1
|
Ye F, Li Q, Huang L, Liao N. Reliable high-PAP-1-loaded polymeric micelles for cancer therapy: preparation, characterization, and evaluation of anti-tumor efficacy. Drug Deliv 2025; 32:2490269. [PMID: 40207975 PMCID: PMC11986873 DOI: 10.1080/10717544.2025.2490269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
The mitochondrial potassium channel Kv1.3 is a critical therapeutic target, as its blockade induces cancer cell apoptosis, highlighting its therapeutic potential. PAP-1, a potent and selective membrane-permeant Kv1.3 inhibitor, faces solubility challenges affecting its bioavailability and antitumor efficacy. To circumvent these challenges, we developed a tumor-targeting drug delivery system by encapsulating PAP-1 within pH-responsive mPEG-PAE polymeric micelles. These self-assembled micelles exhibited high entrapment efficiency (91.35%) and drug loading level (8.30%). As pH decreased, the micelles exhibited a significant increase in particle size and zeta potential, accompanied by a surge in PAP-1 release. Molecular simulations revealed that PAE's tertiary amine protonation affected the self-assembly process, modifying hydrophobicity and resulting in larger, loosely packed particles. Furthermore, compared to free PAP-1 or PAP-1 combined with MDR inhibitors, PAP-1-loaded micelles significantly enhanced cytotoxicity and apoptosis induction in Jurkat and B16F10 cells, through mechanisms involving decreased mitochondrial membrane potential and elevated caspase-3 activity. In vivo, while free PAP-1 failed to reduce tumor size in a B16F10 melanoma mouse model, PAP-1-loaded micelles substantially suppressed tumors, reducing volume by up to 94.26%. Fluorescent-marked micelles effectively accumulated in mouse tumors, confirming their targeting efficiency. This strategy holds promise for significantly improving PAP-1's antitumor efficacy in tumor therapy.
Collapse
Affiliation(s)
- Fang Ye
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Qi Li
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, P. R. China
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Longping Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, P. R. China
- Clinical Laboratory, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Naikai Liao
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, P. R. China
| |
Collapse
|
2
|
Fotakopoulos G, Gatos C, Georgakopoulou VE, Christodoulidis G, Kagkouras I, Trakas N, Foroglou N. Exploring the Role of Sigma Receptors in the Treatment of Cancer: A Narrative Review. Cureus 2024; 16:e70946. [PMID: 39502961 PMCID: PMC11537387 DOI: 10.7759/cureus.70946] [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] [Accepted: 10/06/2024] [Indexed: 11/08/2024] Open
Abstract
This study investigated the association of sigma receptors (SRs) and their selective ligands (because the molecular characteristics of the same SRs, particularly sigma-2 receptor {S2R}, are not completely clear) in carcinogenesis, their potential use as antitumor agents, and their great utility in tumor imaging. The ion channels and transporters enhance the cell's ability to adapt to the metabolic conditions encountered in the tumor tissue. The high expression of SRs in the proliferating cells compared with those at rest indicates that this is a significant clinical biomarker for determining the proliferative status of solid tumors using functional PET imaging techniques. The association of SRs in the pathophysiology of cancer cells is a result of the high concentration of S1R and S2R binding sites observed in various tumor cell lines and tissues. It would also be remarkable to determine if SRs are involved in metastasis and other metastatic cell behaviors such as adhesion, secretion, motility, and penetration. An absolute challenge for research in this field is to develop an integrated model that describes the molecular mechanisms of sigma receptors, incorporating their known biological and pathophysiological roles.
Collapse
Affiliation(s)
| | - Charalabos Gatos
- Neurosurgery, General University Hospital of Larissa, Larissa, GRC
| | | | | | | | | | - Nikolaos Foroglou
- Neurosurgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, GRC
| |
Collapse
|
3
|
Alberghini-Dos-Santos JV, Sanchez CA, Bordon KDCF, Pucca MB, Antunes LMG, Arantes EC, Oliveira ISD. Effects of crotamine in human prostate cancer cell line. Toxicon 2024; 243:107746. [PMID: 38704124 DOI: 10.1016/j.toxicon.2024.107746] [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: 02/14/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
Our study presents the anticancer potential of crotamine from Crotalus durissus terrificus in human prostate cancer cell line DU-145. Crotamine isolation was conducted through RP-FPLC, its molecular mass analyzed by MALDI-TOF was 4881.4 kDa, and N-terminal sequencing confirmed crotamine identity. Crotamine demonstrated no toxicity and did not inhibit migration in HUVEC cells. Although no cell death occurred in DU-145 cells, crotamine inhibited their migration. Thus, crotamine presented potential to be a prototype of anticancer drug.
Collapse
Affiliation(s)
- João Victor Alberghini-Dos-Santos
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Caroline Andolfato Sanchez
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Karla de Castro Figueiredo Bordon
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Manuela Berto Pucca
- Health and Sciences Postgraduate Program, Federal University of Roraima, Boa Vista, RR, Brazil; Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Isadora Sousa de Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
4
|
Dupuy M, Gueguinou M, Potier-Cartereau M, Lézot F, Papin M, Chantôme A, Rédini F, Vandier C, Verrecchia F. SK Ca- and Kv1-type potassium channels and cancer: Promising therapeutic targets? Biochem Pharmacol 2023; 216:115774. [PMID: 37678626 DOI: 10.1016/j.bcp.2023.115774] [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: 06/28/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023]
Abstract
Ion channels are transmembrane structures that allow the passage of ions across cell membranes such as the plasma membrane or the membranes of various organelles like the nucleus, endoplasmic reticulum, Golgi apparatus or mitochondria. Aberrant expression of various ion channels has been demonstrated in several tumor cells, leading to the promotion of key functions in tumor development, such as cell proliferation, resistance to apoptosis, angiogenesis, invasion and metastasis. The link between ion channels and these key biological functions that promote tumor development has led to the classification of cancers as oncochannelopathies. Among all ion channels, the most varied and numerous, forming the largest family, are the potassium channels, with over 70 genes encoding them in humans. In this context, this review will provide a non-exhaustive overview of the role of plasma membrane potassium channels in cancer, describing 1) the nomenclature and structure of potassium channels, 2) the role of these channels in the control of biological functions that promotes tumor development such as proliferation, migration and cell death, and 3) the role of two particular classes of potassium channels, the SKCa- and Kv1- type potassium channels in cancer progression.
Collapse
Affiliation(s)
- Maryne Dupuy
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| | | | | | - Frédéric Lézot
- Sorbonne University, INSERM UMR933, Hôpital Trousseau (AP-HP), Paris F-75012, France
| | - Marion Papin
- N2C UMR 1069, University of Tours, INSERM, Tours, France
| | | | - Françoise Rédini
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France
| | | | - Franck Verrecchia
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000 Nantes, France.
| |
Collapse
|
5
|
Sesti F, Bortolami A, Kathera-Ibarra EF. Non-conducting functions of potassium channels in cancer and neurological disease. CURRENT TOPICS IN MEMBRANES 2023; 92:199-231. [PMID: 38007268 DOI: 10.1016/bs.ctm.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Cancer and neurodegenerative disease, albeit fundamental differences, share some common pathogenic mechanisms. Accordingly, both conditions are associated with aberrant cell proliferation and migration. Here, we review the causative role played by potassium (K+) channels, a fundamental class of proteins, in cancer and neurodegenerative disease. The concept that emerges from the review of the literature is that K+ channels can promote the development and progression of cancerous and neurodegenerative pathologies by dysregulating cell proliferation and migration. K+ channels appear to control these cellular functions in ways that not necessarily depend on their conducting properties and that involve the ability to directly or indirectly engage growth and survival signaling pathways. As cancer and neurodegenerative disease represent global health concerns, identifying commonalities may help understand the molecular basis for those devastating conditions and may facilitate the design of new drugs or the repurposing of existing drugs.
Collapse
Affiliation(s)
- Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States.
| | - Alessandro Bortolami
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States
| | - Elena Forzisi Kathera-Ibarra
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States
| |
Collapse
|
6
|
Angi B, Muccioli S, Szabò I, Leanza L. A Meta-Analysis Study to Infer Voltage-Gated K+ Channels Prognostic Value in Different Cancer Types. Antioxidants (Basel) 2023; 12:antiox12030573. [PMID: 36978819 PMCID: PMC10045123 DOI: 10.3390/antiox12030573] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 03/02/2023] Open
Abstract
Potassium channels are often highly expressed in cancer cells with respect to healthy ones, as they provide proliferative advantages through modulating membrane potential, calcium homeostasis, and various signaling pathways. Among potassium channels, Shaker type voltage-gated Kv channels are emerging as promising pharmacological targets in oncology. Here, we queried publicly available cancer patient databases to highlight if a correlation exists between Kv channel expression and survival rate in five different cancer types. By multiple gene comparison analysis, we found a predominant expression of KCNA2, KCNA3, and KCNA5 with respect to the other KCNA genes in skin cutaneous melanoma (SKCM), uterine corpus endometrial carcinoma (UCEC), stomach adenocarcinoma (STAD), lung adenocarcinoma (LUAD), and lung squamous cell carcinoma (LUSC). This analysis highlighted a prognostic role of KCNA3 and KCNA5 in SKCM, LUAD, LUSC, and STAD, respectively. Interestingly, KCNA3 was associated with a positive prognosis in SKCM and LUAD but not in LUSC. Results obtained by the analysis of KCNA3-related differentially expressed genes (DEGs); tumor immune cell infiltration highlighted differences that may account for such differential prognosis. A meta-analysis study was conducted to investigate the role of KCNA channels in cancer using cancer patients’ datasets. Our study underlines a promising correlation between Kv channel expression in tumor cells, in infiltrating immune cells, and survival rate.
