pretreatment of the TMD2 peptide by 1,1,1,3,3,3-hexafluoro-2-propanol solvent or a conversion from methionine to leucine in the MXXXM motif changes the aggregation structure of the peptide and decreases the binding affinity by 1 order of magnitude. Our results suggest that the intrinsic interaction of the second transmembrane domain itself may be closely associated with the formation of hCtr1 pore in cellular membranes, and two methionine residues in the MXXXM motif may be important for TMD2 both in the trimeric assembly and in a higher-affinity binding to Ag(I).
Collapse
Affiliation(s)
- Zhe Dong
- †State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Avenue, Changchun 130012, PR China
| | - Yunrui Wang
- †State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Avenue, Changchun 130012, PR China
| | - Chunyu Wang
- †State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Avenue, Changchun 130012, PR China
| | | | - Liping Guan
- †State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Avenue, Changchun 130012, PR China
| | | | - Fei Li
- †State Key Laboratory of Supramolecular Structure and Materials, Jilin University, 2699 Qianjin Avenue, Changchun 130012, PR China
| |
Collapse
|
26
|
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: 3.2] [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
|
27
|
Differential Natural Selection of Human Zinc Transporter Genes between African and Non-African Populations. Sci Rep 2015; 5:9658. [PMID: 25927708 PMCID: PMC5386188 DOI: 10.1038/srep09658] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/13/2015] [Indexed: 12/22/2022] Open
Abstract
Zinc transporters play important roles in all eukaryotes by maintaining the rational zinc concentration in cells. However, the diversity of zinc transporter genes (ZTGs) remains poorly studied. Here, we investigated the genetic diversity of 24 human ZTGs based on the 1000 Genomes data. Some ZTGs show small population differences, such as SLC30A6 with a weighted-average FST (WA-FST = 0.015), while other ZTGs exhibit considerably large population differences, such as SLC30A9 (WA-FST = 0.284). Overall, ZTGs harbor many more highly population-differentiated variants compared with random genes. Intriguingly, we found that SLC30A9 was underlying natural selection in both East Asians (EAS) and Africans (AFR) but in different directions. Notably, a non-synonymous variant (rs1047626) in SLC30A9 is almost fixed with 96.4% A in EAS and 92% G in AFR, respectively. Consequently, there are two different functional haplotypes exhibiting dominant abundance in AFR and EAS, respectively. Furthermore, a strong correlation was observed between the haplotype frequencies of SLC30A9 and distributions of zinc contents in soils or crops. We speculate that the genetic differentiation of ZTGs could directly contribute to population heterogeneity in zinc transporting capabilities and local adaptations of human populations in regard to the local zinc state or diets, which have both evolutionary and medical implications.
Collapse
|
28
|
Rose MC, Kostyanovskaya E, Huang RS. Pharmacogenomics of cisplatin sensitivity in non-small cell lung cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2014; 12:198-209. [PMID: 25449594 PMCID: PMC4411417 DOI: 10.1016/j.gpb.2014.10.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/11/2014] [Accepted: 10/13/2014] [Indexed: 01/13/2023]
Abstract
Cisplatin, a platinum-based chemotherapeutic drug, has been used for over 30 years in a wide variety of cancers with varying degrees of success. In particular, cisplatin has been used to treat late stage non-small cell lung cancer (NSCLC) as the standard of care. However, therapeutic outcomes vary from patient to patient. Considerable efforts have been invested to identify biomarkers that can be used to predict cisplatin sensitivity in NSCLC. Here we reviewed current evidence for cisplatin sensitivity biomarkers in NSCLC. We focused on several key pathways, including nucleotide excision repair, drug transport and metabolism. Both expression and germline DNA variation were evaluated in these key pathways. Current evidence suggests that cisplatin-based treatment could be improved by the use of these biomarkers.
