1
|
Sakellakis M, Yoon SM, Reet J, Chalkias A. Novel insights into voltage-gated ion channels: Translational breakthroughs in medical oncology. Channels (Austin) 2024; 18:2297605. [PMID: 38154047 PMCID: PMC10761148 DOI: 10.1080/19336950.2023.2297605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/05/2023] [Indexed: 12/30/2023] Open
Abstract
Preclinical evidence suggests that voltage gradients can act as a kind of top-down master regulator during embryogenesis and orchestrate downstream molecular-genetic pathways during organ regeneration or repair. Moreover, electrical stimulation shifts response to injury toward regeneration instead of healing or scarring. Cancer and embryogenesis not only share common phenotypical features but also commonly upregulated molecular pathways. Voltage-gated ion channel activity is directly or indirectly linked to the pathogenesis of cancer hallmarks, while experimental and clinical studies suggest that their modulation, e.g., by anesthetic agents, may exert antitumor effects. A large recent clinical trial served as a proof-of-principle for the benefit of preoperative use of topical sodium channel blockade as a potential anticancer strategy against early human breast cancers. Regardless of whether ion channel aberrations are primary or secondary cancer drivers, understanding the functional consequences of these events may guide us toward the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Minas Sakellakis
- Department of Medicine, Jacobi North Central Bronx Hospital, Bronx, USA
| | - Sung Mi Yoon
- Department of Medicine, Jacobi North Central Bronx Hospital, Bronx, USA
| | - Jashan Reet
- Department of Medicine, Jacobi North Central Bronx Hospital, Bronx, USA
| | - Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Outcomes Research Consortium, Cleveland, OH, USA
| |
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
|
Balboni A, D'Angelo C, Collura N, Brusco S, Di Berardino C, Targa A, Massoti B, Mastrangelo E, Milani M, Seneci P, Broccoli V, Muzio L, Galli R, Menegon A. Acid-sensing ion channel 3 is a new potential therapeutic target for the control of glioblastoma cancer stem cells growth. Sci Rep 2024; 14:20421. [PMID: 39227705 PMCID: PMC11372124 DOI: 10.1038/s41598-024-71623-9] [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: 02/02/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain cancer that, despite recent advances in the understanding of its pathogenesis, remains incurable. GBM contains a subpopulation of cells with stem cell-like properties called cancer stem cells (CSCs). Several studies have demonstrated that CSCs are resistant to conventional chemotherapy and radiation thus representing important targets for novel anti-cancer therapies. Proton sensing receptors expressed by CSCs could represent important factors involved in the adaptation of tumours to the extracellular environment. Accordingly, the expression of acid-sensing ion channels (ASICs), proton-gated sodium channels mainly expressed in the neurons of peripheral (PNS) and central nervous system (CNS), has been demonstrated in several tumours and linked to an increase in cell migration and proliferation. In this paper we report that the ASIC3 isoform, usually absent in the CNS and present in the PNS, is enriched in human GBM CSCs while poorly expressed in the healthy human brain. We propose here a novel therapeutic strategy based on the pharmacological activation of ASIC3, which induces a significant GBM CSCs damage while being non-toxic for neurons. This approach might offer a promising and appealing new translational pathway for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Andrea Balboni
- Experimental Imaging Centre, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | - Camilla D'Angelo
- Experimental Imaging Centre, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | - Nicoletta Collura
- Experimental Imaging Centre, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | - Simone Brusco
- Division of Neuroscience, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
- Electrophysiology Unit, Axxam S.P.A., Via Meucci 3, Bresso, 20091, Milan, Italy
| | - Claudia Di Berardino
- Division of Neuroscience, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | - Altea Targa
- Experimental Imaging Centre, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | - Beatrice Massoti
- Experimental Imaging Centre, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | | | | | | | - Vania Broccoli
- Division of Neuroscience, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
- CNR-Institute of Neuroscience, Milan, Italy
| | - Luca Muzio
- INsPE, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy
| | - Rossella Galli
- Neural Stem Cell Biology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Menegon
- Experimental Imaging Centre, San Raffaele Scientific Institute IRCCS, 20132, Milan, Italy.
| |
Collapse
|
4
|
Jia Y, Xu X, Liu Y, Shen H, Sun S, Sun G. Effect of calcium concentration on metastasis of hepatocellular carcinoma cells cultured in alginate gel beads. Colloids Surf B Biointerfaces 2024; 245:114201. [PMID: 39255748 DOI: 10.1016/j.colsurfb.2024.114201] [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: 07/11/2024] [Revised: 08/28/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024]
Abstract
Changes in sodium alginate and calcium ion concentrations have a considerable impact on the structural properties of calcium alginate gel (ALG) beads, consequently influencing the biological characteristics of the cells encapsulated within them. This study aimed to examine the effects of calcium on the metastatic potential of hepatocellular carcinoma (HCC) cells encapsulated in ALG beads. The results showed that the invasion ability of HCC cells significantly increased when they were encapsulated in beads prepared with a calcium concentration of 200 mM compared to those prepared with a calcium concentration of 50 mM. Furthermore, the expression levels of genes related to metastasis were significantly elevated in ALG beads prepared with a calcium concentration of 200 mM. Specifically, the expression of activated matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), and urokinase-type plasminogen activator system proteins was found to be high. Conversely, the expression of phosphatase and tensin homolog deleted on chromosome 10 was observed to be significantly reduced. These findings indicate that manipulating the calcium ion concentration during the fabrication of ALG beads enables the generation of three-dimensional HCC cells with varying metastatic capacities. This model offers a valuable tool for investigating the mechanisms underlying liver cancer metastasis and screening potential therapeutic drugs.
Collapse
Affiliation(s)
- Yunbo Jia
- Innovative Engineering Technology Research Center for Cell Therapy, Shengjing Hospital of China Medical University, Shenyang, China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoxi Xu
- Laboratory of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yang Liu
- Innovative Engineering Technology Research Center for Cell Therapy, Shengjing Hospital of China Medical University, Shenyang, China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China; Laboratory of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Hongfei Shen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Siyu Sun
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guangwei Sun
- Innovative Engineering Technology Research Center for Cell Therapy, Shengjing Hospital of China Medical University, Shenyang, China; Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China; Laboratory of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China.
| |
Collapse
|
5
|
Aravind A, Mathew RT, Kuruba L, Vijayakumar M, Prasad TSK. Proteomic analysis of peripheral blood mononuclear cells from OSCC patients reveals potential immune checkpoints to enable personalized treatment. Mol Omics 2024. [PMID: 39177064 DOI: 10.1039/d4mo00112e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most prevalent cancers worldwide, with high mortality and prevalence rates. OSCC is defined as an immunogenic tumor with the potential to be recognized and targeted by the immune system. It is characterized by the extensive infiltration of immune cells and plays a vital role in tumorigenesis. Peripheral blood mononuclear cells (PBMC) are a functional subset of immune cells readily accessible through minimally invasive procedures. The molecular characterization of immune cells aids in understanding their functional roles in various pathophysiological conditions. Proteomic analysis of PBMCs from cancer patients provides insight into the mechanism of immunoregulation and the role of immune cells in impeding tumor development and progression. Therefore, the present study investigated the immune cell proteome of a cancer control cohort within OSCC, leveraging data-independent acquisition analysis by mass spectrometry (DIA-MS). Among the differentially abundant proteins in OSCC, we identified promising molecular targets, including LMNB1, CTSB, CD14, CD177, and SPI1. Further exploration of the signaling pathways related to the candidate molecules demonstrated their involvement in cancer immunomodulation. Therefore, this study can serve as a platform for identifying new candidate proteins to further investigate their potential as immunotherapeutic targets and prognostic markers.
Collapse
Affiliation(s)
- Anjana Aravind
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka - 575018, India.
| | - Rohan Thomas Mathew
- Department of Surgical Oncology, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, Karnataka - 575018, India.
| | - Lepakshi Kuruba
- Department of Medical Oncology, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, Karnataka - 575018, India
| | - Manavalan Vijayakumar
- Department of Surgical Oncology, Yenepoya Medical College, Yenepoya (Deemed to be University), Mangalore, Karnataka - 575018, India.
| | | |
Collapse
|
6
|
Haustrate A, Cordier C, Shapovalov G, Mihalache A, Desruelles E, Soret B, Essonghé NC, Spriet C, Yassine M, Barras A, Marines J, Alcaraz LB, Szunerits S, Robin G, Gosset P, Prevarskaya N, Lehen'kyi V. Trpv6 channel targeting using monoclonal antibody induces prostate cancer cell apoptosis and tumor regression. Cell Death Dis 2024; 15:419. [PMID: 38879621 PMCID: PMC11180136 DOI: 10.1038/s41419-024-06809-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/19/2024]
Abstract
TRPV6 calcium channel is a prospective target in prostate cancer (PCa) since it is not expressed in healthy prostate while its expression increases during cancer progression. Despite the role of TRPV6 in PCa cell survival and apoptotic resistance has been already established, no reliable tool to target TRPV6 channel in vivo and thus to reduce tumor burden is known to date. Here we report the generation of mouse monoclonal antibody mAb82 raised against extracellular epitope of the pore region of the channel. mAb82 inhibited TRPV6 currents by 90% at 24 µg/ml in a dose-dependent manner while decreasing store-operated calcium entry to 56% at only 2.4 µg/ml. mAb82 decreased PCa survival rate in vitro by 71% at 12 µg/ml via inducing cell death through the apoptosis cascade via activation of the protease calpain, following bax activation, mitochondria enlargement, and loss of cristae, Cyt C release, pro-caspase 9 cleavage with the subsequent activation of caspases 3/7. In vivo, mice bearing either PC3Mtrpv6+/+ or PC3Mtrpv6-/-+pTRPV6 tumors were successfully treated with mAb82 at the dose as low as 100 µg/kg resulting in a significant reduction tumor growth by 31% and 90%, respectively. The survival rate was markedly improved by 3.5 times in mice treated with mAb82 in PC3Mtrpv6+/+ tumor group and completely restored in PC3Mtrpv6-/-+pTRPV6 tumor group. mAb82 showed a TRPV6-expression dependent organ distribution and virtually no toxicity in the same way as mAbAU1, a control antibody of the same Ig2a isotype. Overall, our data demonstrate for the first time the use of an anti-TRPV6 monoclonal antibody in vitro and in vivo in the treatment of the TRPV6-expressing PCa tumors.
Collapse
Affiliation(s)
- Aurélien Haustrate
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
- FONDATION ARC, 9 rue Guy Môquet, 94830, Villejuif, France
| | - Clément Cordier
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - George Shapovalov
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - Adriana Mihalache
- Service d'Anatomie et de Cytologie Pathologiques, Groupement des Hôpitaux de l'Institut Catholique de Lille (GHICL), 59000, Lille, France
| | - Emilie Desruelles
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - Benjamin Soret
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - Nadège Charlène Essonghé
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - Corentin Spriet
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, F-59000, Lille, France
| | - Maya Yassine
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - Alexandre Barras
- University of Lille, CNRS, University Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | | | | | - Sabine Szunerits
- University of Lille, CNRS, University Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Gautier Robin
- Mabqi, Cap Sigma, Zac Euromédecine II, Grabels, France
| | - Pierre Gosset
- Service d'Anatomie et de Cytologie Pathologiques, Groupement des Hôpitaux de l'Institut Catholique de Lille (GHICL), 59000, Lille, France
| | - Natalia Prevarskaya
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France
| | - V'yacheslav Lehen'kyi
- Department of Biology, Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Faculty of Science and Technologies, University of Lille, 59650, Villeneuve d'Ascq, France.