Collapse
|
7
|
Fraser SP, Tesi A, Bonito B, Ka Ming Hui M, Arcangeli A, Djamgoz MB. Potassium Channel Blockage and Invasiveness of Strongly Metastatic Prostate and Breast Cancer Cells. Bioelectricity 2021. [DOI: 10.1089/bioe.2020.0041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Scott P. Fraser
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom
| | - Alessandra Tesi
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Benedetta Bonito
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Marcus Ka Ming Hui
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mustafa B.A. Djamgoz
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, London, United Kingdom
- Biotechnology Research Centre, Cyprus International University, Haspolat, Nicosia, North Cyprus, Turkey
| |
Collapse
|
8
|
Capatina AL, Lagos D, Brackenbury WJ. Targeting Ion Channels for Cancer Treatment: Current Progress and Future Challenges. Rev Physiol Biochem Pharmacol 2020; 183:1-43. [PMID: 32865696 DOI: 10.1007/112_2020_46] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ion channels are key regulators of cancer cell pathophysiology. They contribute to a variety of processes such as maintenance of cellular osmolarity and membrane potential, motility (via interactions with the cytoskeleton), invasion, signal transduction, transcriptional activity and cell cycle progression, leading to tumour progression and metastasis. Ion channels thus represent promising targets for cancer therapy. Ion channels are attractive targets because many of them are expressed at the plasma membrane and a broad range of existing inhibitors are already in clinical use for other indications. However, many of the ion channels identified in cancer cells are also active in healthy normal cells, so there is a risk that certain blockers may have off-target effects on normal physiological function. This review describes recent research advances into ion channel inhibitors as anticancer therapeutics. A growing body of evidence suggests that a range of existing and novel Na+, K+, Ca2+ and Cl- channel inhibitors may be effective for suppressing cancer cell proliferation, migration and invasion, as well as enhancing apoptosis, leading to suppression of tumour growth and metastasis, either alone or in combination with standard-of-care therapies. The majority of evidence to date is based on preclinical in vitro and in vivo studies, although there are several examples of ion channel-targeting strategies now reaching early phase clinical trials. Given the strong links between ion channel function and regulation of tumour growth, metastasis and chemotherapy resistance, it is likely that further work in this area will facilitate the development of new therapeutic approaches which will reach the clinic in the future.
Collapse
Affiliation(s)
| | - Dimitris Lagos
- Hull York Medical School, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - William J Brackenbury
- Department of Biology, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
9
|
Ion Channel Profiling in Prostate Cancer: Toward Cell Population-Specific Screening. Rev Physiol Biochem Pharmacol 2020; 181:39-56. [PMID: 32737754 DOI: 10.1007/112_2020_22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the last three decades, a growing number of studies have implicated ion channels in all essential processes of prostate carcinogenesis, including cell proliferation, apoptosis, migration, and angiogenesis. The changes in the expression of individual ion channels show a specific profile, making these proteins promising clinical biomarkers that may enable better molecular subtyping of the disease and lead to more rapid and accurate clinical decision-making. Expression profiles and channel function are mainly based on the tumoral tissue itself, in this case, the epithelial cancer cell population. To date, little data on the ion channel profile of the cancerous prostate stroma are available, even though tumor interactions with the microenvironment are crucial in carcinogenesis and each distinct population plays a specific role in tumor progression. In this review, we describe ion channel expression profiles specific for the distinct cell population of the tumor microenvironment (stromal, endothelial, neuronal, and neuroendocrine cell populations) and the technical approaches used for efficient separation and screening of these cell populations.
Collapse
|
10
|
George K, Thomas NS, Malathi R. 4,4'-Diisothiocyanatostilbene-2,2'-disulfonate modulates voltage-gated K + current and influences cell cycle arrest in androgen sensitive and insensitive human prostate cancer cell lines. Toxicol Mech Methods 2020; 30:358-369. [PMID: 32193973 DOI: 10.1080/15376516.2020.1745343] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The stilbene derivative, 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid (DIDS), an anion channel blocker is used in the present study to evaluate its modulatory effect on voltage-gated K+ current (IK) in human prostate cancer cell lines (LNCaP and PC-3). Voltage-gated K+ (KV) channels in the plasma membrane are critically involved in the proliferation of tumor cells. Therefore, KV channels are considered as a novel potential target for cancer treatment. The results of the present study show that the external perfusion of DIDS activates IK in a concentration-dependent manner, although the known K+ channel blocker TEA failed to block the DIDS activated IK in PC-3 cells. Whereas, in LNCaP cells, the higher concentration of DIDS blocked IK, though this effect was not completely recovered after washout. The difference in function of DIDS might be due to the expression of different Kv channel isoforms in LNCaP and PC-3 cells. Further, the anticancer studies show that treatment of DIDS significantly induced G2/M phase cell cycle arrest and induced moderate and low level of cell death in LNCaP and PC-3 cells respectively. This finding reveals that DIDS modulates IK and exerts cell cycle arrest and cell death in LNCaP and PC-3 cells.
Collapse
Affiliation(s)
- Kiran George
- Department of Biomedical Engineering, Chennai Institute of Technology, Chennai, India.,Bio Engineering Lab, Department of Electronics and Instrumentation Engineering, Annamalai University, Annamalai Nagar, India
| | - Nisha Susan Thomas
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, India
| | - Raman Malathi
- Bio Engineering Lab, Department of Electronics and Instrumentation Engineering, Annamalai University, Annamalai Nagar, India
| |
Collapse
|
11
|
Teisseyre A, Palko-Labuz A, Sroda-Pomianek K, Michalak K. Voltage-Gated Potassium Channel Kv1.3 as a Target in Therapy of Cancer. Front Oncol 2019; 9:933. [PMID: 31612103 PMCID: PMC6769076 DOI: 10.3389/fonc.2019.00933] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 09/06/2019] [Indexed: 12/11/2022] Open
Abstract
Voltage-gated potassium channel Kv1.3 is an integral membrane protein, which is selectively permeable for potassium ions and is activated upon a change of membrane potential. Channel activation enables transportation of potassium ions down their electrochemical gradient. Kv1.3 channel is expressed in many cell types, both normal and cancer. Activity of the channel plays an important role in cell proliferation and apoptosis. Inhibition of Kv1.3 channel may be beneficial in therapy of several diseases including some cancer disorders. This review focuses on Kv1.3 channel as a new potentially attractive molecular target in cancer therapy. In the first part, changes in the channel expression in selected cancer disorders are described. Then, the role of the channel activity in cancer cell proliferation and apoptosis is presented. Finally, it is shown that some low molecular weight organic inhibitors of the channel including selected biologically active plant-derived polycyclic compounds may selectively induce apoptosis of Kv1.3-expressing cancer cells while sparing normal cells and healthy organs. These compounds may be promising candidates for putative application in therapy of some cancer disorders, such as melanoma, pancreatic ductal adenocarcinoma (PDAC), or B-type chronic lymphocytic leukemia (B-CLL).
Collapse
Affiliation(s)
- Andrzej Teisseyre
- Department of Biophysics, Wroclaw Medical University, Wrocław, Poland
| | - Anna Palko-Labuz
- Department of Biophysics, Wroclaw Medical University, Wrocław, Poland
| | | | - Krystyna Michalak
- Department of Biophysics, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
12
|
Abstract
As the leading cause of death in cancer, there is an urgent need to develop treatments to target the dissemination of primary tumor cells to secondary organs, known as metastasis. Bioelectric signaling has emerged in the last century as an important controller of cell growth, and with the development of current molecular tools we are now beginning to identify its role in driving cell migration and metastasis in a variety of cancer types. This review summarizes the currently available research for bioelectric signaling in solid tumor metastasis. We review the steps of metastasis and discuss how these can be controlled by bioelectric cues at the level of a cell, a population of cells, and the tissue. The role of ion channel, pump, and exchanger activity and ion flux is discussed, along with the importance of the membrane potential and the relationship between ion flux and membrane potential. We also provide an overview of the evidence for control of metastasis by external electric fields (EFs) and draw from examples in embryogenesis and regeneration to discuss the implications for endogenous EFs. By increasing our understanding of the dynamic properties of bioelectric signaling, we can develop new strategies that target metastasis to be translated into the clinic.