Collapse
Affiliation(s)
- Maimon C Rose
- Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | | | - R Stephanie Huang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
29
|
Tsai CY, Larson CA, Safaei R, Howell SB. Molecular modulation of the copper and cisplatin transport function of CTR1 and its interaction with IRS-4. Biochem Pharmacol 2014; 90:379-87. [PMID: 24967972 DOI: 10.1016/j.bcp.2014.06.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 01/29/2023]
Abstract
The copper influx transporter CTR1 is also a major influx transporter for cisplatin (cDDP) in tumor cells. It influences the cytotoxicity of cDDP both in vivo and in vitro. Whereas Cu triggers internalization of CTR1 from the plasma membrane, cDDP does not. To investigate the mechanisms of these effects, myc-tagged forms of wild type hCTR1 and variants in which Y103 was converted to alanine, C189 was converted to serine, or the K178/K179 dilysine motif was converted to alanines were re-expressed in mouse embryo cells in which both alleles of CTR1 had been knocked out and also in HEK293T cells. The Y103A mutation and to a lesser extent the C189S mutation reduced internalization of CTR1 induced by Cu while the K178A/K179A had little effect. Both Y103 and C189 were required for Cu and cDDP transport whereas the K178/K179 motif was not. While Y103 lies in an YXXM motif that, when phosphorylated, is a potential docking site for phosphatidylinositol 3-kinase and other proteins involved in endocytosis, Western blot analysis of immunoprecipitated myc-CTR1, and proteomic analysis of peptides derived from CTR1, failed to identify any basal or Cu-induced phosphorylation. However, proteomic analysis did identify an interaction of CTR1 with IRS-4 and this was confirmed by co-immunoprecipitation from HEK cells expressing either FLAG-CTR1 or myc-CTR1. The interaction was greater in the Y103A-expressing cells. We conclude that Y103 is required for the internalization of hCTR1 in response to Cu, that this occurs by a mechanism other than phosphorylation and that mutation of Y103 modulates the interaction with IRS-4.
Collapse
Affiliation(s)
- Cheng-Yu Tsai
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, Mail Code 0819, La Jolla, CA 92093-0819, USA.
| | - Christopher A Larson
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, Mail Code 0819, La Jolla, CA 92093-0819, USA.
| | - Roohangiz Safaei
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, Mail Code 0819, La Jolla, CA 92093-0819, USA
| | - Stephen B Howell
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, Mail Code 0819, La Jolla, CA 92093-0819, USA; Department of Medicine, University of California, San Diego, 3855 Health Sciences Drive, Mail Code 0819, La Jolla, CA 92093-0819, USA.
| |
Collapse
|
30
|
Quail JF, Tsai CY, Howell SB. Characterization of a monoclonal antibody capable of reliably quantifying expression of human Copper Transporter 1 (hCTR1). J Trace Elem Med Biol 2014; 28:151-159. [PMID: 24447817 PMCID: PMC3989404 DOI: 10.1016/j.jtemb.2013.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 11/16/2013] [Accepted: 12/10/2013] [Indexed: 12/20/2022]
Abstract
Human copper transporter 1 (hCTR1) is the high-affinity copper influx transporter in mammalian cells that also mediates the influx of cisplatin. Loss of hCTR1 expression has been implicated in the development of resistance to this cancer chemotherapeutic agent. It has turned out to be very difficult to develop antibodies to hCTR1 and polyclonal antibodies produced by different laboratories have yielded conflicting results. We have characterized a newly-available rabbit monoclonal antibody that reacts with an epitope on the N-terminal end of hCTR1 that now permits rigorous identification and quantification of hCTR1 using Western blot analysis. Postnuclear membrane (PNM) preparations made from cells engineered to express high levels of myc-tagged hCTR1, and cells in which the expression of hCTR1 was knocked down, were used to characterize the antibody. The identity of the bands detected was confirmed by immunoprecipitation, surface biotinylation and deglycosylation of myc-tagged hCTR1. Despite the specificity expected of a monoclonal antibody, the anti-hCTR1 detected a variety of bands in whole cell lysates (WCL), which made it difficult to quantify hCTR1. This problem was overcome by isolating post-nuclear membranes and using these for further analysis. Three bands were identified using this antibody in PNM preparations that migrated at 28, 33-35 and 62-64kDa. Multiple lines of evidence presented here suggest that the 33-35 and 62-64kDa bands are hCTR1 whereas the 28kDa band is a cross-reacting protein of unknown identify. The 33-35kDa band is consistent with the expected MW of the glycosylated hCTR1 monomer. This analysis now permits rigorous identification and quantification of hCTR1.