- FONDATION ARC, 9 rue Guy Môquet, 94830, Villejuif, France.
| |
Collapse
|
7
|
Doldi V, Tortoreto M, Colecchia M, Maffezzini M, Percio S, Giammello F, Brandalise F, Gandellini P, Zaffaroni N. Repositioning of antiarrhythmics for prostate cancer treatment: a novel strategy to reprogram cancer-associated fibroblasts towards a tumor-suppressive phenotype. J Exp Clin Cancer Res 2024; 43:161. [PMID: 38858661 PMCID: PMC11165820 DOI: 10.1186/s13046-024-03081-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/25/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) play a significant role in fueling prostate cancer (PCa) progression by interacting with tumor cells. A previous gene expression analysis revealed that CAFs up-regulate genes coding for voltage-gated cation channels, as compared to normal prostate fibroblasts (NPFs). In this study, we explored the impact of antiarrhythmic drugs, known cation channel inhibitors, on the activated state of CAFs and their interaction with PCa cells. METHODS The effect of antiarrhythmic treatment on CAF activated phenotype was assessed in terms of cell morphology and fibroblast activation markers. CAF contractility and migration were evaluated by 3D gel collagen contraction and scratch assays, respectively. The ability of antiarrhythmics to impair CAF-PCa cell interplay was investigated in CAF-PCa cell co-cultures by assessing tumor cell growth and expression of epithelial-to-mesenchymal transition (EMT) markers. The effect on in vivo tumor growth was assessed by subcutaneously injecting PCa cells in SCID mice and intratumorally administering the medium of antiarrhythmic-treated CAFs or in co-injection experiments, where antiarrhythmic-treated CAFs were co-injected with PCa cells. RESULTS Activated fibroblasts show increased membrane conductance for potassium, sodium and calcium, consistently with the mRNA and protein content analysis. Antiarrhythmics modulate the expression of fibroblast activation markers. Although to a variable extent, these drugs also reduce CAF motility and hinder their ability to remodel the extracellular matrix, for example by reducing MMP-2 release. Furthermore, conditioned medium and co-culture experiments showed that antiarrhythmics can, at least in part, reverse the protumor effects exerted by CAFs on PCa cell growth and plasticity, both in androgen-sensitive and castration-resistant cell lines. Consistently, the transcriptome of antiarrhythmic-treated CAFs resembles that of tumor-suppressive NPFs. In vivo experiments confirmed that the conditioned medium or the direct coinjection of antiarrhythmic-treated CAFs reduced the tumor growth rate of PCa xenografts. CONCLUSIONS Collectively, such data suggest a new therapeutic strategy for PCa based on the repositioning of antiarrhythmic drugs with the aim of normalizing CAF phenotype and creating a less permissive tumor microenvironment.
Collapse
Affiliation(s)
- Valentina Doldi
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCSS Istituto Nazionale Dei Tumori, Milan, 20133, Italy.
| | - Monica Tortoreto
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCSS Istituto Nazionale Dei Tumori, Milan, 20133, Italy
| | - Maurizio Colecchia
- Vita-Salute San Raffaele University, IRCCS San Raffaele Hospital and Scientific Institute, Milan, 20132, Italy
| | - Massimo Maffezzini
- Department of Urology, Hospitals of Legnano and Magenta, Milan, 20013, Italy
| | - Stefano Percio
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCSS Istituto Nazionale Dei Tumori, Milan, 20133, Italy
| | | | | | - Paolo Gandellini
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCSS Istituto Nazionale Dei Tumori, Milan, 20133, Italy
| |
Collapse
|
8
|
Arponen O, McLean MA, Nanaa M, Manavaki R, Baxter GC, Gill AB, Riemer F, Kennerley AJ, Woitek R, Kaggie JD, Brackenbury WJ, Gilbert FJ. 23Na MRI: inter-reader reproducibility of normal fibroglandular sodium concentration measurements at 3 T. Eur Radiol Exp 2024; 8:75. [PMID: 38853182 PMCID: PMC11162986 DOI: 10.1186/s41747-024-00465-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 04/08/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND To study the reproducibility of 23Na magnetic resonance imaging (MRI) measurements from breast tissue in healthy volunteers. METHODS Using a dual-tuned bilateral 23Na/1H breast coil at 3-T MRI, high-resolution 23Na MRI three-dimensional cones sequences were used to quantify total sodium concentration (TSC) and fluid-attenuated sodium concentration (FASC). B1-corrected TSC and FASC maps were created. Two readers manually measured mean, minimum and maximum TSC and mean FASC values using two sampling methods: large regions of interest (LROIs) and small regions of interest (SROIs) encompassing fibroglandular tissue (FGT) and the highest signal area at the level of the nipple, respectively. The reproducibility of the measurements and correlations between density, age and FGT apparent diffusion coefficient (ADC) values were evaluatedss. RESULTS Nine healthy volunteers were included. The inter-reader reproducibility of TSC and FASC using SROIs and LROIs was excellent (intraclass coefficient range 0.945-0.979, p < 0.001), except for the minimum TSC LROI measurements (p = 0.369). The mean/minimum LROI TSC and mean LROI FASC values were lower than the respective SROI values (p < 0.001); the maximum LROI TSC values were higher than the SROI TSC values (p = 0.009). TSC correlated inversely with age but not with FGT ADCs. The mean and maximum FGT TSC and FASC values were higher in dense breasts in comparison to non-dense breasts (p < 0.020). CONCLUSIONS The chosen sampling method and the selected descriptive value affect the measured TSC and FASC values, although the inter-reader reproducibility of the measurements is in general excellent. RELEVANCE STATEMENT 23Na MRI at 3 T allows the quantification of TSC and FASC sodium concentrations. The sodium measurements should be obtained consistently in a uniform manner. KEY POINTS • 23Na MRI allows the quantification of total and fluid-attenuated sodium concentrations (TSC/FASC). • Sampling method (large/small region of interest) affects the TSC and FASC values. • Dense breasts have higher TSC and FASC values than non-dense breasts. • The inter-reader reproducibility of TSC and FASC measurements was, in general, excellent. • The results suggest the importance of stratifying the sodium measurements protocol.
Collapse
Affiliation(s)
- Otso Arponen
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK.
| | - Mary A McLean
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - Muzna Nanaa
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - Roido Manavaki
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - Gabrielle C Baxter
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - Andrew B Gill
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - Frank Riemer
- Department of Radiology, Mohn Medical Imaging and Visualization Centre (MMIV), Haukeland University Hospital, Bergen, Norway
| | - Aneurin J Kennerley
- York Biomedical Research Institute, University of York, York, UK
- Department of Sports and Exercise Science, Institute of Sport, Manchester Metropolitan University, Manchester, UK
| | - Ramona Woitek
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
- Research Center for Medical Image Analysis and AI (MIAAI), Danube Private University, Krems, Austria
| | - Joshua D Kaggie
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| | - William J Brackenbury
- York Biomedical Research Institute, University of York, York, UK
- Department of Biology, University of York, York, UK
| | - Fiona J Gilbert
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Box 218, Cambridge, CB2 0QQ, UK
| |
Collapse
|
9
|
Michaels AM, Zoccarato A, Hoare Z, Firth G, Chung YJ, Kuchel PW, Shah AM, Shattock MJ, Southworth R, Eykyn TR. Disrupting Na + ion homeostasis and Na +/K + ATPase activity in breast cancer cells directly modulates glycolysis in vitro and in vivo. Cancer Metab 2024; 12:15. [PMID: 38783368 PMCID: PMC11119389 DOI: 10.1186/s40170-024-00343-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Glycolytic flux is regulated by the energy demands of the cell. Upregulated glycolysis in cancer cells may therefore result from increased demand for adenosine triphosphate (ATP), however it is unknown what this extra ATP turnover is used for. We hypothesise that an important contribution to the increased glycolytic flux in cancer cells results from the ATP demand of Na+/K+-ATPase (NKA) due to altered sodium ion homeostasis in cancer cells. METHODS Live whole-cell measurements of intracellular sodium [Na+]i were performed in three human breast cancer cells (MDA-MB-231, HCC1954, MCF-7), in murine breast cancer cells (4T1), and control human epithelial cells MCF-10A using triple quantum filtered 23Na nuclear magnetic resonance (NMR) spectroscopy. Glycolytic flux was measured by 2H NMR to monitor conversion of [6,6-2H2]D-glucose to [2H]-labelled L-lactate at baseline and in response to NKA inhibition with ouabain. Intracellular [Na+]i was titrated using isotonic buffers with varying [Na+] and [K+] and introducing an artificial Na+ plasma membrane leak using the ionophore gramicidin-A. Experiments were carried out in parallel with cell viability assays, 1H NMR metabolomics of intracellular and extracellular metabolites, extracellular flux analyses and in vivo measurements in a MDA-MB-231 human-xenograft mouse model using 2-deoxy-2-[18F]fluoroglucose (18F-FDG) positron emission tomography (PET). RESULTS Intracellular [Na+]i was elevated in human and murine breast cancer cells compared to control MCF-10A cells. Acute inhibition of NKA by ouabain resulted in elevated [Na+]i and inhibition of glycolytic flux in all three human cancer cells which are ouabain sensitive, but not in the murine cells which are ouabain resistant. Permeabilization of cell membranes with gramicidin-A led to a titratable increase of [Na+]i in MDA-MB-231 and 4T1 cells and a Na+-dependent increase in glycolytic flux. This was attenuated with ouabain in the human cells but not in the murine cells. 18FDG PET imaging in an MDA-MB-231 human-xenograft mouse model recorded lower 18FDG tumour uptake when treated with ouabain while murine tissue uptake was unaffected. CONCLUSIONS Glycolytic flux correlates with Na+-driven NKA activity in breast cancer cells, providing evidence for the 'centrality of the [Na+]i-NKA nexus' in the mechanistic basis of the Warburg effect.
Collapse
Affiliation(s)
- Aidan M Michaels
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Anna Zoccarato
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Zoe Hoare
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - George Firth
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Yu Jin Chung
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Philip W Kuchel
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Michael J Shattock
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Richard Southworth
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK
| | - Thomas R Eykyn
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
10
|
Langthaler S, Zumpf C, Rienmüller T, Shrestha N, Fuchs J, Zhou R, Pelzmann B, Zorn-Pauly K, Fröhlich E, Weinberg SH, Baumgartner C. The bioelectric mechanisms of local calcium dynamics in cancer cell proliferation: an extension of the A549 in silico cell model. Front Mol Biosci 2024; 11:1394398. [PMID: 38770217 PMCID: PMC11102976 DOI: 10.3389/fmolb.2024.1394398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction Advances in molecular targeting of ion channels may open up new avenues for therapeutic approaches in cancer based on the cells' bioelectric properties. In addition to in-vitro or in-vivo models, in silico models can provide deeper insight into the complex role of electrophysiology in cancer and reveal the impact of altered ion channel expression and the membrane potential on malignant processes. The A549 in silico model is the first computational cancer whole-cell ion current model that simulates the bioelectric mechanisms of the human non-small cell lung cancer cell line A549 during the different phases of the cell cycle. This work extends the existing model with a detailed mathematical description of the store-operated Ca2+ entry (SOCE) and the complex local intracellular calcium dynamics, which significantly affect the entire electrophysiological properties of the cell and regulate cell cycle progression. Methods The initial model was extended by a multicompartmental approach, addressing the heterogenous calcium profile and dynamics in the ER-PM junction provoked by local calcium entry of store-operated calcium channels (SOCs) and uptake by SERCA pumps. Changes of cytosolic calcium levels due to diffusion from the ER-PM junction, release from the ER by RyR channels and IP3 receptors, as well as corresponding PM channels were simulated and the dynamics evaluated based on calcium imaging data. The model parameters were fitted to available data from two published experimental studies, showing the function of CRAC channels and indirectly of IP3R, RyR and PMCA via changes of the cytosolic calcium levels. Results The proposed calcium description accurately reproduces the dynamics of calcium imaging data and simulates the SOCE mechanisms. In addition, simulations of the combined A549-SOCE model in distinct phases of the cell cycle demonstrate how Ca2+ - dynamics influence responding channels such as KCa, and consequently modulate the membrane potential accordingly. Discussion Local calcium distribution and time evolution in microdomains of the cell significantly impact the overall electrophysiological properties and exert control over cell cycle progression. By providing a more profound description, the extended A549-SOCE model represents an important step on the route towards a valid model for oncological research and in silico supported development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sonja Langthaler
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
| | - Christian Zumpf
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
| | - Theresa Rienmüller
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
| | - Niroj Shrestha
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Julia Fuchs
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
- Research Unit on Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Rui Zhou
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
| | - Brigitte Pelzmann
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Klaus Zorn-Pauly
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Seth H. Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
- Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Christian Baumgartner
- Institute of Health Care Engineering with European Testing Center for Medical Devices, Graz University of Technology, Graz, Austria
| |
Collapse
|
11
|
Bozdag A, Kuloglu T, Artas G, Aydin S. Investigation of Trpa1 and Trpc1 Immunreactivities in Colon Adenocarcinomas. Cancer Manag Res 2024; 16:377-384. [PMID: 38699653 PMCID: PMC11063473 DOI: 10.2147/cmar.s447549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Purpose As the normal colon epithelium differentiates into adenoma, invasive cancer and metastatic cancer, the cell acquires new characteristics such as apoptosis, proliferation, differentiation, invasion and metastasis. Many mechanisms are effective in acquiring these qualities. One of these is the regulation of the functioning of ion channels. This study aimed to examine TRPA1 and TRPC1 expression in colorectal adenocarcinomas showing different degrees of differentiation. Patients and Methods We examined the biopsy specimens of 60 patients diagnosed with colorectal adenocarcinomas, including those of patients with well-differentiated (n = 20), moderately differentiated (n = 20) and poorly differentiated (n = 20) carcinomas. Moreover, 20 biopsy specimens of individuals with normal colonic mucosa were examined. Histoscores were calculated for TRPA1 and TRPC1 based on the extent of diffusion and intensity of immunoreactivity, and these scores were compared statistically. Results A statistically significant increase in both TRPA1 and TRPC1 immunoreactivity was observed in low-grade and high-grade colon adenocarcinomas compared to the control group (p<0.001). A statistically significant decrease in both TRPA1 and TRPC1 immunoreactivity was observed in high-grade colon adenocarcinomas compared to low-grade colon adenocarcinomas (p<0.001). Conclusion TRPA1 and TRPC1 immunoreactivites are increased in colorectal adenocarcinoma tissue compared with the healthy tissue. Furthermore, the immunoreactivity decreases as the grade of cancer increases.