Collapse
Affiliation(s)
- Samantha L. Payne
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
13
|
Ion Channel Targeting with Antibodies and Antibody Fragments for Cancer Diagnosis. Antibodies (Basel) 2019; 8:antib8020033. [PMID: 31544839 PMCID: PMC6640718 DOI: 10.3390/antib8020033] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022] Open
Abstract
The antibody era has greatly impacted cancer management in recent decades. Indeed, antibodies are currently applied for both cancer diagnosis and therapy. For example, monoclonal antibodies are the main constituents of several in vitro diagnostics, which are applied at many levels of cancer diagnosis. Moreover, the great improvement provided by in vivo imaging, especially for early-stage cancer diagnosis, has traced the path for the development of a complete new class of antibodies, i.e., engineered antibody fragments. The latter embody the optimal characteristics (e.g., low renal retention, rapid clearance, and small size) which make them ideal for in vivo applications. Furthermore, the present review focuses on reviewing the main applications of antibodies and antibody fragments for solid cancer diagnosis, both in vitro and in vivo. Furthermore, we review the scientific evidence showing that ion channels represent an almost unexplored class of ideal targets for both in vitro and in vivo diagnostic purposes. In particular, we review the applications, in solid cancers, of monoclonal antibodies and engineered antibody fragments targeting the voltage-dependent ion channel Kv 11.1, also known as hERG1.
Collapse
|
14
|
Guzel RM, Ogmen K, Ilieva KM, Fraser SP, Djamgoz MBA. Colorectal cancer invasiveness in vitro: Predominant contribution of neonatal Nav1.5 under normoxia and hypoxia. J Cell Physiol 2018; 234:6582-6593. [PMID: 30341901 DOI: 10.1002/jcp.27399] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/17/2018] [Indexed: 12/28/2022]
Abstract
Functional expression of voltage-gated Na+ channels (VGSCs) occurs in human carcinomas and promotes invasiveness in vitro and metastasis in vivo. Both neonatal and adult forms of Nav1.5 (nNav1.5 and aNav1.5, respectively) have been reported to be expressed at messenger RNA (mRNA) level in colorectal cancer (CRCa) cells. Here, three CRCa cell lines (HT29, HCT116 and SW620) were studied and found to express nNav1.5 mRNA and protein. In SW620 cells, adopted as a model, effects of gene silencing (by several small interfering RNAs [siRNAs]) selectively targeting nNav1.5 or aNav1.5 were determined on (a) channel activity and (b) invasiveness in vitro. Silencing nNav1.5 made the currents more "adult-like" and suppressed invasion by up to 73%. Importantly, subsequent application of the highly specific, general VGSC blocker, tetrodotoxin (TTX), had no further effect. Conversely, silencing aNav1.5 made the currents more "neonatal-like" but suppressed invasion by only 17% and TTX still induced a significant effect. Hypoxia increased invasiveness and this was also blocked completely by siRNA targeting nNav1.5. The effect of hypoxia was suppressed dose dependently by ranolazine, but its effect was lost in cells pretreated with nNav1.5-siRNA. We conclude that (a) functional nNav1.5 expression is common to human CRCa cells, (b) hypoxia increases the invasiveness of SW620 cells, (c) the VGSC-dependent invasiveness is driven predominantly by nNav1.5 under both normoxic and hypoxic conditions and (d) the hypoxia-induced increase in invasiveness is likely to be mediated by the persistent current component of nNav1.5.
Collapse
Affiliation(s)
- R Mine Guzel
- Department of Life Sciences, Imperial College London, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London, UK
| | - Kazim Ogmen
- Department of Life Sciences, Imperial College London, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London, UK
| | - Kristina M Ilieva
- Department of Life Sciences, Imperial College London, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London, UK
| | - Scott P Fraser
- Department of Life Sciences, Imperial College London, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London, UK
| | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London, UK.,Biotechnology Research Centre, Cyprus International University, Haspolat, Mersin 10, North Cyprus
| |
Collapse
|
15
|
Zhang Y, Feng Y, Chen L, Zhu J. Effects of Intermediate-Conductance Ca(2+)-Activated K(+) Channels on Human Endometrial Carcinoma Cells. Cell Biochem Biophys 2017; 72:515-25. [PMID: 25608633 DOI: 10.1007/s12013-014-0497-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The objective of this study was to investigate the effect of intermediate-conductance Ca(2+)-activated K(+) (KCa3.1) channels on the cell proliferation, cell cycle, apoptosis, migration, and invasion in endometrial cancer (EC) cells. Human EC cell lines HEC-1-A and Ishikawa were cultured in vitro and transfected with recombinant plasmid containing KCa3.1-targeting shRNA. RT-qPCR and Western blot were used to examine the mRNA and protein expression levels of KCa3.1 channels in transfected cells. In addition, the specific inhibitor of KCa3.1, TRAM-34, was used to examine the effect of KCa3.1 blockage on migration capacity and invasiveness of EC cells using transwell assay. Proliferation and apoptotic rates of EC cells transfected with KCa3.1 shRNA or treated with TRAM-34 were analyzed using MTT, BrdU incorporation assay, and flow cytometry. Expression of cell cycle proteins and metalloproteinase-2 (MMP-2) was evaluated by RT-qPCR and Western blotting. TRAM-34 treatment and KCa3.1 silencing using shRNA dramatically suppressed both the mRNA and protein expression of KCa3.1 channels (P < 0.01) compared with control groups. Blockage of KCa3.1 by TRAM-34 treatment and KCa3.1 shRNA transfection exerted inhibitory effect on cell growth of both EC cell lines, as demonstrated by increased cell population at G0-G1 phase and decreased cell population at S phase. However, both the treatments did not result in significant changes in the apoptotic rate (P > 0.05) compared to controls. Protein expressions of cyclin D1, cyclin E, and survivin were significantly decreased in the experimental groups comparing to control. We showed that TRAM-34 treatment led to significantly inhibited migration, invasion, and MMP-2 expression in HEC-1-A and Ishikawa cells, compared with the control group (P < 0.01). Blockage of KCa3.1 channel activity or expression inhibits cell proliferation and cell cycle progression without inducing apoptosis in EC cells. Moreover, TRAM-34 could reduce the ability of EC cells to migrate and invade, which might be related to reduced expression of MMP-2.
Collapse
Affiliation(s)
- Yingli Zhang
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022, China
| | - Youji Feng
- Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, 200080, China
| | - Lu Chen
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022, China.
| | - Jianqing Zhu
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, 310022, China
| |
Collapse
|
16
|
Shahabi A, Lewinger JP, Ren J, April C, Sherrod AE, Hacia JG, Daneshmand S, Gill I, Pinski JK, Fan JB, Stern MC. Novel Gene Expression Signature Predictive of Clinical Recurrence After Radical Prostatectomy in Early Stage Prostate Cancer Patients. Prostate 2016; 76:1239-56. [PMID: 27272349 PMCID: PMC9015679 DOI: 10.1002/pros.23211] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/16/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Current clinical tools have limited accuracy in differentiating patients with localized prostate cancer who are at risk of recurrence from patients with indolent disease. We aimed to identify a gene expression signature that jointly with clinical variables could improve upon the prediction of clinical recurrence after RP for patients with stage T2 PCa. METHODS The study population includes consented patients who underwent a radical retropubic prostatectomy (RP) and bilateral pelvic lymph node dissection at the University of Southern California in the PSA-era (1988-2008). We used a nested case-control study of 187 organ-confined patients (pT2N0M0): 154 with no recurrence ("controls") and 33 with clinical recurrence ("cases"). RNA was obtained from laser capture microdissected malignant glands representative of the overall Gleason score of each patient. Whole genome gene expression profiles (29,000 transcripts) were obtained using the Whole Genome DASL HT platform (Illumina, Inc). A gene expression signature of PCa clinical recurrence was identified using stability selection with elastic net regularized logistic regression. Three existing datasets generated with the Affymetrix Human Exon 1.0ST array were used for validation: Mayo Clinic (MC, n = 545), Memorial Sloan Kettering Cancer Center (SKCC, n = 150), and Erasmus Medical Center (EMC, n = 48). The areas under the ROC curve (AUCs) were obtained using repeated fivefold cross-validation. RESULTS A 28-gene expression signature was identified that jointly with key clinical variables (age, Gleason score, pre-operative PSA level, and operation year) was predictive of clinical recurrence (AUC of clinical variables only was 0.67, AUC of clinical variables, and 28-gene signature was 0.99). The AUC of this gene signature fitted in each of the external datasets jointly with clinical variables was 0.75 (0.72-0.77) (MC), 0.90 (0.86-0.94) (MSKCC), and 0.82 (0.74-0.91) (EMC), whereas the AUC for clinical variables only in each dataset was 0.72 (0.70-0.74), 0.86 (0.82-0.91), and 0.76 (0.67-0.85), respectively. CONCLUSIONS We report a novel gene-expression based classifier identified using agnostic approaches from whole genome expression profiles that can improve upon the accuracy of clinical indicators to stratify early stage localized patients at risk of clinical recurrence after RP. Prostate 76:1239-1256, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ahva Shahabi
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Juan Pablo Lewinger
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California
| | - Jie Ren
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California
| | | | - Andy E. Sherrod
- Department of Pathology, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
| | - Joseph G. Hacia
- Department of Biochemistry and Molecular Biology, Keck School of Medicine of USC, Los Angeles, California
| | - Siamak Daneshmand
- Department of Urology and USC Institute of Urology, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
| | - Inderbir Gill
- Department of Urology and USC Institute of Urology, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
| | - Jacek K. Pinski
- Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
| | - Jian-Bing Fan
- Illumina, Inc., San Diego, California
- AnchorDx Corporation, Guangzhou, China
| | - Mariana C. Stern
- Department of Preventive Medicine, Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California
- Department of Urology and USC Institute of Urology, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, California
- Correspondence to: Dr. Mariana C. Stern, University of Southern California Keck School of Medicine, Norris Comprehensive Cancer Center, 1441 Eastlake Avenue, Room 5421A, Los Angeles, CA 90089.