Collapse
Affiliation(s)
- Jacob F Quail
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, United States
| | - Cheng-Yu Tsai
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, United States
| | - Stephen B Howell
- Moores UCSD Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093, United States.
| |
Collapse
|
31
|
Lin X, Shang X, Manorek G, Fofana M, Stephen B H. Integrin αV modulates the cellular pharmacology of copper and cisplatin by regulating expression of the influx transporter CTR1. Oncoscience 2014; 1:185-195. [PMID: 25594011 PMCID: PMC4278295 DOI: 10.18632/oncoscience.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 03/23/2014] [Indexed: 01/07/2023] Open
Abstract
The αV integrin is expressed in most cancer cells where it regulates a diverse array of cellular functions essential to the initiation, progression and metastasis of solid tumors. However, little is known about how αV integrin modulates cellular sensitivity to chemotherapeutic agents, particularly the platinum drugs. In this study, we found that down-regulation of αV sensitized human M21 cells to cisplatin (cDDP) through up-regulation of the copper influx transporter CTR1. Cells selected for low αV integrin expression (M21L) were more sensitive to cDDP, accompanied by increase in CTR1 mRNA and CTR1 protein levels, more intracellular cDDP accumulation and cDDP DNA adduct formation. Basal copper (Cu) content, Cu uptake, and Cu cytotoxicity were also increased. Transfection of a luciferase reporter construct containing the hCTR1 promoter sequence revealed an increase of the hCTR1 transcription activity in M21L cells. The basis for the increased hCTR1 transcription was related to an increase in the steady-state level of Sp1, a transcription factor known to drive hCTR1 expression. These results indicate that the αV integrin modulates sensitivity of human cells to the cytotoxic effect of cDDP by regulating expression of the Cu transporter CTR1, and introduce the concept that αV expression is linked to Cu homeostasis.
Collapse
Affiliation(s)
- Xinjian Lin
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Xiying Shang
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Gerald Manorek
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Mariama Fofana
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Howell Stephen B
- Department of Medicine and UC San Diego Moores Cancer Center, University of California, San Diego, La Jolla, CA
| |
Collapse
|
32
|
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: 4.6] [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
|
33
|
Ctr2 regulates biogenesis of a cleaved form of mammalian Ctr1 metal transporter lacking the copper- and cisplatin-binding ecto-domain. Proc Natl Acad Sci U S A 2013; 110:E4279-88. [PMID: 24167251 DOI: 10.1073/pnas.1311749110] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Copper is an essential catalytic cofactor for enzymatic activities that drive a range of metabolic biochemistry including mitochondrial electron transport, iron mobilization, and peptide hormone maturation. Copper dysregulation is associated with fatal infantile disease, liver, and cardiac dysfunction, neuropathy, and anemia. Here we report that mammals regulate systemic copper acquisition and intracellular mobilization via cleavage of the copper-binding ecto-domain of the copper transporter 1 (Ctr1). Although full-length Ctr1 is critical to drive efficient copper import across the plasma membrane, cleavage of the ecto-domain is required for Ctr1 to mobilize endosomal copper stores. The biogenesis of the truncated form of Ctr1 requires the structurally related, previously enigmatic copper transporter 2 (Ctr2). Ctr2(-/-) mice are defective in accumulation of truncated Ctr1 and exhibit increased tissue copper levels, and X-ray fluorescence microscopy demonstrates that copper accumulates as intracellular foci. These studies identify a key regulatory mechanism for mammalian copper transport through Ctr2-dependent accumulation of a Ctr1 variant lacking the copper- and cisplatin-binding ecto-domain.
Collapse
|
34
|
Du X, Li H, Wang X, Liu Q, Ni J, Sun H. Kinetics and thermodynamics of metal binding to the N-terminus of a human copper transporter, hCTR1. Chem Commun (Camb) 2013; 49:9134-6. [PMID: 23962988 DOI: 10.1039/c3cc45360j] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The N-terminus of hCTR1 was demonstrated to bind three Cu(+) ions tightly (log K = 14.92) and reversibly via its Met-rich motifs. Ag(+) binds to the protein with the same stoichiometry but much lower affinities than Cu(+). The protein also coordinates two Cu(2+) ions through its ATCUN motif and His-rich motif with lower affinity. This study provides an insight into the selectivity of the transporter.