Collapse
Affiliation(s)
- Ahmet Bozdag
- Department of General Surgery, School of Medicine, Firat University, Elazig, Turkey
| | - Tuncay Kuloglu
- Department of Histology and Embryology, School of Medicine, Firat University, Elazig, Turkey
| | - Gokhan Artas
- Department of Pathology, School of Medicine, Firat University, Elazig, Turkey
| | - Suleyman Aydin
- Department of Biochemistry, School of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
12
|
Wang H, Cai H, Li L. Comprehensive analysis of m6A reader YTHDF2 prognosis, immune infiltration, and related regulatory networks in hepatocellular carcinoma. Heliyon 2024; 10:e23204. [PMID: 38163150 PMCID: PMC10756983 DOI: 10.1016/j.heliyon.2023.e23204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024] Open
Abstract
Background N6-Methyladenosine (m6A) RNA modification is the most prevalent internal modification pattern in eukaryotic mRNAs and plays critical roles in diverse physiological and pathological processes. However, the expression of m6A regulator YTHDF2, its prognostic value, its biological function, its correlation with tumor microenvironment (TME) immune infiltrates, and related regulatory networks in hepatocellular carcinoma (HCC) remain determined. Methods TCGA, GTEx, and GEO databases were used to investigate the expression profile of YTHDF2 in HCC. We performed differentially expressed genes (DEGs) analysis and constructed a PPI network to explore the biological processes of YTHDF2 in HCC. Kaplan-Meier curves and Cox regression analysis were used to assess the prognostic value of YTHDF2 and then a clinical prognostic nomogram was constructed. Additionally, ssGSEA was performed to assess the correlation between YTHDF2 and immune infiltration levels. The TISIDB database was applied to explore the expression of YTHDF2 in immune and molecular subtypes of HCC. GSEA identifies the YTHDF2-related signaling pathways. Finally, we utilized miRNet and starBase database to construct regulatory networks for HCC based on lncRNA-miRNA and miRNA-YTHDF2 interactions. Results YTHDF2 was significantly upregulated in HCC tumor tissues compared with the adjacent normal tissues. HCC patients in the high YTHDF2 expression group had poorer survival. Multivariate Cox analysis suggested that YTHDF2 may be a new independent prognostic indicator for HCC patients, with the prognostic nomogram exhibiting satisfactory results. YTHDF2 expression was significantly correlated with TME immune cell-infiltrating characteristics. Strong correlations were also shown in immune subtypes, molecular subtypes and immune checkpoints. Further analysis revealed that the combination of YTHDF2 expression and immune cell score was considerably associated with survival outcome in HCC patients. GESA analysis demonstrated that high YTHDF2 expression is associated with multiple biological processes and oncogenic pathways. Moreover, 14 possible regulatory networks were constructed, which are associated with HCC progression. Conclusion Our findings revealed that YTHDF2 may serve as a promising prognostic biomarker for HCC and may regulate the tumor immune microenvironment to provide effective therapeutic strategies.
Collapse
Affiliation(s)
- Hang Wang
- Shengli Clinical Medical College of Fujian Medical University, Department of Health Management, Fujian Provincial Hospital, Fuzhou, Fujian, China
- Shengli Clinical Medical College of Fujian Medical University, Department of Disease Prevention and Healthcare, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Hui Cai
- Shengli Clinical Medical College of Fujian Medical University, Department of Health Management, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Li Li
- Shengli Clinical Medical College of Fujian Medical University, Department of Health Management, Fujian Provincial Hospital, Fuzhou, Fujian, China
- Shengli Clinical Medical College of Fujian Medical University, Department of Disease Prevention and Healthcare, Fujian Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
13
|
Maliszewska-Olejniczak K, Bednarczyk P. Novel insights into the role of ion channels in cellular DNA damage response. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 793:108488. [PMID: 38266668 DOI: 10.1016/j.mrrev.2024.108488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
The DNA damage response (DDR) is a complex and highly regulated cellular process that detects and repairs DNA damage. The integrity of the DNA molecule is crucial for the proper functioning and survival of cells, as DNA damage can lead to mutations, genomic instability, and various diseases, including cancer. The DDR safeguards the genome by coordinating a series of signaling events and repair mechanisms to maintain genomic stability and prevent the propagation of damaged DNA to daughter cells. The study of an ion channels in the context of DDR is a promising avenue in biomedical research. Lately, it has been reported that the movement of ions through channels plays a crucial role in various physiological processes, including nerve signaling, muscle contraction, cell signaling, and maintaining cell membrane potential. Knowledge regarding the involvement of ion channels in the DDR could support refinement of our approach to several pathologies, mainly cancer, and perhaps lead to innovative therapies. In this review, we focused on the ion channel's possible role in the DDR. We present an analysis of the involvement of ion channels in DDR, their role in DNA repair mechanisms, and cellular outcomes. By addressing these areas, we aim to provide a comprehensive perspective on ion channels in the DDR and potentially guide future research in this field. It is worth noting that the interplay between ion channels and the cellular DDR is complex and multifaceted. More research is needed to fully understand the underlying mechanisms and potential therapeutic implications of these interactions.
Collapse
Affiliation(s)
- Kamila Maliszewska-Olejniczak
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland.
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| |
Collapse
|
14
|
Murugan NJ, Cariba S, Abeygunawardena S, Rouleau N, Payne SL. Biophysical control of plasticity and patterning in regeneration and cancer. Cell Mol Life Sci 2023; 81:9. [PMID: 38099951 PMCID: PMC10724343 DOI: 10.1007/s00018-023-05054-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Cells and tissues display a remarkable range of plasticity and tissue-patterning activities that are emergent of complex signaling dynamics within their microenvironments. These properties, which when operating normally guide embryogenesis and regeneration, become highly disordered in diseases such as cancer. While morphogens and other molecular factors help determine the shapes of tissues and their patterned cellular organization, the parallel contributions of biophysical control mechanisms must be considered to accurately predict and model important processes such as growth, maturation, injury, repair, and senescence. We now know that mechanical, optical, electric, and electromagnetic signals are integral to cellular plasticity and tissue patterning. Because biophysical modalities underly interactions between cells and their extracellular matrices, including cell cycle, metabolism, migration, and differentiation, their applications as tuning dials for regenerative and anti-cancer therapies are being rapidly exploited. Despite this, the importance of cellular communication through biophysical signaling remains disproportionately underrepresented in the literature. Here, we provide a review of biophysical signaling modalities and known mechanisms that initiate, modulate, or inhibit plasticity and tissue patterning in models of regeneration and cancer. We also discuss current approaches in biomedical engineering that harness biophysical control mechanisms to model, characterize, diagnose, and treat disease states.
Collapse
Affiliation(s)
- Nirosha J Murugan
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada.
- Allen Discovery Center, Tufts University, Medford, MA, USA.
| | - Solsa Cariba
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
- Allen Discovery Center, Tufts University, Medford, MA, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Samantha L Payne
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
15
|
Brackenbury WJ, Palmieri C. Blocking channels to metastasis: targeting sodium transport in breast cancer. Breast Cancer Res 2023; 25:140. [PMID: 37950273 PMCID: PMC10638823 DOI: 10.1186/s13058-023-01741-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
The development of therapies that can suppress invasion and prevent metastasis, 'anti-metastatic drugs', is an important area of unmet therapeutic need. The new results of a recent open-label, multicentre randomised trial published in J Clin Oncol showed a significant disease-free survival (DFS) benefit for breast cancer patients receiving presurgical, peritumoral injection of lidocaine, an amide local anaesthetic, which blocks voltage-gated sodium channels (VGSCs). VGSCs are expressed on electrically excitable cells, including neurons and cardiomyocytes, where they sustain rapid membrane depolarisation during action potential firing. As a result of this key biophysical function, VGSCs are important drug targets for excitability-related disorders, including epilepsy, neuropathic pain, affective disorders and cardiac arrhythmia. A growing body of preclinical evidence highlights VGSCs as key protagonists in regulating altered sodium influx in breast cancer cells, thus driving invasion and metastasis. Furthermore, prescription of certain VGSC-inhibiting medications has been associated with reduced cancer incidence and improved survival in several observational studies. Thus, VGSC-inhibiting drugs already in clinical use may be ideal candidates for repurposing as possible anti-metastatic therapies. While these results are promising, further work is required to establish whether other VGSC inhibitors may afford superior metastasis suppression. Finally, increasing preclinical evidence suggests that several other ion channels are also key drivers of cancer hallmarks; thus, there are undoubtedly further opportunities to harness ion transport inhibition that should also be explored.
Collapse
Affiliation(s)
- William J Brackenbury
- Department of Biology, University of York, Wentworth Way, Heslington, York, YO10 5DD, UK.
- York Biomedical Research Institute, University of York, Wentworth Way, Heslington, York, YO10 5DD, UK.
| | - Carlo Palmieri
- Institute of Systems, Molecular and Integrative Biology, Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
16
|
Ruffinatti FA, Scarpellino G, Chinigò G, Visentin L, Munaron L. The Emerging Concept of Transportome: State of the Art. Physiology (Bethesda) 2023; 38:0. [PMID: 37668550 DOI: 10.1152/physiol.00010.2023] [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: 04/12/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
The array of ion channels and transporters expressed in cell membranes, collectively referred to as the transportome, is a complex and multifunctional molecular machinery; in particular, at the plasma membrane level it finely tunes the exchange of biomolecules and ions, acting as a functionally adaptive interface that accounts for dynamic plasticity in the response to environmental fluctuations and stressors. The transportome is responsible for the definition of membrane potential and its variations, participates in the transduction of extracellular signals, and acts as a filter for most of the substances entering and leaving the cell, thus enabling the homeostasis of many cellular parameters. For all these reasons, physiologists have long been interested in the expression and functionality of ion channels and transporters, in both physiological and pathological settings and across the different domains of life. Today, thanks to the high-throughput technologies of the postgenomic era, the omics approach to the study of the transportome is becoming increasingly popular in different areas of biomedical research, allowing for a more comprehensive, integrated, and functional perspective of this complex cellular apparatus. This article represents a first effort for a systematic review of the scientific literature on this topic. Here we provide a brief overview of all those studies, both primary and meta-analyses, that looked at the transportome as a whole, regardless of the biological problem or the models they used. A subsequent section is devoted to the methodological aspect by reviewing the most important public databases annotating ion channels and transporters, along with the tools they provide to retrieve such information. Before conclusions, limitations and future perspectives are also discussed.