| |
Collapse
|
17
|
Fraser SP, Hemsley F, Djamgoz MBA. Caffeic acid phenethyl ester: Inhibition of metastatic cell behaviours via voltage-gated sodium channel in human breast cancer in vitro. Int J Biochem Cell Biol 2015; 71:111-118. [PMID: 26724521 DOI: 10.1016/j.biocel.2015.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/09/2015] [Accepted: 12/21/2015] [Indexed: 11/28/2022]
Abstract
Caffeic acid phenethyl ester, derived from natural propolis, has been reported to have anti-cancer properties. Voltage-gated sodium channels are upregulated in many cancers where they promote metastatic cell behaviours, including invasiveness. We found that micromolar concentrations of caffeic acid phenethyl ester blocked voltage-gated sodium channel activity in several invasive cell lines from different cancers, including breast (MDA-MB-231 and MDA-MB-468), colon (SW620) and non-small cell lung cancer (H460). In the MDA-MB-231 cell line, which was adopted as a 'model', long-term (48 h) treatment with 18 μM caffeic acid phenethyl ester reduced the peak current density by 91% and shifted steady-state inactivation to more hyperpolarized potentials and slowed recovery from inactivation. The effects of long-term treatment were also dose-dependent, 1 μM caffeic acid phenethyl ester reducing current density by only 65%. The effects of caffeic acid phenethyl ester on metastatic cell behaviours were tested on the MDA-MB-231 cell line at a working concentration (1 μM) that did not affect proliferative activity. Lateral motility and Matrigel invasion were reduced by up to 14% and 51%, respectively. Co-treatment of caffeic acid phenethyl ester with tetrodotoxin suggested that the voltage-gated sodium channel inhibition played a significant intermediary role in these effects. We conclude, first, that caffeic acid phenethyl ester does possess anti-metastatic properties. Second, the voltage-gated sodium channels, commonly expressed in strongly metastatic cancers, are a novel target for caffeic acid phenethyl ester. Third, more generally, ion channel inhibition can be a significant mode of action of nutraceutical compounds.
Collapse
Affiliation(s)
- Scott P Fraser
- Imperial College London, Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London SW7 2AZ, UK.
| | - Faye Hemsley
- Imperial College London, Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London SW7 2AZ, UK
| | - Mustafa B A Djamgoz
- Imperial College London, Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, South Kensington Campus, London SW7 2AZ, UK; Biotechnology Research Centre (BRC), Cyprus International University, Haspolat, Lefkosa, North Cyprus, Mersin 10, Turkey
| |
Collapse
|
18
|
Abstract
This review compares the biological and physiological function of Sigma receptors [σRs] and their potential therapeutic roles. Sigma receptors are widespread in the central nervous system and across multiple peripheral tissues. σRs consist of sigma receptor one (σ1R) and sigma receptor two (σ2R) and are expressed in numerous regions of the brain. The sigma receptor was originally proposed as a subtype of opioid receptors and was suggested to contribute to the delusions and psychoses induced by benzomorphans such as SKF-10047 and pentazocine. Later studies confirmed that σRs are non-opioid receptors (not an µ opioid receptor) and play a more diverse role in intracellular signaling, apoptosis and metabolic regulation. σ1Rs are intracellular receptors acting as chaperone proteins that modulate Ca2+ signaling through the IP3 receptor. They dynamically translocate inside cells, hence are transmembrane proteins. The σ1R receptor, at the mitochondrial-associated endoplasmic reticulum membrane, is responsible for mitochondrial metabolic regulation and promotes mitochondrial energy depletion and apoptosis. Studies have demonstrated that they play a role as a modulator of ion channels (K+ channels; N-methyl-d-aspartate receptors [NMDAR]; inositol 1,3,5 triphosphate receptors) and regulate lipid transport and metabolism, neuritogenesis, cellular differentiation and myelination in the brain. σ1R modulation of Ca2+ release, modulation of cardiac myocyte contractility and may have links to G-proteins. It has been proposed that σ1Rs are intracellular signal transduction amplifiers. This review of the literature examines the mechanism of action of the σRs, their interaction with neurotransmitters, pharmacology, location and adverse effects mediated through them.
Collapse
Affiliation(s)
- Colin G Rousseaux
- a Department of Pathology and Laboratory Medicine , University of Ottawa , Ottawa , ON , Canada and
| | | |
Collapse
|
19
|
Involvement of potassium channels in the progression of cancer to a more malignant phenotype. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2477-92. [PMID: 25517985 DOI: 10.1016/j.bbamem.2014.12.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 12/01/2014] [Accepted: 12/08/2014] [Indexed: 12/22/2022]
Abstract
Potassium channels are a diverse group of pore-forming transmembrane proteins that selectively facilitate potassium flow through an electrochemical gradient. They participate in the control of the membrane potential and cell excitability in addition to different cell functions such as cell volume regulation, proliferation, cell migration, angiogenesis as well as apoptosis. Because these physiological processes are essential for the correct cell function, K+ channels have been associated with a growing number of diseases including cancer. In fact, different K+ channel families such as the voltage-gated K+ channels, the ether à-go-go K+ channels, the two pore domain K+ channels and the Ca2+-activated K+ channels have been associated to tumor biology. Potassium channels have a role in neoplastic cell-cycle progression and their expression has been found abnormal in many types of tumors and cancer cells. In addition, the expression and activity of specific K+ channels have shown a significant correlation with the tumor malignancy grade. The aim of this overview is to summarize published data on K+ channels that exhibit oncogenic properties and have been linked to a more malignant cancer phenotype. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.
Collapse
|
20
|
Ru Q, Tian X, Pi MS, Chen L, Yue K, Xiong Q, Ma BM, Li CY. Voltage‑gated K+ channel blocker quinidine inhibits proliferation and induces apoptosis by regulating expression of microRNAs in human glioma U87‑MG cells. Int J Oncol 2014; 46:833-40. [PMID: 25420507 DOI: 10.3892/ijo.2014.2777] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/10/2014] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence has proved that potassium channels (K+ channels) are involved in regulating cell proliferation, cell cycle progression and apoptosis of tumor cells. However, the precise cellular mechanisms are still unknown. In the present study, we investigated the effect and mechanisms of quinidine, a commonly used voltage-gated K+ channel blocker, on cell proliferation and apoptosis of human glioma U87-MG cells. We found that quinidine significantly inhibited the proliferation of U87-MG cells and induced apoptosis in a dose-dependent manner. The results of caspase colorimetric assay showed that the mitochondrial pathway was the main mode involved in the quinidine-induced apoptotic process. Furthermore, the concentration range of quinidine, which inhibited voltage-gated K+ channel currents in electrophysiological assay, was consistent with that of quinidine inhibiting cell proliferation and inducing cell apoptosis. In U87-MG cells treated with quinidine (100 µmol/l), 11 of 2,042 human microRNAs (miRNAs) were upregulated and 16 were downregulated as detected with the miRNA array analysis. The upregulation of miR-149-3p and downregulation of miR-424-5p by quinidine treatment were further verified by using quantitative real-time PCR. In addition, using miRNA target prediction program, putative target genes related to cell proliferation and apoptosis for two differentially expressed miRNAs were predicted. Taken together, these data suggested that the anti-proliferative and pro-apoptosis effect of voltage-gated K+ channel blocker quinidine in human glioma cells was mediated at least partly through regulating expression of miRNAs, and provided further support for the mechanisms of voltage-gated K+ channels in mediating cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Qin Ru
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Xiang Tian
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Ming-Shan Pi
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Lin Chen
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Kai Yue
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Qi Xiong
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Bao-Miao Ma
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| | - Chao-Ying Li
- Wuhan Institutes of Biomedical Sciences, Jianghan University, Wuhan, Hubei 430056, P.R. China
| |
Collapse
|
21
|
Fraser SP, Peters A, Fleming-Jones S, Mukhey D, Djamgoz MBA. Resveratrol: Inhibitory Effects on Metastatic Cell Behaviors and Voltage-Gated Na+Channel Activity in Rat Prostate Cancer In Vitro. Nutr Cancer 2014; 66:1047-58. [DOI: 10.1080/01635581.2014.939291] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
22
|
Pier DM, Shehatou GSG, Giblett S, Pullar CE, Trezise DJ, Pritchard CA, Challiss RAJ, Mitcheson JS. Long-term channel block is required to inhibit cellular transformation by human ether-à-go-go-related gene (hERG1) potassium channels. Mol Pharmacol 2014; 86:211-21. [PMID: 24830940 PMCID: PMC4127929 DOI: 10.1124/mol.113.091439] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 05/14/2014] [Indexed: 11/22/2022] Open
Abstract
Both human ether-à-go-go-related gene (hERG1) and the closely related human ether-à-go-go (hEAG1) channel are aberrantly expressed in a large proportion of human cancers. In the present study, we demonstrate that transfection of hERG1 into mouse fibroblasts is sufficient to induce many features characteristic of malignant transformation. An important finding of this work is that this transformation could be reversed by chronic incubation (for 2-3 weeks) with the hERG channel blocker dofetilide (100 nM), whereas more acute applications (for 1-2 days) were ineffective. The hERG1 expression resulted in a profound loss of cell contact inhibition, multiple layers of overgrowing cells, and high saturation densities. Cells also changed from fibroblast-like to a more spindle-shaped morphology, which was associated with a smaller cell size, a dramatic increase in cell polarization, a reduction in the number of actin stress fibers, and less punctate labeling of focal adhesions. Analysis of single-cell migration and scratch-wound closure clearly demonstrated that hERG1-expressing cells migrated more rapidly than vector-transfected control cells. In contrast to previous studies on hEAG1, there were no increases in rates of proliferation, or loss of growth factor dependency; however, hERG1-expressing cells were capable of substrate-independent growth. Allogeneic transplantation of hERG1-expressing cells into nude mice resulted in an increased incidence of tumors. In contrast to hEAG1, the mechanism of cellular transformation is dependent on ion conduction. Trafficking-deficient and conduction-deficient hERG1 mutants also prevented cellular transformation. These results provide evidence that hERG1 expression is sufficient to induce cellular transformation by a mechanism distinct from hEAG1. The most important conclusion of this study is that selective hERG1 channel blockers have therapeutic potential in the treatment of hERG1-expressing cancers.