Collapse
Affiliation(s)
- Xiubo Du
- Department of Chemistry, The University of Hong Kong, Hong Kong, China.
| | | | | | | | | | | |
Collapse
|
35
|
Ivy KD, Kaplan JH. A re-evaluation of the role of hCTR1, the human high-affinity copper transporter, in platinum-drug entry into human cells. Mol Pharmacol 2013; 83:1237-46. [PMID: 23543413 PMCID: PMC3657103 DOI: 10.1124/mol.113.085068] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/29/2013] [Indexed: 01/11/2023] Open
Abstract
Cisplatin (cDDP) is an anticancer drug used in a number of malignancies, including testicular, ovarian, cervical, bladder, lung, head, and neck cancers. Its use is limited by the development of resistance, often rationalized via effects on cellular uptake. It has been claimed that human copper transporter 1 (hCTR1), the human high-affinity copper transporter, is the major entry pathway for cDDP and related drugs via a mechanism that mimics copper. This is an unexpected property of hCTR1, a highly selective copper (I) transporter. We compared the uptake rates of copper with cDDP (and several analogs) into human embryonic kidney 293 cells overexpressing wild-type or mutant hCTR1, mouse embryonic fibroblasts that do or do not express CTR1, and human ovarian tumor cells that are sensitive or resistant to cDDP. We have also compared the effects of extracellular copper, which causes regulatory endocytosis of hCTR1, to those of cDDP. We confirm the correlation between higher hCTR1 levels and higher platinum drug uptake in tumor cells sensitive to the drug. However, we show that hCTR1 is not the major entry route of platinum drugs, and that the copper transporter is not internalized in response to extracellular drug. Our data suggest the major entry pathway for platinum drugs is not saturable at relevant concentrations and not protein-mediated. Clinical trials have been initiated that depend upon regulating membrane levels of hCTR1. If reduced drug uptake is a major factor in resistance, hCTR1 is unlikely to be a productive target in attempts to enhance efficacy, although the proteins involved in copper homeostasis may play a role.
Collapse
Affiliation(s)
- Kristin D Ivy
- Department of Biochemistry & Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60607, USA
| | | |
Collapse
|
36
|
Wang E, Xi Z, Li Y, Li L, Zhao L, Ma G, Liu Y. Interaction between Platinum Complexes and the C-Terminal Motif of Human Copper Transporter 1. Inorg Chem 2013; 52:6153-9. [DOI: 10.1021/ic400495w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Erqiong Wang
- CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry
and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, China
| | - Zhaoyong Xi
- CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry
and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, China
| | - Yan Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China
| | - Lianzhi Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China
| | - Linhong Zhao
- CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry
and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, China
| | - Guolin Ma
- CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry
and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, China
| | - Yangzhong Liu
- CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry
and Materials Science, University of Science and Technology of China, Hefei Anhui 230026, China
| |
Collapse
|
37
|
Wee NK, Weinstein DC, Fraser ST, Assinder SJ. The mammalian copper transporters CTR1 and CTR2 and their roles in development and disease. Int J Biochem Cell Biol 2013; 45:960-3. [DOI: 10.1016/j.biocel.2013.01.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 01/14/2013] [Accepted: 01/25/2013] [Indexed: 12/26/2022]
|
38
|
Nguyen TH, Arnesano F, Scintilla S, Rossetti G, Ippoliti E, Carloni P, Natile G. Structural Determinants of Cisplatin and Transplatin Binding to the Met-Rich Motif of Ctr1: A Computational Spectroscopy Approach. J Chem Theory Comput 2012; 8:2912-20. [DOI: 10.1021/ct300167m] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Trung Hai Nguyen
- Computational Biophysics, German Research School for Simulation Sciences, D-52425 Jülich, Germany,
and Institute for Advanced Simulation, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Fabio Arnesano
- Department Farmaco-Chimico, University of Bari “A. Moro”, via Edoardo
Orabona 4, 70125 Bari, Italy
| | - Simone Scintilla
- Department Farmaco-Chimico, University of Bari “A. Moro”, via Edoardo
Orabona 4, 70125 Bari, Italy
| | - Giulia Rossetti
- Computational Biophysics, German Research School for Simulation Sciences, D-52425 Jülich, Germany,
and Institute for Advanced Simulation, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Emiliano Ippoliti
- Computational Biophysics, German Research School for Simulation Sciences, D-52425 Jülich, Germany,
and Institute for Advanced Simulation, Forschungszentrum Jülich, D-52425 Jülich, Germany