Collapse
Affiliation(s)
- Federico Alessandro Ruffinatti
- Turin Cell Physiology Laboratory (TCP-Lab), Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Giorgia Scarpellino
- Turin Cell Physiology Laboratory (TCP-Lab), Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Giorgia Chinigò
- Turin Cell Physiology Laboratory (TCP-Lab), Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Luca Visentin
- Turin Cell Physiology Laboratory (TCP-Lab), Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| | - Luca Munaron
- Turin Cell Physiology Laboratory (TCP-Lab), Department of Life Sciences and Systems Biology, University of Turin, Turin, Italy
| |
Collapse
|
17
|
Marini M, Titiz M, Souza Monteiro de Araújo D, Geppetti P, Nassini R, De Logu F. TRP Channels in Cancer: Signaling Mechanisms and Translational Approaches. Biomolecules 2023; 13:1557. [PMID: 37892239 PMCID: PMC10605459 DOI: 10.3390/biom13101557] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Ion channels play a crucial role in a wide range of biological processes, including cell cycle regulation and cancer progression. In particular, the transient receptor potential (TRP) family of channels has emerged as a promising therapeutic target due to its involvement in several stages of cancer development and dissemination. TRP channels are expressed in a large variety of cells and tissues, and by increasing cation intracellular concentration, they monitor mechanical, thermal, and chemical stimuli under physiological and pathological conditions. Some members of the TRP superfamily, namely vanilloid (TRPV), canonical (TRPC), melastatin (TRPM), and ankyrin (TRPA), have been investigated in different types of cancer, including breast, prostate, lung, and colorectal cancer. TRP channels are involved in processes such as cell proliferation, migration, invasion, angiogenesis, and drug resistance, all related to cancer progression. Some TRP channels have been mechanistically associated with the signaling of cancer pain. Understanding the cellular and molecular mechanisms by which TRP channels influence cancer provides new opportunities for the development of targeted therapeutic strategies. Selective inhibitors of TRP channels are under initial scrutiny in experimental animals as potential anti-cancer agents. In-depth knowledge of these channels and their regulatory mechanisms may lead to new therapeutic strategies for cancer treatment, providing new perspectives for the development of effective targeted therapies.
Collapse
Affiliation(s)
| | | | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, 50139 Florence, Italy; (M.M.); (M.T.); (D.S.M.d.A.); (P.G.); (F.D.L.)
| | | |
Collapse
|
18
|
Malcolm JR, Sajjaboontawee N, Yerlikaya S, Plunkett-Jones C, Boxall PJ, Brackenbury WJ. Voltage-gated sodium channels, sodium transport and progression of solid tumours. CURRENT TOPICS IN MEMBRANES 2023; 92:71-98. [PMID: 38007270 DOI: 10.1016/bs.ctm.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Sodium (Na+) concentration in solid tumours of different origin is highly dysregulated, and this corresponds to the aberrant expression of Na+ transporters. In particular, the α subunits of voltage gated Na+ channels (VGSCs) raise intracellular Na+ concentration ([Na+]i) in malignant cells, which influences the progression of solid tumours, predominantly driving cancer cells towards a more aggressive and metastatic phenotype. Conversely, re-expression of VGSC β subunits in cancer cells can either enhance tumour progression or promote anti-tumourigenic properties. Metastasis is the leading cause of cancer-related mortality, highlighting an important area of research which urgently requires improved therapeutic interventions. Here, we review the extent to which VGSC subunits are dysregulated in solid tumours, and consider the implications of such dysregulation on solid tumour progression. We discuss current understanding of VGSC-dependent mechanisms underlying increased invasive and metastatic potential of solid tumours, and how the complex relationship between the tumour microenvironment (TME) and VGSC expression may further drive tumour progression, in part due to the interplay of infiltrating immune cells, cancer-associated fibroblasts (CAFs) and insufficient supply of oxygen (hypoxia). Finally, we explore past and present clinical trials that investigate utilising existing VGSC modulators as potential pharmacological options to support adjuvant chemotherapies to prevent cancer recurrence. Such research demonstrates an exciting opportunity to repurpose therapeutics in order to improve the disease-free survival of patients with aggressive solid tumours.
Collapse
Affiliation(s)
- Jodie R Malcolm
- Department of Biology, University of York, Heslington, York, United Kingdom
| | - Nattanan Sajjaboontawee
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom
| | - Serife Yerlikaya
- Department of Biology, University of York, Heslington, York, United Kingdom; Istanbul Medipol University, Research Institute for Health Sciences and Technologies, Istanbul, Turkey
| | | | - Peter J Boxall
- Department of Biology, University of York, Heslington, York, United Kingdom; York and Scarborough Teaching Hospitals NHS Foundation Trust, York, United Kingdom
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York, United Kingdom; York Biomedical Research Institute, University of York, Heslington, York, United Kingdom.
| |
Collapse
|
19
|
Yang P, Feng J, Zhu Y, Hao Y. A Novel Cell Volume Sensor for Real-Time Analysis of Ca 2+-Activated K + Channel. ACS Biomater Sci Eng 2023; 9:5255-5259. [PMID: 37639544 DOI: 10.1021/acsbiomaterials.3c00771] [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] [Indexed: 08/31/2023]
Abstract
Potassium channels play a vital role in cell volume regulation. A cell volume sensor was constructed by integrating regulatory volume decrease (RVD) with quartz-crystal microbalance (QCM) for studying potassium channels and their expression. The sensor successfully monitored the K+ channel's activities during RVD by sensitive and noninvasive means. It showed that Ca2+ activated the K+ channel (KCa) and enhanced the RVD level. The inhibition of blockers on K+ channels exhibited an obvious difference in RVD level between normal and cancerous nasopharyngeal cells, suggesting that the KCa channel contributes a dominant role to the RVD function and provides an approach to identify the activation of various K+ channels.
Collapse
Affiliation(s)
- Peihui Yang
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, People's Republic of China
| | - Jingwei Feng
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, People's Republic of China
| | - Yeyan Zhu
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, People's Republic of China
| | - Yan Hao
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, People's Republic of China
| |
Collapse
|
20
|
Son GY, Tu NH, Santi MD, Lopez SL, Souza Bomfim GH, Vinu M, Zhou F, Chaloemtoem A, Alhariri R, Idaghdour Y, Khanna R, Ye Y, Lacruz RS. The Ca 2+ channel ORAI1 is a regulator of oral cancer growth and nociceptive pain. Sci Signal 2023; 16:eadf9535. [PMID: 37669398 PMCID: PMC10747475 DOI: 10.1126/scisignal.adf9535] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/15/2023] [Indexed: 09/07/2023]
Abstract
Oral cancer causes pain associated with cancer progression. We report here that the function of the Ca2+ channel ORAI1 is an important regulator of oral cancer pain. ORAI1 was highly expressed in tumor samples from patients with oral cancer, and ORAI1 activation caused sustained Ca2+ influx in human oral cancer cells. RNA-seq analysis showed that ORAI1 regulated many genes encoding oral cancer markers such as metalloproteases (MMPs) and pain modulators. Compared with control cells, oral cancer cells lacking ORAI1 formed smaller tumors that elicited decreased allodynia when inoculated into mouse paws. Exposure of trigeminal ganglia neurons to MMP1 evoked an increase in action potentials. These data demonstrate an important role of ORAI1 in oral cancer progression and pain, potentially by controlling MMP1 abundance.
Collapse
Affiliation(s)
- Ga-Yeon Son
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
| | - Nguyen Huu Tu
- NYU Dentistry Translational Research Center, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010
| | - Maria Daniela Santi
- NYU Dentistry Translational Research Center, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010
| | - Santiago Loya Lopez
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
- New York University Pain Research Center, New York University, New York, NY 10010
| | | | - Manikandan Vinu
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Fang Zhou
- Department of Pathology, New York University Langone Health, New York, NY 10010
| | - Ariya Chaloemtoem
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Rama Alhariri
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Youssef Idaghdour
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi, 129188, Saadiyat Island, Abu Dhabi, United Arab Emirates
| | - Rajesh Khanna
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
- New York University Pain Research Center, New York University, New York, NY 10010
| | - Yi Ye
- NYU Dentistry Translational Research Center, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY 10010
- New York University Pain Research Center, New York University, New York, NY 10010
| | - Rodrigo S. Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010
| |
Collapse
|
21
|
Zhang D, Yin G, Zheng S, Chen Q, Li Y. Construction of a prediction model for prognosis of bladder cancer based on the expression of ion channel-related genes. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:499-509. [PMID: 37643983 PMCID: PMC10495249 DOI: 10.3724/zdxbyxb-2023-0051] [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: 02/07/2023] [Accepted: 07/06/2023] [Indexed: 08/24/2023]
Abstract
OBJECTIVES To construct a prediction model for the prognosis of bladder cancer patients based on the expression of ion channel-related genes (ICRGs). METHODS ICRGs were obtained from the existing researches. The clinical information and the expression of ICRGs mRNA in breast cancer patients were obtained from the Cancer Genome Atlas database. Cox regression analysis, minimum absolute shrinkage and selection operator regression analysis were used to screen breast cancer prognosis related genes, which were verified by immunohistochemistry and qRT-PCR. The risk scoring equation for predicting the prognosis of patients with bladder cancer was constructed, and the patients were divided into high-risk group and low-risk group according to the median risk score. Immune cell infiltration was compared between the two groups. Kaplan-Meier survival curve and receiver operating characteristic (ROC) curve were used to evaluate the accuracy and clinical application value of the risk scoring equation. The factors related to the prognosis of bladder cancer patients were analyzed by univariate and multivariate Cox regression, and a nomogram for predicting the prognosis of bladder cancer patients was constructed. RESULTS By comparing the expression levels of ICRGs in bladder cancer tissues and normal bladder tissues, 73 differentially expressed ICRGs were dentified, of which 11 were related to the prognosis of bladder cancer patients. Kaplan-Meier survival curve suggested that the risk score based on these 11 genes was negatively correlated with the prognosis of patients. The area under the ROC curve of the risk score for predicting the prognosis of patients at 1, 3 and 5 year was 0.634, 0.665 and 0.712, respectively. Stratified analysis showed that the ICRGs-based risk score performed well in predicting the prognosis of patients with American Joint Committee on Cancer (AJCC) stage Ⅲ-Ⅳ bladder cancer (P<0.05), while it had a poor value in predicting the prognosis of patients with AJCC stage Ⅰ-Ⅱ (P>0.05). There were significant differences in the infiltration of plasma cells, activated natural killer cells, resting mast cells and M2 macrophages between the high-risk group and the low-risk group. Cox regression analysis showed that risk score, smoking, age and AJCC stage were independently associated with the prognosis of patients with bladder cancer (P<0.05). The nomogram constructed by combining risk score and clinical parameters has high accuracy in predicting the 1, 3 and 5 year overall survival rate of bladder cancer patients. CONCLUSIONS The study shows the potential value of ICRGs in the prognostic risk assessment of bladder cancer patients. The constructed prognostic nomogram based on ICRGs risk score has high accuracy in predicting the prognosis of bladder cancer patients.