Collapse
Affiliation(s)
- David M Pier
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - George S G Shehatou
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Susan Giblett
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Christine E Pullar
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Derek J Trezise
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - Catrin A Pritchard
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - R A John Challiss
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| | - John S Mitcheson
- Department of Cell Physiology and Pharmacology (D.M.P., G.S.G.S., C.E.P., R.A.J.C., J.S.M.) and Department of Biochemistry (S.G., C.A.P.), University of Leicester, Leicester, United Kingdom; Molecular Discovery Research, GlaxoSmithKline R&D, Harlow, Essex, United Kingdom (D.J.T.); Department of Pharmacology and Toxicology, University of Mansoura, Egypt (G.S.G.S.); Essen Bioscience Ltd., Welwyn Garden City, UK (D.J.T.); School of Clinical Sciences, University of Edinburgh, United Kingdom (D.M.P.)
| |
Collapse
|
23
|
Comes N, Bielanska J, Vallejo-Gracia A, Serrano-Albarrás A, Marruecos L, Gómez D, Soler C, Condom E, Ramón Y Cajal S, Hernández-Losa J, Ferreres JC, Felipe A. The voltage-dependent K(+) channels Kv1.3 and Kv1.5 in human cancer. Front Physiol 2013; 4:283. [PMID: 24133455 PMCID: PMC3794381 DOI: 10.3389/fphys.2013.00283] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 09/18/2013] [Indexed: 11/20/2022] Open
Abstract
Voltage-dependent K+ channels (Kv) are involved in a number of physiological processes, including immunomodulation, cell volume regulation, apoptosis as well as differentiation. Some Kv channels participate in the proliferation and migration of normal and tumor cells, contributing to metastasis. Altered expression of Kv1.3 and Kv1.5 channels has been found in several types of tumors and cancer cells. In general, while the expression of Kv1.3 apparently exhibits no clear pattern, Kv1.5 is induced in many of the analyzed metastatic tissues. Interestingly, evidence indicates that Kv1.5 channel shows inversed correlation with malignancy in some gliomas and non-Hodgkin's lymphomas. However, Kv1.3 and Kv1.5 are similarly remodeled in some cancers. For instance, expression of Kv1.3 and Kv1.5 correlates with a certain grade of tumorigenicity in muscle sarcomas. Differential remodeling of Kv1.3 and Kv1.5 expression in human cancers may indicate their role in tumor growth and their importance as potential tumor markers. However, despite of this increasing body of information, which considers Kv1.3 and Kv1.5 as emerging tumoral markers, further research must be performed to reach any conclusion. In this review, we summarize what it has been lately documented about Kv1.3 and Kv1.5 channels in human cancer.
Collapse
Affiliation(s)
- Núria Comes
- Molecular Physiology Laboratory, Departament de Bioquímica i Biologia Molecular, Institut de Biomedicina, Universitat de Barcelona Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Frede J, Fraser SP, Oskay-Özcelik G, Hong Y, Ioana Braicu E, Sehouli J, Gabra H, Djamgoz MB. Ovarian cancer: Ion channel and aquaporin expression as novel targets of clinical potential. Eur J Cancer 2013; 49:2331-44. [DOI: 10.1016/j.ejca.2013.03.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 01/29/2013] [Accepted: 03/10/2013] [Indexed: 01/11/2023]
|
25
|
The inhibitor of Ca(2+)-dependent K+ channels TRAM-34 blocks growth of hepatocellular carcinoma cells via downregulation of estrogen receptor alpha mRNA and nuclear factor-kappaB. Invest New Drugs 2012; 31:452-7. [PMID: 23054207 DOI: 10.1007/s10637-012-9879-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 09/16/2012] [Indexed: 01/03/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common liver malignancy still demanding for novel therapeutic options. Since the ion channel inhibitor TRAM-34 (1-[(2-chlorophenyl) diphenylmethyl]-1H-pyrazole) was shown to block growth in various cancer cells, we investigated anti-tumor effects of TRAM-34 in human HCC cell lines. We found that TRAM-34 reduced HCC cell proliferation without induction of apoptosis. This was due to a decreased mRNA expression of estrogen receptor alpha (ESR1) and a reduced activation of NF-kappaB, which both are implicated in the development of HCC. Therefore, TRAM-34 might represent a novel therapeutic target for the treatment of HCC.
Collapse
|
26
|
Huang X, Dubuc AM, Hashizume R, Berg J, He Y, Wang J, Chiang C, Cooper MK, Northcott PA, Taylor MD, Barnes MJ, Tihan T, Chen J, Hackett CS, Weiss WA, James CD, Rowitch DH, Shuman MA, Jan YN, Jan LY. Voltage-gated potassium channel EAG2 controls mitotic entry and tumor growth in medulloblastoma via regulating cell volume dynamics. Genes Dev 2012; 26:1780-96. [PMID: 22855790 DOI: 10.1101/gad.193789.112] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.
Collapse
Affiliation(s)
- Xi Huang
- Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Ion channels and G-protein-coupled receptors (GPCRs) play a fundamental role in cancer progression by influencing Ca(2+) influx and signaling pathways in transformed cells. Transformed cells thrive in a hostile environment that is characterized by extracellular acidosis that promotes the pathological phenotype. The pathway(s) by which extracellular protons achieve this remain unclear. Here, a role for proton-sensing ion channels and GPCRs as mediators of the effects of extracellular protons in cancer cells is discussed.
Collapse
Affiliation(s)
- Maike Glitsch
- Department of Physiology, Anatomy and Genetics, Oxford University, Oxford, United Kingdom.
| |
Collapse
|
28
|
Abstract
Ion channels are involved in a variety of tumors. In particular, potassium channels are expressed abnormally in many cancer types, where their pharmacologic manipulation impairs tumor progression. Since this group of molecules has been successfully targeted for decades in other therapeutic areas, there is a significant body of knowledge on the pharmacology of potassium channels. Several groups of potassium channels with defined molecular identities have been proposed as candidates for therapeutic intervention. The strategies put forward range from classical small molecule blockade to gene therapy approaches, and include the use of potassium channels as targets for adjuvant therapy. We will discuss the reasons for these proposals and explore possible future developments.
Collapse
|
29
|
Enyedi P, Czirják G. Molecular background of leak K+ currents: two-pore domain potassium channels. Physiol Rev 2010; 90:559-605. [PMID: 20393194 DOI: 10.1152/physrev.00029.2009] [Citation(s) in RCA: 670] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Two-pore domain K(+) (K(2P)) channels give rise to leak (also called background) K(+) currents. The well-known role of background K(+) currents is to stabilize the negative resting membrane potential and counterbalance depolarization. However, it has become apparent in the past decade (during the detailed examination of the cloned and corresponding native K(2P) channel types) that this primary hyperpolarizing action is not performed passively. The K(2P) channels are regulated by a wide variety of voltage-independent factors. Basic physicochemical parameters (e.g., pH, temperature, membrane stretch) and also several intracellular signaling pathways substantially and specifically modulate the different members of the six K(2P) channel subfamilies (TWIK, TREK, TASK, TALK, THIK, and TRESK). The deep implication in diverse physiological processes, the circumscribed expression pattern of the different channels, and the interesting pharmacological profile brought the K(2P) channel family into the spotlight. In this review, we focus on the physiological roles of K(2P) channels in the most extensively investigated cell types, with special emphasis on the molecular mechanisms of channel regulation.