| | - Paolo Carloni
- Computational Biophysics, German Research School for Simulation Sciences, D-52425 Jülich, Germany,
and Institute for Advanced Simulation, Forschungszentrum Jülich, D-52425 Jülich, Germany
- Statistical and Biological Physics Sector, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265,
I-34136 Trieste, Italy
| | - Giovanni Natile
- Department Farmaco-Chimico, University of Bari “A. Moro”, via Edoardo
Orabona 4, 70125 Bari, Italy
| |
Collapse
|
39
|
Skvortsov AN, Zatulovskiy EA, Puchkova LV. Structure-functional organization of eukaryotic high-affinity copper importer CTR1 determines its ability to transport copper, silver, and cisplatin. Mol Biol 2012. [DOI: 10.1134/s0026893312010219] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Wang X, Li H, Du X, Harris J, Guo Z, Sun H. Activation of carboplatin and nedaplatin by the N-terminus of human copper transporter 1 (hCTR1). Chem Sci 2012. [DOI: 10.1039/c2sc20738a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
41
|
Du X, Wang X, Li H, Sun H. Comparison between copper and cisplatin transport mediated by human copper transporter 1 (hCTR1). Metallomics 2012; 4:679-85. [DOI: 10.1039/c2mt20021j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
42
|
Kommuguri UN, Bodiga S, Sankuru S, Bodiga VL. Copper deprivation modulates CTR1 and CUP1 expression and enhances cisplatin cytotoxicity in Saccharomyces cerevisiae. J Trace Elem Med Biol 2012; 26:13-9. [PMID: 22365074 DOI: 10.1016/j.jtemb.2011.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 10/19/2011] [Accepted: 12/04/2011] [Indexed: 01/19/2023]
Abstract
Saccharomyces cerevisiae has been established as a model system for cancer studies, due to the widely conserved family of genes involved in cell cycle progression, proliferation and apoptosis. In the current study, we sought to determine whether copper deprivation modulates sensitivity of yeast to cisplatin. Yeast cultures grown in low copper medium and exposed to bathocuproiene disulfate (BCS) resulted in significant reduction of intracellular copper. We report here that low copper medium rendered BY4741 hypersensitive to cisplatin (CDDP). Yeast grown in low copper medium exhibited ∼2.0 fold enhanced cytotoxicity in survival and colony-forming ability, compared to copper adequate control cells grown in YPD. The effect of copper restriction on CDDP sensitivity appeared to be associated with the up regulation of CTR1, facilitating enhanced uptake and accumulation of CDDP. Also, CDDP further lowered copper deprivation-induced changes in CUP1 metallothionein levels, SOD activity and GSH levels. These changes were associated with increased protein oxidation and lipid peroxidation induced by CDDP. These results thus suggest that cisplatin cytotoxicity is potentiated under low copper conditions due to enhanced uptake and accumulation of cisplatin and also in part due to lowered antioxidant defense and increased oxidative stress imposed by copper deprivation.
Collapse
|
43
|
Li Q, Peng X, Yang H, Rodriguez JA, Shu Y. Contribution of organic cation transporter 3 to cisplatin cytotoxicity in human cervical cancer cells. J Pharm Sci 2011; 101:394-404. [PMID: 21905038 DOI: 10.1002/jps.22752] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/18/2011] [Accepted: 08/16/2011] [Indexed: 01/11/2023]
Abstract
This study was conducted to investigate whether drug transporters play a role in determination of cisplatin resistance in cervical cancer cells. The transcript levels of the transporter genes previously associated with cisplatin transport and/or resistance were compared between the cisplatin-sensitive cervical adenocarcinoma KB-3-1 and its derivative cisplatin-resistant KB-CP20 cells. The expression of the efflux transporter gene multidrug resistance-associated protein 2 (MRP2) was significantly reduced in KB-CP20 cells, in support of previous studies indicating that MRP2 is unlikely responsible for cisplatin resistance in these cells. We observed that the expression of the uptake transporter organic cation transporter 3 (OCT3) was extremely downregulated in KB-CP20 compared with KB-3-1 cells. Consistently, the transport function for organic cations in the former was considerably low. OCT3 overexpression significantly increased cisplatin cellular accumulation and cytotoxicity in KB-3-1 cells, while its downregulation by short hairpin RNA or chemical inhibition increased the resistance. Interestingly, there was no effect of OCT3 overexpression on cisplatin accumulation and cytotoxicity in human embryonic kidney 293 cells. The present study indicates that OCT3 partially contributes to the sensitivity of cervical adenocarcinoma cells to cisplatin cytotoxicity. Further studies are required to determine OCT3 activity in cervical cancer tissues of different cisplatin chemoresponses and to elucidate the underlying mechanisms of different OCT3 function in different cell types.