Collapse
Affiliation(s)
- Dianfeng Zhang
- Department of Urology, Xuchang Central Hospital of Henan Province, Xuchang 461000, Henan Province, China.
| | - Guicao Yin
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Shengqi Zheng
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Qiu Chen
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China
| | - Yifan Li
- Department of Urology, the Affiliated Hospital of Yangzhou University, Yangzhou 225000, Jiangsu Province, China.
| |
Collapse
|
22
|
Grasso G, Colella F, Forciniti S, Onesto V, Iuele H, Siciliano AC, Carnevali F, Chandra A, Gigli G, Del Mercato LL. Fluorescent nano- and microparticles for sensing cellular microenvironment: past, present and future applications. NANOSCALE ADVANCES 2023; 5:4311-4336. [PMID: 37638162 PMCID: PMC10448310 DOI: 10.1039/d3na00218g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/13/2023] [Indexed: 08/29/2023]
Abstract
The tumor microenvironment (TME) demonstrates distinct hallmarks, including acidosis, hypoxia, reactive oxygen species (ROS) generation, and altered ion fluxes, which are crucial targets for early cancer biomarker detection, tumor diagnosis, and therapeutic strategies. Various imaging and sensing techniques have been developed and employed in both research and clinical settings to visualize and monitor cellular and TME dynamics. Among these, ratiometric fluorescence-based sensors have emerged as powerful analytical tools, providing precise and sensitive insights into TME and enabling real-time detection and tracking of dynamic changes. In this comprehensive review, we discuss the latest advancements in ratiometric fluorescent probes designed for the optical mapping of pH, oxygen, ROS, ions, and biomarkers within the TME. We elucidate their structural designs and sensing mechanisms as well as their applications in in vitro and in vivo detection. Furthermore, we explore integrated sensing platforms that reveal the spatiotemporal behavior of complex tumor cultures, highlighting the potential of high-resolution imaging techniques combined with computational methods. This review aims to provide a solid foundation for understanding the current state of the art and the future potential of fluorescent nano- and microparticles in the field of cellular microenvironment sensing.
Collapse
Affiliation(s)
- Giuliana Grasso
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Francesco Colella
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Stefania Forciniti
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Helena Iuele
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Anna Chiara Siciliano
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Federica Carnevali
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Anil Chandra
- Centre for Research in Pure and Applied Sciences, Jain (Deemed-to-be-university) Bangalore Karnataka 560078 India
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
- Department of Mathematics and Physics ''Ennio De Giorgi", University of Salento c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| | - Loretta L Del Mercato
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC) c/o Campus Ecotekne, via Monteroni 73100 Lecce Italy
| |
Collapse
|
23
|
Reisenauer KN, Aroujo J, Tao Y, Ranganathan S, Romo D, Taube JH. Therapeutic vulnerabilities of cancer stem cells and effects of natural products. Nat Prod Rep 2023; 40:1432-1456. [PMID: 37103550 PMCID: PMC10524555 DOI: 10.1039/d3np00002h] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Covering: 1995 to 2022Tumors possess both genetic and phenotypic heterogeneity leading to the survival of subpopulations post-treatment. The term cancer stem cells (CSCs) describes a subpopulation that is resistant to many types of chemotherapy and which also possess enhanced migratory and anchorage-independent growth capabilities. These cells are enriched in residual tumor material post-treatment and can serve as the seed for future tumor re-growth, at both primary and metastatic sites. Elimination of CSCs is a key goal in enhancing cancer treatment and may be aided by application of natural products in conjunction with conventional treatments. In this review, we highlight molecular features of CSCs and discuss synthesis, structure-activity relationships, derivatization, and effects of six natural products with anti-CSC activity.
Collapse
Affiliation(s)
| | - Jaquelin Aroujo
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | - Yongfeng Tao
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | | | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor Univesrity, Waco, TX, USA
| | - Joseph H Taube
- Department of Biology, Baylor University, Waco, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
24
|
Zheng Q, Wang Y, Zhao R, Han P, Zhao J, Li L, Zhou X, Li P, Mo Y, Pan X, Luo W, Zhou X. Inactivation of epithelial sodium ion channel molecules serves as effective diagnostic biomarkers in clear cell renal cell carcinoma. Genes Genomics 2023; 45:855-866. [PMID: 37133722 DOI: 10.1007/s13258-023-01376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 03/08/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Non-voltage-gated sodium channel, also known as the epithelial sodium channel (ENaC), formed by heteromeric complexes consisting of SCNN1A, SCNN1B, and SCNN1G, is responsible for maintaining sodium ion and body fluid homeostasis in epithelial cells. However, no systematic study of SCNN1 family members has been conducted in renal clear cell carcinoma (ccRCC) to date. OBJECTIVE To investigate the abnormal expression of SCNN1 family in ccRCC and its potential correlation with clinical parameters. METHODS The transcription and protein expression levels of SCNN1 family members in ccRCC were analyzed based on the TCGA database, and were confirmed by quantitative RT-PCR and immunohistochemical staining assays, respectively. The area under curve (AUC) was used to evaluate the diagnostic value of SCNN1 family members for ccRCC patients. RESULTS The mRNA and protein expression of SCNN1 family members was significantly downregulated in ccRCC compared with normal kidney tissues, which might be due to DNA hypermethylation in the promoter region. It is worth noting that the AUC of SCNN1A, SCNN1B, and SCNN1G were 0.965, 0.979, and 0.988 based on the TCGA database (p < 0.0001), respectively. The diagnostic value was even higher when combing these three members together (AUC = 0.997, p < 0.0001). Intriguingly, the mRNA level of SCNN1A was significantly lower in females compared with males, while SCNN1B and SCNN1G were increased with the progression of ccRCC and remarkably associated with a worse outcome for patients. CONCLUSION The aberrantly decrease of SCNN1 family members might serve as valuable biomarkers for the diagnosis of ccRCC.
Collapse
Affiliation(s)
- Qian Zheng
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Yifang Wang
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Ran Zhao
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Peipei Han
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Jun Zhao
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Limei Li
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Ping Li
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Yingxi Mo
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Xinli Pan
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, China
| | - Wenqi Luo
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, China.
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China.
| |
Collapse
|
25
|
Mlayah-Bellalouna S, Aissaoui-Zid D, Chantome A, Jebali J, Souid S, Ayedi E, Mejdoub H, Belghazi M, Marrakchi N, Essafi-Benkhadir K, Vandier C, Srairi-Abid N. Insights into the mechanisms governing P01 scorpion toxin effect against U87 glioblastoma cells oncogenesis. Front Pharmacol 2023; 14:1203247. [PMID: 37426811 PMCID: PMC10326281 DOI: 10.3389/fphar.2023.1203247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
The emerging concept of small conductance Ca2+-activated potassium channels (SKCa) as pharmacological target for cancer treatment has significantly increased in recent years. In this study, we isolated the P01 toxin from Androctonus australis (Aa) scorpion venom and investigated its effect on biological properties of glioblastoma U87, breast MDA-MB231 and colon adenocarcinoma LS174 cancer cell lines. Our results showed that P01 was active only on U87 glioblastoma cells. It inhibited their proliferation, adhesion and migration with IC50 values in the micromolar range. We have also shown that P01 reduced the amplitude of the currents recorded in HEK293 cells expressing SK2 channels with an IC50 value of 3 pM, while it had no effect on those expressing SK3 channels. The investigation of the SKCa channels expression pattern showed that SK2 transcripts were expressed differently in the three cancer cell lines. Particularly, we highlighted the presence of SK2 isoforms in U87 cells, which could explain and rely on the specific activity of P01 on this cell line. These experimental data highlighted the usefulness of scorpion peptides to decipher the role of SKCa channels in the tumorigenesis process, and develop potential therapeutic molecules targeting glioblastoma with high selectivity.
Collapse
Affiliation(s)
- Saoussen Mlayah-Bellalouna
- LR20IPT01 Biomolécules, Venins et Application Théranostique, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Dorra Aissaoui-Zid
- LR20IPT01 Biomolécules, Venins et Application Théranostique, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Aurelie Chantome
- N2C UMR 1069, Institut national de la santé et de la recherche médicale, University of Tours, Tours, France
| | - Jed Jebali
- LR20IPT01 Biomolécules, Venins et Application Théranostique, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Soumaya Souid
- LR16IPT04 Laboratoire d’Epidémiologie Moléculaire et Pathologie Expérimentale, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Emna Ayedi
- LR20IPT01 Biomolécules, Venins et Application Théranostique, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Hafedh Mejdoub
- USCR Séquenceur de Protéines, Faculté des Sciences de Sfax, Route de Soukra, Sfax, Tunisia
| | - Maya Belghazi
- Aix Marseille Université, CNRS, Plateforme Protéomique, IMM FR3479, Marseille Protéomique (MaP), Marseille, France
| | - Naziha Marrakchi
- LR20IPT01 Biomolécules, Venins et Application Théranostique, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Khadija Essafi-Benkhadir
- LR16IPT04 Laboratoire d’Epidémiologie Moléculaire et Pathologie Expérimentale, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| | - Christophe Vandier
- N2C UMR 1069, Institut national de la santé et de la recherche médicale, University of Tours, Tours, France
| | - Najet Srairi-Abid
- LR20IPT01 Biomolécules, Venins et Application Théranostique, Institut Pasteur de Tunis, Université de Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
26
|
Ramírez A, Ogonaga-Borja I, Acosta B, Chiliquinga AJ, de la Garza J, Gariglio P, Ocádiz-Delgado R, Bañuelos C, Camacho J. Ion Channels and Personalized Medicine in Gynecological Cancers. Pharmaceuticals (Basel) 2023; 16:800. [PMID: 37375748 DOI: 10.3390/ph16060800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Targeted therapy against cancer plays a key role in delivering safer and more efficient treatments. In the last decades, ion channels have been studied for their participation in oncogenic processes because their aberrant expression and/or function have been associated with different types of malignancies, including ovarian, cervical, and endometrial cancer. The altered expression or function of several ion channels have been associated with tumor aggressiveness, increased proliferation, migration, invasion, and metastasis of cancer cells and with poor prognosis in gynecological cancer patients. Most ion channels are integral membrane proteins easily accessible by drugs. Interestingly, a plethora of ion channel blockers have demonstrated anticancer activity. Consequently, some ion channels have been proposed as oncogenes, cancer, and prognostic biomarkers, as well as therapeutic targets in gynecological cancers. Here, we review the association of ion channels with the properties of cancer cells in these tumors, which makes them very promising candidates to be exploited in personalized medicine. The detailed analysis of the expression pattern and function of ion channels could help to improve the clinical outcomes in gynecological cancer patients.
Collapse
Affiliation(s)
- Ana Ramírez
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial Internacional, Tijuana 22390, Mexico
| | - Ingrid Ogonaga-Borja
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Brenda Acosta
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Andrea Jazmín Chiliquinga
- Grupo de Investigación de Ciencias en Red, Universidad Técnica del Norte, Av. 17 de Julio 5-21, Ibarra 100105, Ecuador
| | - Jaime de la Garza
- Unidad de Oncología Torácica y Laboratorio de Medicina Personalizada, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Ciudad de Mexico14080, Mexico
| | - Patricio Gariglio
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Rodolfo Ocádiz-Delgado
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Cecilia Bañuelos
- Programa Transdisciplinario en Desarrollo Científico y Tecnológico para la Sociedad, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av. Instituto Politécnico Nacional 2508, Ciudad de Mexico 07360, Mexico
| |
Collapse
|
27
|
Nasimi Shad A, Fanoodi A, Maharati A, Akhlaghipour I, Moghbeli M. Molecular mechanisms of microRNA-301a during tumor progression and metastasis. Pathol Res Pract 2023; 247:154538. [PMID: 37209575 DOI: 10.1016/j.prp.2023.154538] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023]
Abstract
Cancer is known as one of the leading causes of human deaths globally. Late diagnosis is considered as one of the main reasons for the high mortality rate among cancer patients. Therefore, the introduction of early diagnostic tumor markers can improve the efficiency of therapeutic modalities. MicroRNAs (miRNAs) have a key role in regulation of cell proliferation and apoptosis. MiRNAs deregulation has been frequently reported during tumor progressions. Since, miRNAs have a high stability in body fluids; they can be used as the reliable non-invasive tumor markers. Here, we discussed the role of miR-301a during tumor progressions. MiR-301a mainly functions as an oncogene via the modulation of transcription factors, autophagy, epithelial-mesenchymal transition (EMT), and signaling pathways. This review paves the way to suggest miR-301a as a non-invasive marker for the early tumor diagnosis. MiR-301a can also be suggested as an effective target in cancer therapy.