Collapse
Affiliation(s)
- Péter Enyedi
- Department of Physiology, Semmelweis University, Budapest, Hungary.
| | | |
Collapse
|
30
|
Ralph SJ, Rodríguez-Enríquez S, Neuzil J, Saavedra E, Moreno-Sánchez R. The causes of cancer revisited: "mitochondrial malignancy" and ROS-induced oncogenic transformation - why mitochondria are targets for cancer therapy. Mol Aspects Med 2010; 31:145-70. [PMID: 20206201 DOI: 10.1016/j.mam.2010.02.008] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 02/19/2010] [Indexed: 12/17/2022]
Abstract
The role of oncoproteins and tumor suppressor proteins in promoting the malignant transformation of mammalian cells by affecting properties such as proliferative signalling, cell cycle regulation and altered adhesion is well established. Chemicals, viruses and radiation are also generally accepted as agents that commonly induce mutations in the genes encoding these cancer-causing proteins, thereby giving rise to cancer. However, more recent evidence indicates the importance of two additional key factors imposed on proliferating cells that are involved in transformation to malignancy and these are hypoxia and/or stressful conditions of nutrient deprivation (e.g. lack of glucose). These two additional triggers can initiate and promote the process of malignant transformation when a low percentage of cells overcome and escape cellular senescence. It is becoming apparent that hypoxia causes the progressive elevation in mitochondrial ROS production (chronic ROS) which over time leads to stabilization of cells via increased HIF-2alpha expression, enabling cells to survive with sustained levels of elevated ROS. In cells under hypoxia and/or low glucose, DNA mismatch repair processes are repressed by HIF-2alpha and they continually accumulate mitochondrial ROS-induced oxidative DNA damage and increasing numbers of mutations driving the malignant transformation process. Recent evidence also indicates that the resulting mutated cancer-causing proteins feedback to amplify the process by directly affecting mitochondrial function in combinatorial ways that intersect to play a major role in promoting a vicious spiral of malignant cell transformation. Consequently, many malignant processes involve periods of increased mitochondrial ROS production when a few cells survive the more common process of oxidative damage induced cell senescence and death. The few cells escaping elimination emerge with oncogenic mutations and survive to become immortalized tumors. This review focuses on evidence highlighting the role of mitochondria as drivers of elevated ROS production during malignant transformation and hence, their potential as targets for cancer therapy. The review is organized into five main sections concerning different aspects of "mitochondrial malignancy". The first concerns the functions of mitochondrial ROS and its importance as a pacesetter for cell growth versus senescence and death. The second considers the available evidence that cellular stress in the form of hypoxic and/or hypoglycaemic conditions represent two of the major triggering events for cancer and how oncoproteins reinforce this process by altering gene expression to bring about a common set of changes in mitochondrial function and activity in cancer cells. The third section presents evidence that oncoproteins and tumor suppressor proteins physically localize to the mitochondria in cancer cells where they directly regulate malignant mitochondrial programs, including apoptosis. The fourth section covers common mutational changes in the mitochondrial genome as they relate to malignancy and the relationship to the other three areas. The last section concerns the relevance of these findings, their importance and significance for novel targeted approaches to anti-cancer therapy and selective triggering in cancer cells of the mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Stephen J Ralph
- Genomic Research Centre, Griffith Institute of Health and Medical Research, School of Medical Science, Griffith University, Parklands Avenue, Southport, 4222 Qld, Australia.
| | | | | | | | | |
Collapse
|
31
|
|
32
|
Millar ID, Wang S, Brown PD, Barrand MA, Hladky SB. Kv1 and Kir2 potassium channels are expressed in rat brain endothelial cells. Pflugers Arch 2007; 456:379-91. [DOI: 10.1007/s00424-007-0377-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Accepted: 10/23/2007] [Indexed: 12/01/2022]
|
33
|
Abdul M, Hoosein N. Reduced Kv1.3 Potassium Channel Expression in Human Prostate Cancer. J Membr Biol 2007; 214:99-102. [PMID: 17546508 DOI: 10.1007/s00232-006-0065-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Revised: 11/13/2006] [Indexed: 10/23/2022]
Abstract
The presence of Kv1.3 voltage-gated potassium channels in rat and human prostate epithelial cells has been previously reported. We examined, by immunohistochemistry, Kv1.3 levels in 10 normal human prostate, 18 benign prostatic hyperplasia (BPH) and 147 primary human prostate cancer (Pca) specimens. We found high epithelial expression of Kv1.3 in all normal prostate, 16 BPH and 77 (52%) Pca specimens. Compared to normal, Kv1.3 levels were reduced in 1 (6%) BPH specimen and in 70 (48%) Pca specimens. We found a significant inverse correlation between Kv1.3 levels and tumor grade (r = -0.25, P = 0.003) as well as tumor stage (r = -0.27, P = 0.001). Study of an additional 30 primary Pca specimens showed that 15 (50%) had reduced Kv1.3 immunostaining compared to matched normal prostate tissue. Our data suggest that in Pca reduced Kv1.3 expression occurs frequently and may be associated with a poor outcome.
Collapse
Affiliation(s)
- Mansoor Abdul
- Biology Department, Claflin University, 400 Magnolia Street, Orangeburg, SC 29115, USA
| | | |
Collapse
|
34
|
Prevarskaya N, Skryma R, Bidaux G, Flourakis M, Shuba Y. Ion channels in death and differentiation of prostate cancer cells. Cell Death Differ 2007; 14:1295-304. [PMID: 17479110 DOI: 10.1038/sj.cdd.4402162] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Plasma membrane ion channels contribute to virtually all basic cellular processes, including such crucial ones for maintaining tissue homeostasis as proliferation, differentiation, and apoptosis. Enhanced proliferation, aberrant differentiation, and impaired ability to die are the prime reasons for abnormal tissue growth, which can eventually turn into uncontrolled expansion and invasion, characteristic of cancer. Prostate cancer (PCa) cells express a variety of plasma membrane ion channels. By providing the influx of essential signaling ions, perturbing intracellular ion concentrations, regulating cell volume, and maintaining membrane potential, PCa cells are critically involved in proliferation, differentiation, and apoptosis. PCa cells of varying metastatic ability can be distinguished by their ion channel characteristics. Increased malignancy and invasiveness of androgen-independent PCa cells is generally associated with the shift to a 'more excitable' phenotype of their plasma membrane. This shift is manifested by the appearance of voltage-gated Na(+) and Ca(2+) channels which contribute to their enhanced apoptotic resistance together with downregulated store-operated Ca(2+) influx, altered expression of different K(+) channels and members of the Transient Receptor Potential (TRP) channel family, and strengthened capability for maintaining volume constancy. The present review examines channel types expressed by PCa cells and their involvement in metastatic behaviors.
Collapse
Affiliation(s)
- N Prevarskaya
- Laboratoire de Physiologie Cellulaire, Equipe labellisée par la Ligue contre le cancer, INSERM U800, Université de Lille 1, Villeneuve d'Ascq F-59650, France.
| | | | | | | | | |
Collapse
|
35
|
Wang ZH, Shen B, Yao HL, Jia YC, Ren J, Feng YJ, Wang YZ. Blockage of intermediate-conductance-Ca(2+) -activated K(+) channels inhibits progression of human endometrial cancer. Oncogene 2007; 26:5107-14. [PMID: 17310992 DOI: 10.1038/sj.onc.1210308] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Potassium (K(+)) channels have been implicated in proliferation of some tumor cells. However, whether K(+) channels are important to the pathogenesis of endometrial cancer (EC) remains unknown. In the present study, we report that intermediate-conductance Ca(2+)-activated K(+) (IKCa1) channels play a critical role in the development of EC. The expression of IKCa1 at both mRNA and protein levels in EC tissues was greatly increased than that in atypical hyperplasia and normal tissues. Treatment of EC cells with clotrimazole and TRAM-34, two agents known to inhibit IKCa1 channels, suppressed the proliferation of EC cells and blocked EC cell cycle at G(0)/G(1) phase. Similarly, downregulation of IKCa1 by siRNA against IKCa1 inhibited EC cell proliferation and arrested its cell cycle at G(0)/G(1) phase. A clotrimazole-sensitive K(+) current was induced in EC cells in response to the increased Ca(2+). The current density induced by Ca(2+) was greatly reduced by clotrimazole, TRAM-34, charybdotoxin or downregulation of IKCa1 by the siRNA against IKCa1. Furthermore, TRAM-34 and clotrimazole slowed the formation in nude mice of tumor generated by injection of EC cells. Our results suggest that increased activity of IKCa1 channel is necessary for the development of EC.
Collapse
Affiliation(s)
- Z H Wang
- The Obstetrics and Gynecology Hospital of Medical Center of Fudan University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Teisseyre A, Mozrzymas JW. The influence of protons and zinc ions on the steady-state inactivation of Kv1.3 potassium channels. Cell Mol Biol Lett 2006; 12:220-30. [PMID: 17160583 PMCID: PMC6275785 DOI: 10.2478/s11658-006-0067-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Accepted: 10/12/2006] [Indexed: 11/20/2022] Open
Abstract
Using the whole-cell patch-clamp technique, we investigated the influence of extracellular pH and zinc ions (Zn(2+)) on the steady-state inactivation of Kv1.3 channels expressed in human lymphocytes. The obtained data showed that lowering the extracellular pH from 7.35 to 6.8 shifted the inactivation midpoint (V(i)) by 17.4 +/- 1.12 mV (n = 6) towards positive membrane potentials. This shift was statistically significant (p < 0.05). Applying 100 microM Zn(2+) at pH 6.8 further shifted the Vi value by 16.55 +/- 1.80 mV (n = 6) towards positive membrane potentials. This shift was also statistically significant (p < 0.05). The total shift of the Vi by protons and Zn(2+) was 33.95 +/- 1.90 mV (n = 6), which was significantly higher (p < 0.05) than the shift caused by Zn(2+) alone. The Zn(2+)-induced shift of the V(i) at pH 6.8 was almost identical to the shift at pH = 7.35. Thus, the proton-and Zn(2+)-induced shifts of the V(i) value were additive. The steady-state inactivation curves as a function of membrane voltage were compared with the functions of the steady-state activation. The total shift of the steady-state inactivation was almost identical to the total shift of the steady-state activation (32.01 +/- 2.10 mV, n = 10). As a result, the "windows" of membrane potentials in which the channels can be active under physiological conditions were also markedly shifted towards positive membrane potentials. The values of membrane voltage and the normalised chord conductance corresponding to the points of intersection of the curves of steady-state activation and inactivation were also calculated. The possible physiological significance of the observed modulatory effects is discussed herein.