Collapse
Affiliation(s)
- Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | | | |
Collapse
|
44
|
Haas KL, Putterman AB, White DR, Thiele DJ, Franz KJ. Model peptides provide new insights into the role of histidine residues as potential ligands in human cellular copper acquisition via Ctr1. J Am Chem Soc 2011; 133:4427-37. [PMID: 21375246 PMCID: PMC3247019 DOI: 10.1021/ja108890c] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cellular acquisition of copper in eukaryotes is primarily accomplished through the Ctr family of copper transport proteins. In both humans and yeast, methionine-rich "Mets" motifs in the amino-terminal extracellular domain of Ctr1 are thought to be responsible for recruitment of copper at the cell surface. Unlike yeast, mammalian Ctr1 also contains extracellular histidine-rich motifs, although a role for these regions in copper uptake has not been explored in detail. Herein, synthetic model peptides containing the first 14 residues of the extracellular domain of human Ctr1 (MDHSHHMGMSYMDS) have been prepared and evaluated for their apparent binding affinity to both Cu(I) and Cu(II). These studies reveal a high affinity Cu(II) binding site (log K = 11.0 ± 0.3 at pH 7.4) at the amino-terminus of the peptide as well as a high affinity Cu(I) site (log K = 10.2 ± 0.2 at pH 7.4) that utilizes adjacent HH residues along with an additional His or Met ligand. These model studies suggest that the histidine domains may play a direct role in copper acquisition from serum copper-binding proteins and in facilitating the reduction of Cu(II) to the active Ctr1 substrate, Cu(I). We tested this hypothesis by expressing a Ctr1 mutant lacking only extracellular histidine residues in Ctr1-knockout mouse embryonic fibroblasts. Results from live cell studies support the hypothesis that extracellular amino-terminal His residues directly participate in the copper transport function of Ctr1.
Collapse
Affiliation(s)
- Kathryn L. Haas
- Department of Chemistry, Duke University, Durham, North Carolina 27708
| | | | - Daniel R. White
- Department of Chemistry, Duke University, Durham, North Carolina 27708
| | - Dennis J. Thiele
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27708
| | | |
Collapse
|
45
|
Wang X, Du X, Li H, Chan DSB, Sun H. The effect of the extracellular domain of human copper transporter (hCTR1) on cisplatin activation. Angew Chem Int Ed Engl 2011; 50:2706-11. [PMID: 21387471 DOI: 10.1002/anie.201006739] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 01/19/2011] [Indexed: 01/05/2023]
Affiliation(s)
- Xinghao Wang
- Department of Chemistry, The University of Hong Kong, PR China
| | | | | | | | | |
Collapse
|
46
|
Wang X, Du X, Li H, Chan DS, Sun H. The Effect of the Extracellular Domain of Human Copper Transporter (hCTR1) on Cisplatin Activation. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201006739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Xinghao Wang
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong (P.R. China), Fax: (+852) 2857‐1586
| | - Xiubo Du
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong (P.R. China), Fax: (+852) 2857‐1586
| | - Hongyan Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong (P.R. China), Fax: (+852) 2857‐1586
| | - Denise So‐Bik Chan
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong (P.R. China), Fax: (+852) 2857‐1586
| | - Hongzhe Sun
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong (P.R. China), Fax: (+852) 2857‐1586
| |
Collapse
|
47
|
Regulation of Cisplatin cytotoxicity by cu influx transporters. Met Based Drugs 2011; 2010:317581. [PMID: 21274436 PMCID: PMC3025362 DOI: 10.1155/2010/317581] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 12/07/2010] [Indexed: 12/30/2022] Open
Abstract
Platinum drugs are an important class of cancer chemotherapeutics. However, the use of these drugs is limited by the development of resistance during treatment with decreased accumulation being a common mechanism. Both Cu transporters CTR1 and CTR2 influence the uptake and cytotoxicity of cisplatin. Although it is structurally similar to CTR1, CTR2 functions in a manner opposite to that of CTR1 with respect to Pt drug uptake. Whereas knockout of CTR1 reduces Pt drug uptake, knockdown of CTR2 enhances cisplatin uptake and cytotoxicity. CTR2 is subject to transcriptional and posttranscriptional regulation by both Cu and cisplatin; this regulation is partly dependent on the Cu chaperone ATOX1. Insight into the mechanisms by which CTR1 and CTR2 regulate sensitivity to the Pt-containing drugs has served as the basis for novel pharmacologic strategies for improving their efficacy.
Collapse
|