Collapse
Affiliation(s)
- Arya Nasimi Shad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Fanoodi
- Student Research Committee, Faculty of Medicine, Birjand University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
28
|
Rickard BP, Overchuk M, Chappell VA, Kemal Ruhi M, Sinawang PD, Nguyen Hoang TT, Akin D, Demirci U, Franco W, Fenton SE, Santos JH, Rizvi I. Methods to Evaluate Changes in Mitochondrial Structure and Function in Cancer. Cancers (Basel) 2023; 15:2564. [PMID: 37174030 PMCID: PMC10177605 DOI: 10.3390/cancers15092564] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Mitochondria are regulators of key cellular processes, including energy production and redox homeostasis. Mitochondrial dysfunction is associated with various human diseases, including cancer. Importantly, both structural and functional changes can alter mitochondrial function. Morphologic and quantifiable changes in mitochondria can affect their function and contribute to disease. Structural mitochondrial changes include alterations in cristae morphology, mitochondrial DNA integrity and quantity, and dynamics, such as fission and fusion. Functional parameters related to mitochondrial biology include the production of reactive oxygen species, bioenergetic capacity, calcium retention, and membrane potential. Although these parameters can occur independently of one another, changes in mitochondrial structure and function are often interrelated. Thus, evaluating changes in both mitochondrial structure and function is crucial to understanding the molecular events involved in disease onset and progression. This review focuses on the relationship between alterations in mitochondrial structure and function and cancer, with a particular emphasis on gynecologic malignancies. Selecting methods with tractable parameters may be critical to identifying and targeting mitochondria-related therapeutic options. Methods to measure changes in mitochondrial structure and function, with the associated benefits and limitations, are summarized.
Collapse
Affiliation(s)
- Brittany P. Rickard
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marta Overchuk
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27695, USA
| | - Vesna A. Chappell
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Mustafa Kemal Ruhi
- Institute of Biomedical Engineering, Boğaziçi University, Istanbul 34684, Turkey
| | - Prima Dewi Sinawang
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Tina Thuy Nguyen Hoang
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Demir Akin
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
- Center for Cancer Nanotechnology Excellence for Translational Diagnostics (CCNE-TD), School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Palo Alto, CA 94304, USA
| | - Walfre Franco
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Suzanne E. Fenton
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, and North Carolina State University, Raleigh, NC 27695, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Center for Environmental Health and Susceptibility, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
29
|
Kang Y, Xu L, Dong J, Huang Y, Yuan X, Li R, Chen L, Wang Z, Ji X. Calcium-based nanotechnology for cancer therapy. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
30
|
Saldías MP, Cruz P, Silva I, Orellana-Serradell O, Lavanderos B, Maureira D, Pinto R, Cerda O. The Cytoplasmic Region of SARAF Reduces Triple-Negative Breast Cancer Metastasis through the Regulation of Store-Operated Calcium Entry. Int J Mol Sci 2023; 24:ijms24065306. [PMID: 36982380 PMCID: PMC10049260 DOI: 10.3390/ijms24065306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Triple-negative breast cancer has a poor prognosis and is non-responsive to first-line therapies; hence, new therapeutic strategies are needed. Enhanced store-operated Ca2+ entry (SOCE) has been widely described as a contributing factor to tumorigenic behavior in several tumor types, particularly in breast cancer cells. SOCE-associated regulatory factor (SARAF) acts as an inhibitor of the SOCE response and, therefore, can be a potential antitumor factor. Herein, we generated a C-terminal SARAF fragment to evaluate the effect of overexpression of this peptide on the malignancy of triple-negative breast cancer cell lines. Using both in vitro and in vivo approaches, we showed that overexpression of the C-terminal SARAF fragment reduced proliferation, cell migration, and the invasion of murine and human breast cancer cells by decreasing the SOCE response. Our data suggest that regulating the activity of the SOCE response via SARAF activity might constitute the basis for further alternative therapeutic strategies for triple-negative breast cancer.
Collapse
Affiliation(s)
- María Paz Saldías
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Pablo Cruz
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Ian Silva
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Octavio Orellana-Serradell
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Boris Lavanderos
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Diego Maureira
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Raquel Pinto
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Oscar Cerda
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
- Correspondence: ; Tel.: +56-2-29786909
| |
Collapse
|
31
|
Li J, Liu X, Chen L, Zhu X, Yu Z, Dong L, Zhao X, Zou H, Wei Q, Feng Y, Zhu Y, Chai K, Li Q, Li M. Isopimaric acid, an ion channel regulator, regulates calcium and oxidative phosphorylation pathways to inhibit breast cancer proliferation and metastasis. Toxicol Appl Pharmacol 2023; 462:116415. [PMID: 36754215 DOI: 10.1016/j.taap.2023.116415] [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: 12/01/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Breast cancer is the globally most common malignant tumor and the biggest threat to women. Even though the diagnosis and treatment of breast cancer are progressing continually, a large number of breast cancer patients eventually develop a metastatic tumor, especially triple-negative breast cancer (TNBC). Recently, metal ion homeostasis and ion signaling pathway have become important targets for cancer therapy. In this study, We analyzed the effects and mechanisms of isopimaric acid (IPA), an ion channel regulator, on the proliferation and metastasis of breast cancer cells (4 T1, MDA-MB-231and MCF-7) by cell functional assay, flow cytometry, western blot, proteomics and other techniques in vitro and in vivo. Results found that IPA significantly inhibited the proliferation and metastasis of breast cancer cells (especially 4 T1). Further studies on the anti-tumor mechanism of IPA suggested that IPA might affect EMT and Wnt signaling pathways by targeting mitochondria oxidative phosphorylation and Ca2+ signaling pathways, and then inducing breast cancer cell cycle arrest and apoptosis. Our research reveals the therapeutic value of IPA in breast cancer and provides a theoretical basis for the new treatment of breast cancer.
Collapse
Affiliation(s)
- Jiacheng Li
- College of Life Science, Sichuan Normal University, Chengdu, Sichuan 610101, China; Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Xiaozhen Liu
- Department of Medical and Radiation Oncology, Linyi People's Hospital, Linyi 276000, China
| | - Lin Chen
- Sericultural Research Institute, Zhejiang, Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xinping Zhu
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Zhihong Yu
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Liyao Dong
- College of Life Science, Sichuan Normal University, Chengdu, Sichuan 610101, China; Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Xinyun Zhao
- College of Life Science, Sichuan Normal University, Chengdu, Sichuan 610101, China; Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Hongling Zou
- College of Life Science, Sichuan Normal University, Chengdu, Sichuan 610101, China; Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Qin Wei
- Key Laboratory of Fermentation Resources and Application in Universities of Sichuan Province, Yibin University, Yibin, Sichuan 644000, China
| | - Yongcai Feng
- Xujing (Hangzhou) Biotechnology Research Institute Co., Ltd., Hangzhou 310021, China
| | - Yongqiang Zhu
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Kequn Chai
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Qun Li
- College of Life Science, Sichuan Normal University, Chengdu, Sichuan 610101, China.
| | - Mingqian Li
- Zhejiang Provincial Key Laboratory of Cancer Prevention and Treatment Technology of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China.
| |
Collapse
|
32
|
Sakellakis M, Chalkias A. The Role οf Ion Channels in the Development and Progression of Prostate Cancer. Mol Diagn Ther 2023; 27:227-242. [PMID: 36600143 DOI: 10.1007/s40291-022-00636-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 01/06/2023]
Abstract
Ion channels have major regulatory functions in living cells. Apart from their role in ion transport, they are responsible for cellular electrogenesis and excitability, and may also regulate tissue homeostasis. Although cancer is not officially classified as a channelopathy, it has been increasingly recognized that ion channel aberrations play an important role in virtually all cancer types. Ion channels can exert pro-tumorigenic activities due to genetic or epigenetic alterations, or as a response to molecular signals, such as growth factors, hormones, etc. Increasing evidence suggests that ion channels and pumps play a critical role in the regulation of prostate cancer cell proliferation, apoptosis evasion, migration, epithelial-to-mesenchymal transition, and angiogenesis. There is also evidence suggesting that ion channels might play a role in treatment failure in patients with prostate cancer. Hence, they represent promising targets for diagnosis, staging, and treatment, and their effects may be of particular significance for specific patient populations, including those undergoing anesthesia and surgery. In this article, the role of major types of ion channels involved in the development and progression of prostate cancer are reviewed. Identifying the underlying molecular mechanisms of the pro-tumorigenic effects of ion channels may potentially inform the development of novel therapeutic strategies to counter this malignancy.
Collapse
Affiliation(s)
- Minas Sakellakis
- Hellenic GU Cancer Group, Athens, Greece. .,Department of Medical Oncology, Metropolitan Hospital, 9 Ethnarchou Makariou, 18547, Athens, Greece.
| | - Athanasios Chalkias
- Department of Anesthesiology, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Outcomes Research Consortium, Cleveland, OH, USA
| |
Collapse
|
33
|
Zheng Z, Song Y. Integrated analysis of the voltage-gated potassium channel-associated gene KCNH2 across cancers. BMC Bioinformatics 2023; 24:51. [PMID: 36792990 PMCID: PMC9933257 DOI: 10.1186/s12859-023-05180-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
KCNH2 encodes the human ether-a-go-go-related gene (hERG) potassium channel and is an important repolarization reserve for regulating cardiac electrical activity. Increasing evidence suggests that it is involved in the development of various tumours, yet a thorough analysis of the underlying process has not been performed. Here, we have comprehensively examined the role of KCNH2 in multiple cancers by assessing KCNH2 gene expression, diagnostic and prognostic value, genetic alterations, immune infiltration correlations, RNA modifications, mutations, clinical correlations, interacting proteins, and associated signalling pathways. KCNH2 is differentially expressed in over 30 cancers and has a high diagnostic value for 10 tumours. Survival analysis showed that high expression of KCNH2 was associated with a poor prognosis in glioblastoma multiforme (GBM) and hepatocellular carcinoma (LIHC). Mutations and RNA methylation modifications (especially m6A) of KCNH2 are associated with its expression in multiple tumours. KCNH2 expression is correlated with tumour mutation burden, microsatellite instability, neoantigen load, and mutant-allele tumour heterogeneity. In addition, KCNH2 expression is associated with the tumour immune microenvironment and its immunosuppressive phenotype. KEGG signalling pathway enrichment analysis revealed that KCNH2 and its interacting molecules are involved in a variety of pathways related to carcinogenesis and signal regulation, such as the PI3K/Akt and focal adhesion pathways. Overall, we found that KCNH2 and its interaction molecular are expected to be immune-related biomarkers for cancer diagnosis and prognosis evaluation, and are potential regulatory targets of singalling pathways for tumour development due to their significant role in cancers.
Collapse
Affiliation(s)
- Zequn Zheng
- Department of Cardiovascular Medicine, First Affiliated Hospital of Shantou University Medical College, Shantou University, Shantou, 515000, China
| | - Yongfei Song
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Lihuili Hospital Affiliated to Ningbo University, No. 378 Dongqing Road, Yinzhou District, Ningbo, 315000, Zhejiang, China.