Collapse
Affiliation(s)
- Andrzej Teisseyre
- Department of Biophysics, Wrocław Medical University, Wrocław, Poland.
| | | |
Collapse
|
37
|
Stühmer W, Alves F, Hartung F, Zientkowska M, Pardo LA. Potassium channels as tumour markers. FEBS Lett 2006; 580:2850-2. [PMID: 16783874 DOI: 10.1016/j.febslet.2006.03.062] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An increasing number of ion channels are being found to be causally involved in diseases, giving rise to the new field of "channelopathies". Cancer is no exception, and several ion channels have been linked to tumour progression. Among them is the potassium channel EAG (Ether-a-go-go). Over 75% of tumours have been tested positive using a monoclonal antibody specific for EAG, while inhibition of this channel decreased the proliferation of EAG expressing cells. The inhibition of EAG is accomplished using RNA interference, functional anti-EAG1 antibodies, or (unspecific) EAG channel blockers. Fluorescently labelled recombinant Fab fragments recognizing EAG allow the distribution of EAG to be visualized in an in vivo mouse tumour model.
Collapse
Affiliation(s)
- Walter Stühmer
- Max-Planck Institute for Experimental Medicine, Göttingen, Germany.
| | | | | | | | | |
Collapse
|
38
|
Pardo LA, Contreras-Jurado C, Zientkowska M, Alves F, Stühmer W. Role of voltage-gated potassium channels in cancer. J Membr Biol 2006; 205:115-24. [PMID: 16362499 DOI: 10.1007/s00232-005-0776-1] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Indexed: 01/12/2023]
Abstract
Ion channels are being associated with a growing number of diseases including cancer. This overview summarizes data on voltage-gated potassium channels (VGKCs) that exhibit oncogenic properties: ether-à-go-go type 1 (Eag1). Normally, Eag1 is expressed almost exclusively in tissue of neural origin, but its ectopic expression leads to uncontrolled proliferation, while inhibition of Eag1 expression produces a concomitant reduction in proliferation. Specific monoclonal antibodies against Eag1 recognize an epitope in over 80% of human tumors of diverse origins, endowing it with diagnostic and therapeutic potential. Eag1 also possesses unique electrophysiological properties that simplify its identification. This is particularly important, as specific blockers of Eag1 currents are not available. Molecular imaging of Eag1 in live tumor models has been accomplished with dye-tagged antibodies using 3-D imaging techniques in the near-infrared spectral range.
Collapse
Affiliation(s)
- L A Pardo
- Max-Planck-Institute of Experimental Medicine, Hermann Rein Str. 3, Göttingen, 37075 Germany.
| | | | | | | | | |
Collapse
|
39
|
Fraser SP, Diss JKJ, Chioni AM, Mycielska ME, Pan H, Yamaci RF, Pani F, Siwy Z, Krasowska M, Grzywna Z, Brackenbury WJ, Theodorou D, Koyutürk M, Kaya H, Battaloglu E, De Bella MT, Slade MJ, Tolhurst R, Palmieri C, Jiang J, Latchman DS, Coombes RC, Djamgoz MBA. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis. Clin Cancer Res 2006; 11:5381-9. [PMID: 16061851 DOI: 10.1158/1078-0432.ccr-05-0327] [Citation(s) in RCA: 349] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Ion channel activity is involved in several basic cellular behaviors that are integral to metastasis (e.g., proliferation, motility, secretion, and invasion), although their contribution to cancer progression has largely been ignored. The purpose of this study was to investigate voltage-gated Na(+) channel (VGSC) expression and its possible role in human breast cancer. EXPERIMENTAL DESIGN Functional VGSC expression was investigated in human breast cancer cell lines by patch clamp recording. The contribution of VGSC activity to directional motility, endocytosis, and invasion was evaluated by in vitro assays. Subsequent identification of the VGSC alpha-subunit(s) expressed in vitro was achieved using reverse transcription-PCR, immunocytochemistry, and Western blot techniques and used to investigate VGSCalpha expression and its association with metastasis in vivo. RESULTS VGSC expression was significantly up-regulated in metastatic human breast cancer cells and tissues, and VGSC activity potentiated cellular directional motility, endocytosis, and invasion. Reverse transcription-PCR revealed that Na(v)1.5, in its newly identified "neonatal" splice form, was specifically associated with strong metastatic potential in vitro and breast cancer progression in vivo. An antibody specific for this form confirmed up-regulation of neonatal Na(v)1.5 protein in breast cancer cells and tissues. Furthermore, a strong correlation was found between neonatal Na(v)1.5 expression and clinically assessed lymph node metastasis. CONCLUSIONS Up-regulation of neonatal Na(v)1.5 occurs as an integral part of the metastatic process in human breast cancer and could serve both as a novel marker of the metastatic phenotype and a therapeutic target.
Collapse
Affiliation(s)
- Scott P Fraser
- Neuroscience Solutions to Cancer Research Group, Department of Biological Sciences, Imperial College London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Teisseyre A, Michalak K. Genistein inhibits the activity of kv1.3 potassium channels in human T lymphocytes. J Membr Biol 2006; 205:71-9. [PMID: 16283587 DOI: 10.1007/s00232-005-0764-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 06/21/2005] [Indexed: 10/25/2022]
Abstract
In the present study, the whole-cell patch-clamp technique was applied to follow the inhibitory effect of genistein--a tyrosine kinase inhibitor and a natural anticancer agent--on the activity of voltage-gated potassium channels Kv1.3 expressed in human T lymphocytes (TL). Obtained data provide evidence that genistein application in the concentration range of 1-80 microM reversibly decreased the whole-cell potassium currents in TL in a concentration-dependent manner to about 0.23 of the control value. The half-blocking concentration range of genistein was from 10 to 40 microM. The current inhibition was correlated in time with a significant decrease of the current activation rate. The steady-state activation of the currents was unchanged upon application of genistein, as was the inactivation rate. The inhibitory effect of genistein on the current amplitude and activation kinetics was voltage-independent. The current inhibition was not changed significantly in the presence of 1 mM of sodium orthovanadate, a tyrosine phosphatase inhibitor. Application of daidzein, an inactive genistein analogue, did not affect significantly either the current amplitudes or the activation kinetics. Possible mechanisms of the observed phenomena and their significance for genistein-induced inhibition of cancer cell proliferation are discussed.
Collapse
Affiliation(s)
- A Teisseyre
- Department of Biophysics, Wrocław Medical University, ul. Chałubińskiego 10, Wrocław, Poland.
| | | |
Collapse
|
41
|
Chioni AM, Fraser SP, Pani F, Foran P, Wilkin GP, Diss JKJ, Djamgoz MBA. A novel polyclonal antibody specific for the Na(v)1.5 voltage-gated Na(+) channel 'neonatal' splice form. J Neurosci Methods 2005; 147:88-98. [PMID: 16111763 DOI: 10.1016/j.jneumeth.2005.03.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Revised: 03/22/2005] [Accepted: 03/24/2005] [Indexed: 11/20/2022]
Abstract
Voltage-gated Na(+) channel (VGSC) diversity is achieved through a number of mechanisms: multiple subunits, multiple genes encoding the pore-forming VGSC alpha-subunit and multiple gene isoforms generated by alternative splicing. A major type of VGSCalpha alternative splicing is in D1:S3, which has been proposed to be developmentally regulated. We recently reported a D1:S3 spliced form of Na(v)1.5 in human metastatic breast cancer cells. This novel 'neonatal' isoform differs from the counterpart 'adult' form at seven amino acids (in the extracellular loop between S3-S4 of D1). Here, we generated an anti-peptide polyclonal antibody, named NESOpAb, which specifically recognised 'neonatal' but not 'adult' Na(v)1.5 when tested on cells specifically over-expressing one or other of these Na(v)1.5 spliced forms. The antibody was used to investigate developmental expression of 'neonatal' Na(v)1.5 (nNa(v)1.5) in a range of mouse tissues by immunohistochemistry. Overall, the results were consistent with nNa(v)1.5 protein being more abundantly expressed in selected tissues (particularly heart and brain) from neonate as compared to adult animals. Importantly, NESOpAb blocked functional nNa(v)1.5 ion conductance when applied extracellularly at concentrations as low as 0.05 ng/ml. Possible biological and clinical applications of NESOpAb are discussed.