- Department of Cardiovascular, Lihuili Hospital Facilitated to Ningbo University, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
34
|
He L, Zhang T, Zhu C, Yan T, Liu J. Crown Ether-Based Ion Transporters in Bilayer Membranes. Chemistry 2023; 29:e202300044. [PMID: 36723493 DOI: 10.1002/chem.202300044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/02/2023]
Abstract
Bilayer membranes that enhance the stability of the cell are essential for cell survival, separating and protecting the interior of the cell from its external environment. Membrane-based channel proteins are crucial for sustaining cellular activities. However, dysfunction of these proteins would induce serial channelopathies, which could be substituted by artificial ion channel analogs. Crown ethers (CEs) are widely studied in the area of artificial ion channels owing to their intrinsic host-guest interaction with different kinds of organic and inorganic ions. Other advantages such as lower price, chemical stability, and easier modification also make CE a research hotspot in the field of synthetic transmembrane nanopores. And numerous CEs-based membrane-active synthetic ion channels were designed and fabricated in the past decades. Herein, the recent progress of CEs-based synthetic ion transporters has been comprehensively summarized in this review, including their design principles, functional mechanisms, controllable properties, and biomedical applications. Furthermore, this review has been concluded by discussing the future opportunities and challenges facing this research field. It is anticipated that this review could offer some inspiration for the future fabrication of novel CEs-derived ion transporters with more advanced structures, properties, and practical applications.
Collapse
Affiliation(s)
- Lei He
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, 311121, Hangzhou, P. R. China
| | - Tianlong Zhang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, 311121, Hangzhou, P. R. China
| | - Canhong Zhu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, 311121, Hangzhou, P. R. China
| | - Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, 311121, Hangzhou, P. R. China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology of Zhejiang Province, Hangzhou Normal University, 311121, Hangzhou, P. R. China
| |
Collapse
|
35
|
Aquaporins and Ion Channels as Dual Targets in the Design of Novel Glioblastoma Therapeutics to Limit Invasiveness. Cancers (Basel) 2023; 15:cancers15030849. [PMID: 36765806 PMCID: PMC9913334 DOI: 10.3390/cancers15030849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
Current therapies for Glioblastoma multiforme (GBM) focus on eradicating primary tumors using radiotherapy, chemotherapy and surgical resection, but have limited success in controlling the invasive spread of glioma cells into a healthy brain, the major factor driving short survival times for patients post-diagnosis. Transcriptomic analyses of GBM biopsies reveal clusters of membrane signaling proteins that in combination serve as robust prognostic indicators, including aquaporins and ion channels, which are upregulated in GBM and implicated in enhanced glioblastoma motility. Accumulating evidence supports our proposal that the concurrent pharmacological targeting of selected subclasses of aquaporins and ion channels could impede glioblastoma invasiveness by impairing key cellular motility pathways. Optimal sets of channels to be selected as targets for combined therapies could be tailored to the GBM cancer subtype, taking advantage of differences in patterns of expression between channels that are characteristic of GBM subtypes, as well as distinguishing them from non-cancerous brain cells such as neurons and glia. Focusing agents on a unique channel fingerprint in GBM would further allow combined agents to be administered at near threshold doses, potentially reducing off-target toxicity. Adjunct therapies which confine GBM tumors to their primary sites during clinical treatments would offer profound advantages for treatment efficacy.
Collapse
|
36
|
Self-assembled Supramolecular Artificial Transmembrane Ion Channels: Recent Progress and Application. Chem Res Chin Univ 2023. [DOI: 10.1007/s40242-023-2337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
37
|
Zhang Y, Lu Y, Zhang P, Shang X, Li Y. Brain Injury Induced by Mercury in Common Carp: Novel Insight from Transcriptome Analysis. Biol Trace Elem Res 2023; 201:403-411. [PMID: 35233713 DOI: 10.1007/s12011-022-03161-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 02/12/2022] [Indexed: 01/11/2023]
Abstract
Mercury is a heavy metal which causes irreversible toxicity to fish and is detected in aquatic environment around the world. We aimed to explore the relative mechanism of mercury exposure on the brain injury. In this study, high-throughput sequencing RNA-Seq technology was carried out to analyze the changes of gene expression of brain tissues exposed to mercury. A large number of differentially expressed genes were identified. And 366 genes were up-regulated and 688 genes were down-regulated. Gene Ontology (GO) functional enrichment analysis showed that DNA-templated and transport were highly enriched in the biological process. Membrane, nucleus, and cytoplasm were highly enriched in the cellular component, and metal ion binding and DNA binding were highly enriched in molecular function. The differential genes were enriched in ferroptosis, necroptosis, calcium signaling pathway, and ion channels. Real-time quantitative reverse transcription PCR (qRT-PCR) results demonstrated the selected genes exhibited the same trends with the RNA-Seq results, which indicates the transcriptome sequencing data is reliable. Our results may provide an insightful view for the toxic effects of mercury on brain injury of common carp.
Collapse
Affiliation(s)
- Yue Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, China
| | - Yuting Lu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, China
| | - Peijun Zhang
- Health Monitoring and Inspection Center of Jilin Province, Changchun, 130062, China
| | - Xinchi Shang
- Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Rd 43 Songfa, Daoli District, Harbin, 150070, China
- Key Laboratory of Cold Water Fish Germplasm Resources and Multiplication and Cultivation of Heilongjiang Province, Harbin, 150070, Heilongjiang, China
| | - Yuehong Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
38
|
Haustrate A, Mihalache A, Cordier C, Gosset P, Prevarskaya N, Lehen’kyi V. A Novel Anti-TRPV6 Antibody and Its Application in Cancer Diagnosis In Vitro. Int J Mol Sci 2022; 24:ijms24010419. [PMID: 36613866 PMCID: PMC9820453 DOI: 10.3390/ijms24010419] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Though the first discovery of TRPV6 channel expression in various tissues took place in the early 2000s, reliable tools for its protein detection in various cells and tissues are still missing. Here we show the generation and validation of rabbit polyclonal anti-TRPV6 channel antibodies (rb79-82) against four epitopes of 15 amino acids. Among them, only one antibody, rb79, was capable of detecting the full-length glycosylated form of the TRPV6 channel at around 100 kDa. The generated antibody was shown to be suitable for all in vitro applications, such as immunoblotting, immunoprecipitation, immunocytochemistry, immunofluorescence, etc. One of the most important applications is immunohistochemistry using the paraffin-embedded sections from cancer resection specimens. Using prostate cancer resection specimens, we have confirmed the absence of the TRPV6 protein in both healthy and benign hyperplasia, as well as its expression and correlation to the prostate cancer grades. Thus, the generated rabbit polyclonal anti-TRPV6 channel antibody rb79 is suitable for all in vitro diagnostic applications and particularly for the diagnosis in clinics using paraffin-embedded sections from patients suffering from various diseases and disorders involving the TRPV6 channel.
Collapse
Affiliation(s)
- Aurélien Haustrate
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d’Ascq, France
- FONDATION ARC, 9 rue Guy Môquet, 94830 Villejuif, France
| | - Adriana Mihalache
- Service d’Anatomie et de Cytologie Pathologiques, Groupement des Hôpitaux de l’Institut Catholique de Lille (GHICL), 59000 Lille, France
| | - Clément Cordier
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d’Ascq, France
| | - Pierre Gosset
- Service d’Anatomie et de Cytologie Pathologiques, Groupement des Hôpitaux de l’Institut Catholique de Lille (GHICL), 59000 Lille, France
| | - Natalia Prevarskaya
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d’Ascq, France
| | - V’yacheslav Lehen’kyi
- Laboratory of Cell Physiology, INSERM U1003, Laboratory of Excellence Ion Channels Science and Therapeutics, Department of Biology, Faculty of Science and Technologies, University of Lille, 59650 Villeneuve d’Ascq, France
- FONDATION ARC, 9 rue Guy Môquet, 94830 Villejuif, France
- Correspondence: ; Tel.: +33-3-2033-7078
| |
Collapse
|
39
|
Tiffner A, Hopl V, Derler I. CRAC and SK Channels: Their Molecular Mechanisms Associated with Cancer Cell Development. Cancers (Basel) 2022; 15:101. [PMID: 36612099 PMCID: PMC9817886 DOI: 10.3390/cancers15010101] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca2+-permeating and Ca2+-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca2+ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
Collapse
Affiliation(s)
- Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| | | | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria
| |
Collapse
|
40
|
Paul D, Nedelcu AM. The underexplored links between cancer and the internal body climate: Implications for cancer prevention and treatment. Front Oncol 2022; 12:1040034. [PMID: 36620608 PMCID: PMC9815514 DOI: 10.3389/fonc.2022.1040034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
In order to effectively manage and cure cancer we should move beyond the general view of cancer as a random process of genetic alterations leading to uncontrolled cell proliferation or simply a predictable evolutionary process involving selection for traits that increase cell fitness. In our view, cancer is a systemic disease that involves multiple interactions not only among cells within tumors or between tumors and surrounding tissues but also with the entire organism and its internal "milieu". We define the internal body climate as an emergent property resulting from spatial and temporal interactions among internal components themselves and with the external environment. The body climate itself can either prevent, promote or support cancer initiation and progression (top-down effect; i.e., body climate-induced effects on cancer), as well as be perturbed by cancer (bottom-up effect; i.e., cancer-induced body climate changes) to further favor cancer progression and spread. This positive feedback loop can move the system towards a "cancerized" organism and ultimately results in its demise. In our view, cancer not only affects the entire system; it is a reflection of an imbalance of the entire system. This model provides an integrated framework to study all aspects of cancer as a systemic disease, and also highlights unexplored links that can be altered to both prevent body climate changes that favor cancer initiation, progression and dissemination as well as manipulate or restore the body internal climate to hinder the success of cancer inception, progression and metastasis or improve therapy outcomes. To do so, we need to (i) identify cancer-relevant factors that affect specific climate components, (ii) develop 'body climate biomarkers', (iii) define 'body climate scores', and (iv) develop strategies to prevent climate changes, stop or slow the changes, or even revert the changes (climate restoration).
Collapse
Affiliation(s)
- Doru Paul
- Weill Cornell Medicine, New York, NY, United States
| | - Aurora M. Nedelcu
- Biology Department, University of New Brunswick, Fredericton, NB, Canada
| |
Collapse
|
41
|
TRPV4 Promotes Metastasis in Melanoma by Regulating Cell Motility through Cytoskeletal Rearrangement. Int J Mol Sci 2022; 23:ijms232315155. [PMID: 36499486 PMCID: PMC9737014 DOI: 10.3390/ijms232315155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
The abnormal expression of Transient Receptor Potential cation channel subfamily V member 4 (TRPV4) is closely related to the progression of multiple tumors. In addition, TRPV4 is increasingly being considered a potential target for cancer therapy, especially in tumor metastasis prevention. However, the biological correlation between TRPV4 and tumor metastasis, as well as the specific role of TRPV4 in malignant melanoma metastasis, is poorly understood. In this study, we aimed to examine the role of TRPV4 in melanoma metastasis through experiments and clinical data analysis, and the underlying anticancer mechanism of Baicalin, a natural compound, and its inhibitory effect on TRPV4 with in vivo and in vitro experiments. Our findings suggested that TRPV4 promotes metastasis in melanoma by regulating cell motility via rearranging the cytoskeletal, and Baicalin can inhibit cancer metastasis, whose mechanisms reverse the recruitment of activated cofilin to leading-edge protrusion and the increasing phosphorylation level of cortactin, which is provoked by TRPV4 activation.