Collapse
Affiliation(s)
- Athina-Myrto Chioni
- Division of Cell and Molecular Biology, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | | | | | | | | | | | | |
Collapse
|
42
|
Ding Y, Robbins J, Fraser SP, Grimes JA, Djamgoz MBA. Comparative studies of intracellular Ca2+ in strongly and weakly metastatic rat prostate cancer cell lines. Int J Biochem Cell Biol 2005; 38:366-75. [PMID: 16300989 DOI: 10.1016/j.biocel.2005.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2005] [Revised: 07/21/2005] [Accepted: 07/29/2005] [Indexed: 11/26/2022]
Abstract
The metastatic ability of prostate cancer cells involves differential expression of ionic mechanisms. In the present study, using electrophysiological recordings and intracellular Ca2+ measurements, we investigated Ca2+ related signalling in two rat prostate cancer (MAT-LyLu and AT-2) cell lines of markedly different metastatic potential. Whole-cell voltage clamp experiments indicated the absence of an inward current carried through voltage-dependent Ca2+ channels in either cell line. A Ca2+-dependent component was also absent in the voltage-activated outward K+ currents. Indo-1 microfluorimetry confirmed these results and also revealed marked differences in the resting level of intracellular Ca2+ and the ability of the two cell lines to regulate intracellular Ca2+. The weakly metastatic AT-2 cells displayed a significantly higher resting intracellular Ca2+ than the related but strongly metastatic MAT-LyLu cell line. Increasing extracellular K+ decreased intracellular Ca2+ in the AT-2 but had no effect on intracellular Ca2+ levels in the MAT-LyLu cells. Furthermore, increasing extracellular Ca2+ increased intracellular Ca2+ in AT-2 but, again, had no effect on MAT-LyLu cells. These results suggested the presence of a tonic, voltage-independent Ca2+ permeation mechanism operating specifically in the AT-2 cells. The influx of Ca2+ into the AT-2 cells was suppressed by both CdCl2 (100-300 microM) and SKF-96365 (10-30 microM). It is concluded that the strongly metastatic MAT-LyLu cell line lacks a voltage-independent basal Ca2+ influx mechanism that is present in the weakly metastatic AT-2 cells.
Collapse
Affiliation(s)
- Y Ding
- Department of Biological Sciences, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, UK
| | | | | | | | | |
Collapse
|
43
|
Diss JKJ, Stewart D, Pani F, Foster CS, Walker MM, Patel A, Djamgoz MBA. A potential novel marker for human prostate cancer: voltage-gated sodium channel expression in vivo. Prostate Cancer Prostatic Dis 2005; 8:266-73. [PMID: 16088330 DOI: 10.1038/sj.pcan.4500796] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Functional expression of voltage-gated sodium channel alpha-subunits (VGSCalphas), specifically Nav1.7, is associated with strong metastatic potential in prostate cancer (CaP) in vitro. Furthermore, VGSC activity in vitro directly potentiates processes integral to metastasis. To investigate VGSCalpha expression in CaP in vivo, immunohistochemistry and real-time PCR were performed on human prostate biopsies (n>20). VGSCalpha immunostaining was evident in prostatic tissues and markedly stronger in CaP vs non-CaP patients. Importantly, RT-PCRs identified Nav1.7 as the VGSCalpha most strikingly upregulated (approximately 20-fold) in CaP, and the resultant receiver-operating characteristics curve demonstrated high diagnostic efficacy for the disease. It is concluded that VGSCalpha expression increases significantly in CaP in vivo and that Nav1.7 is a potential functional diagnostic marker.
Collapse
Affiliation(s)
- J K J Diss
- Department of Biological Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London, UK
| | | | | | | | | | | | | |
Collapse
|
44
|
O'Grady SM, Lee SY. Molecular diversity and function of voltage-gated (Kv) potassium channels in epithelial cells. Int J Biochem Cell Biol 2005; 37:1578-94. [PMID: 15882958 DOI: 10.1016/j.biocel.2005.04.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 03/27/2005] [Accepted: 04/05/2005] [Indexed: 01/17/2023]
Abstract
Voltage-gated K+ channels belonging to Kv1-9 subfamilies are widely expressed in excitable cells where they play an essential role in membrane hyperpolarization during an action potential and in the propagation of action potentials along the plasma membrane. Early patch clamp studies on epithelial cells revealed the presence of K+ currents with biophysical and pharmacologic properties characteristic of Kv channels expressed in excitable cells. More recently, molecular approaches including PCR and the availability of more selective antibodies directed against Kv alpha and auxiliary subunits, have demonstrated that epithelial cells from various organ systems, express a remarkable diversity Kv channel subunits. Unlike neurons and myocytes however, epithelial cells do not typically generate action potentials or exhibit dynamic changes in membrane potential necessary for activation of Kv alpha subunits. Moreover, the fact that many Kv channels expressed in epithelial cells exhibit inactivation suggest that their activities are relatively transient, making it difficult to ascribe a functional role for these channels in transepithelial electrolyte or nutrient transport. Other proposed functions have included (i) cell migration and wound healing, (ii) cell proliferation and cancer, (iii) apoptosis and (iv) O2 sensing. Certain Kv channels, particularly Kv1 and Kv2 subfamily members, have been shown to be involved in the proliferation of prostate, colon, lung and breast carcinomas. In some instances, a significant increase in Kv channel expression has been correlated with tumorogenesis suggesting the possibility of using these proteins as markers for transformation and perhaps reducing the rate of tumor growth by selectively inhibiting their functional activity.
Collapse
Affiliation(s)
- Scott M O'Grady
- Department of Physiology, University of Minnesota, 495 Animal Science/Veterinary Medicine Building, 1998 Fitch Avenue, St. Paul, MN 55108, USA.
| | | |
Collapse
|
45
|
Abstract
It is commonly accepted that cells require K(+) channels to proliferate. The role(s) of K(+) channels in the process is, however, poorly understood. Cloning of K(+) channel genes opened the possibility to approach this problem in a way more independent from pharmacological tools. Recent work shows that several identified K(+) channels are important in both physiological and pathological cell proliferation and open a promising pathway for novel targeted therapies.
Collapse
Affiliation(s)
- Luis A Pardo
- Max-Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
46
|
Ding Y, Djamgoz MBA. Serum concentration modifies amplitude and kinetics of voltage-gated Na+ current in the Mat-LyLu cell line of rat prostate cancer. Int J Biochem Cell Biol 2004; 36:1249-60. [PMID: 15109569 DOI: 10.1016/j.biocel.2003.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2003] [Revised: 10/20/2003] [Accepted: 10/20/2003] [Indexed: 11/17/2022]
Abstract
Voltage-gated Na+ channel (VGSC) expression has previously been shown to be upregulated in strongly metastatic prostate cancer cells (rat and human) and its activity shown to potentiate a variety of cellular behaviours integral to the metastatic cascade. However, the mechanism(s) responsible for the Na+ channel upregulation is not known. As a step towards evaluating the role of the extracellular biochemical environment in this regard, we have determined the effects of serum concentration on characteristics of Na+ channel expressed in the strongly metastatic Mat-LyLu rat prostate cancer cell line. Whole-cell patch-clamp recording techniques were used to study the effects of serum concentrations, above and below the normal 1%. Both the amplitude and the kinetics of the currents were analysed. The following results were obtained: (1) Adding 1% foetal calf serum to cells starved of serum for 24h increased Na+ current density; however, increasing serum concentration further (to 5%) caused a reduction. (2) Serum-free medium produced Na+ currents with slower kinetics of activation (time to peak) and inactivation (exponential decay). (3) Increased serum concentration (a) shifted steady-state inactivation to more positive potentials without affecting conductance and (b) increased tetrodotoxin sensitivity. It is concluded that serum concentration is an important determinant of the Na+ channel characteristics leading to possible transcriptional and post-translational modifications of channel expression and/or activity. Experiments are now needed to determine which constituents (protein hormones, growth factors, etc.) are responsible for these effects.
Collapse
Affiliation(s)
- Yanning Ding
- Neuroscience Solutions to Cancer Research Group, Department of Biological Sciences, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| | | |
Collapse
|
47
|
Abstract
The sigma (sigma) receptor and its agonists have been implicated in a myriad of cellular functions, biological processes and diseases. Whereas the precise molecular mechanism(s) of sigma receptors and their involvement in cancer cell biology have not been elucidated, recent work has started to shed some light on these issues. A molecular model has been proposed for the cloned sigma1 receptor; the precise molecular nature of the sigma2 receptor remains unknown. sigma receptors have been found to be frequently up-regulated in human cancer cells and tissues. sigma2 receptor drugs particularly have been shown to have antiproliferative effects. An interesting possibility is that sigma and/or sigma1 drugs could produce anticancerous effects by modulating ion channels. As well as proliferation, a variety of other metastatic cellular behaviors such as adhesion, motility, and secretion may also be affected. Other mechanisms of sigma receptor action may involve interaction with ankyrin and modulation of intracellular Ca(2+) and sphingolipid levels. Although more research is needed to further define the molecular physiology of sigma receptors, their involvement in the cellular pathophysiology of cancer raises the possibility that sigma drugs could be useful as novel therapeutic agents.
Collapse
Affiliation(s)
- Ebru Aydar
- Department of Biological Sciences, Sir Alexander Fleming Building, Imperial College, South Kensington Campus, London SW7 2AZ, United Kingdom.
| | | | | |
Collapse
|