Collapse
|
42
|
Xue C, Gao Y, Li X, Zhang M, Yang Y, Han Q, Sun Z, Bai C, Zhao RC. Mesenchymal stem cells derived from adipose accelerate the progression of colon cancer by inducing a MT-CAFs phenotype via TRPC3/NF-KB axis. STEM CELL RESEARCH & THERAPY 2022; 13:335. [PMID: 35870973 PMCID: PMC9308187 DOI: 10.1186/s13287-022-03017-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/10/2022] [Indexed: 12/02/2022]
Abstract
Background There is increasing evidence that mesenchymal stem cells (MSCs) help shape the tumor microenvironment and promote tumor progression, and ion channels might play a critical role in this process. The objective of the present study was to explore the function and mechanism of MT-CAFs on progression of colon cancer. Methods Here, a gene chip was used for a general analysis of gene expression changes in MSC-transformed CAF cells (MT-CAFs). Bioinformatic tool and western blot screened out the ion channel protein TRPC3 with significantly increased expression, and identify the function through two-photon microscope. The progression of cancer was detected via MTS, transwell and Wound Healing. ELISA deected the secretion of inflammation factors. TRPC3/NF-KB axis was identified by western blot and immunofluorescence. Results TRPC3 can caused calcium influx, which further activated the NF-KB signaling pathway. Knockdown or inhibition of TRPC3 in MSCs significantly reduced the activation of NF-KB, and decreased the growth, migration, and invasion of MT-CAFs. After TRPC3 knockdown, the ability of MT- CAFs to promote tumor migration and invasion was impaired. Conversely, the upregulation of TRPC3 expression in MT-CAFs had the opposite effect. In vivo, TRPC3 expressed on MSCs also contributed to the tumorigenesis and progression of cancer cells. In addition, the Oncomine and GEPIA databases showed that TRPC3 expression is higher in colon cancer tissues compared with normal colon tissues, and was positively correlated with the expression of the CAF genes alpha-smooth muscle (α-SMA/ACTA2) and fibroblast activation protein Alpha. The disease-free survival of patients with positive TRPC3 expression in MSCs was significantly shorter than those with negative expression. Conclusions These results indicate that TRPC3 expressed on MT-CAFs plays a critical role in tumor progression via the NF-KB signaling pathway, and is correlated with poor prognosis in colon cancer patients. Therefore, TRPC3 may be a novel therapeutic target for the treatment of colon cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03017-5.
Collapse
|
43
|
TRPV3 promotes the angiogenesis through HIF-1α-VEGF signaling pathway in A549 cells. Acta Histochem 2022; 124:151955. [DOI: 10.1016/j.acthis.2022.151955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022]
|
44
|
Dewdney B, Ursich L, Fletcher EV, Johns TG. Anoctamins and Calcium Signalling: An Obstacle to EGFR Targeted Therapy in Glioblastoma? Cancers (Basel) 2022; 14:cancers14235932. [PMID: 36497413 PMCID: PMC9740065 DOI: 10.3390/cancers14235932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Glioblastoma is the most common form of high-grade glioma in adults and has a poor survival rate with very limited treatment options. There have been no significant advancements in glioblastoma treatment in over 30 years. Epidermal growth factor receptor is upregulated in most glioblastoma tumours and, therefore, has been a drug target in recent targeted therapy clinical trials. However, while many inhibitors and antibodies for epidermal growth factor receptor have demonstrated promising anti-tumour effects in preclinical models, they have failed to improve outcomes for glioblastoma patients in clinical trials. This is likely due to the highly plastic nature of glioblastoma tumours, which results in therapeutic resistance. Ion channels are instrumental in the development of many cancers and may regulate cellular plasticity in glioblastoma. This review will explore the potential involvement of a class of calcium-activated chloride channels called anoctamins in brain cancer. We will also discuss the integrated role of calcium channels and anoctamins in regulating calcium-mediated signalling pathways, such as epidermal growth factor signalling, to promote brain cancer cell growth and migration.
Collapse
Affiliation(s)
- Brittany Dewdney
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6319-1023
| | - Lauren Ursich
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| | - Terrance G. Johns
- Cancer Centre, Telethon Kids Institute, Perth, WA 6009, Australia
- Centre for Child Health Research, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
45
|
Bioelectronic medicines: Therapeutic potential and advancements in next-generation cancer therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188808. [DOI: 10.1016/j.bbcan.2022.188808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 11/22/2022]
|
46
|
Abstract
Diverse ion channels have dysregulated functional expression in the tumor microenvironment (TME). In this issue of Cell Metabolism, Chen et al. reveal that high intratumoral K+ ions restrict the plasticity of tumor-associated macrophages (TAMs). Inhibition of the Kir2.1 potassium channel induced metabolic reprogramming and repolarization of pro-tumor M2-TAMs to tumoricidal M1-like states.
Collapse
Affiliation(s)
- Umar Al-Sheikh
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China.
| | - Lijun Kang
- Department of Neurobiology and Department of Neurology of the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
47
|
Han Y, Shi Y, Chen B, Wang J, Liu Y, Sheng S, Fu Z, Shen C, Wang X, Yin S, Li H. An ion-channel-gene-based prediction model for head and neck squamous cell carcinoma: Prognostic assessment and treatment guidance. Front Immunol 2022; 13:961695. [PMID: 36389709 PMCID: PMC9650652 DOI: 10.3389/fimmu.2022.961695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/12/2022] [Indexed: 09/18/2023] Open
Abstract
PURPOSE Head and neck squamous cell carcinoma (HNSCC) is a very diverse malignancy with a poor prognosis. The purpose of this study was to develop a new signature based on 12 ion channel genes to predict the outcome and immune status of HNSCC patients. METHODS Clinicopathological information and gene sequencing data of HNSCC patients were generated from the Cancer Genome Atlas and Gene Expression Omnibus databases. A set of 323 ion channel genes was obtained from the HUGO Gene Nomenclature Committee database and literature review. Using univariate Cox regression analysis, the ion channel genes related to HNSCC prognosis were identified. A prognostic signature and nomogram were then created using machine learning methods. Kaplan-Meier analysis was used to explore the relevance of the risk scores and overall survival (OS). We also investigated the association between risk scores, tumor immune infiltration, and gene mutational status. Finally, we detected the expression levels of the signature genes by quantitative real-time polymerase chain reaction, western blotting, and immunohistochemistry. RESULTS We separated the patients into high- and low-risk groups according to the risk scores computed based on these 12 ion channel genes, and the OS of the low-risk group was significantly longer (p<0.001). The area under the curve for predicting 3-year survival was 0.729. Univariate and multivariate analyses showed that the 12-ion-channel-gene risk model was an independent prognostic factor. We also developed a nomogram model based on risk scores and clinicopathological variables to forecast outcomes. Furthermore, immune cell infiltration, gene mutation status, immunotherapy response, and chemotherapeutic treatment sensitivity were all linked to risk scores. Moreover, high expression levels of ANO1, AQP9, and BEST2 were detected in HNSCC tissues, whereas AQP5, SCNN1G, and SCN4A expression was low in HNSCC tissues, as determined by experiments. CONCLUSION The 12-ion-channel-gene prognostic signatures have been demonstrated to be highly efficient in predicting the prognosis, immune microenvironment, gene mutation status, immunotherapy response, and chemotherapeutic sensitivity of HNSCC patients.
Collapse
Affiliation(s)
- Yanxun Han
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
| | - Yangyang Shi
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Bangjie Chen
- Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | | | - Yuchen Liu
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Medical University, Hefei, Anhui, China
| | | | - Ziyue Fu
- Anhui Medical University, Hefei, Anhui, China
| | | | - Xinyi Wang
- Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Siyue Yin
- Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Haiwen Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
48
|
Boyle Y, Johns TG, Fletcher EV. Potassium Ion Channels in Malignant Central Nervous System Cancers. Cancers (Basel) 2022; 14:cancers14194767. [PMID: 36230692 PMCID: PMC9563970 DOI: 10.3390/cancers14194767] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Malignant central nervous system (CNS) cancers are among the most difficult to treat, with low rates of survival and a high likelihood of recurrence. This is primarily due to their location within the CNS, hindering adequate drug delivery and tumour access via surgery. Furthermore, CNS cancer cells are highly plastic, an adaptive property that enables them to bypass targeted treatment strategies and develop drug resistance. Potassium ion channels have long been implicated in the progression of many cancers due to their integral role in several hallmarks of the disease. Here, we will explore this relationship further, with a focus on malignant CNS cancers, including high-grade glioma (HGG). HGG is the most lethal form of primary brain tumour in adults, with the majority of patient mortality attributed to drug-resistant secondary tumours. Hence, targeting proteins that are integral to cellular plasticity could reduce tumour recurrence, improving survival. This review summarises the role of potassium ion channels in malignant CNS cancers, specifically how they contribute to proliferation, invasion, metastasis, angiogenesis, and plasticity. We will also explore how specific modulation of these proteins may provide a novel way to overcome drug resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Yasmin Boyle
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
- Correspondence:
| | - Terrance G. Johns
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| |
Collapse
|
49
|
Ion Channels in Endometrial Cancer. Cancers (Basel) 2022; 14:cancers14194733. [PMID: 36230654 PMCID: PMC9564232 DOI: 10.3390/cancers14194733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Uterine or endometrial cancer is one of the most common types of cancer among the female population. Different alterations of molecules are related to many types of cancer. Some molecules called ion channels have been described as involved in the development of cancer, including endometrial cancer. We review the scientific evidence about the involvement of the ion channels in endometrial cancer and how some treatments can be developed with these molecules as a target. Even though they are involved in the progression of endometrial cancer, since they are present throughout the whole body, some possible treatments based on these could be studied. Abstract Uterine or endometrial cancer (EC) is the sixth most common neoplasia among women worldwide. Cancer can originate from a myriad of causes, and increasing evidence suggests that ion channels (IC) play an important role in the process of carcinogenesis, taking part in many pathways such as self-sufficiency in growth signals, proliferation, evasion of programmed cell death (apoptosis), angiogenesis, cell differentiation, migration, adhesion, and metastasis. Hormones and growth factors are well-known to be involved in the development and/or progression of many cancers and can also regulate some ion channels and pumps. Since the endometrium is responsive and regulated by these factors, the ICs could make an important contribution to the development and progression of endometrial cancer. In this review, we explore what is beyond (ion) flow regulation by investigating the role of the main families of ICs in EC, including as possible targets for EC treatment.
Collapse
|
50
|
Pellegrino M, Ricci E, Ceraldi R, Nigro A, Bonofiglio D, Lanzino M, Morelli C. From HDAC to Voltage-Gated Ion Channels: What's Next? The Long Road of Antiepileptic Drugs Repositioning in Cancer. Cancers (Basel) 2022; 14:cancers14184401. [PMID: 36139561 PMCID: PMC9497059 DOI: 10.3390/cancers14184401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although in the last decades the clinical outcome of cancer patients considerably improved, the major drawbacks still associated with chemotherapy are the unwanted side effects and the development of drug resistance. Therefore, a continuous effort in trying to discover new tumor markers, possibly of diagnostic, prognostic and therapeutic value, is being made. This review is aimed at highlighting the anti-tumor activity that several antiepileptic drugs (AEDs) exert in breast, prostate and other types of cancers, mainly focusing on their ability to block the voltage-gated Na+ and Ca++ channels, as well as to inhibit the activity of histone deacetylases (HDACs), all well-documented tumor markers and/or molecular targets. The existence of additional AEDs molecular targets is highly suspected. Therefore, the repurposing of already available drugs as adjuvants in cancer treatment would have several advantages, such as reductions in dose-related toxicity CVs will be sent in a separate mail to the indicated address of combined treatments, lower production costs, and faster approval for clinical use. Abstract Cancer is a major health burden worldwide. Although the plethora of molecular targets identified in the last decades and the deriving developed treatments, which significantly improved patients’ outcome, the occurrence of resistance to therapies remains the major cause of relapse and mortality. Thus, efforts in identifying new markers to be exploited as molecular targets in cancer therapy are needed. This review will first give a glance on the diagnostic and therapeutic significance of histone deacetylase (HDAC) and voltage gated ion channels (VGICs) in cancer. Nevertheless, HDAC and VGICs have also been reported as molecular targets through which antiepileptic drugs (AEDs) seem to exert their anticancer activity. This should be claimed as a great advantage. Indeed, due to the slowness of drug approval procedures, the attempt to turn to off-label use of already approved medicines would be highly preferable. Therefore, an updated and accurate overview of both preclinical and clinical data of commonly prescribed AEDs (mainly valproic acid, lamotrigine, carbamazepine, phenytoin and gabapentin) in breast, prostate, brain and other cancers will follow. Finally, a glance at the emerging attempt to administer AEDs by means of opportunely designed drug delivery systems (DDSs), so to limit toxicity and improve bioavailability, is also given.
Collapse
Affiliation(s)
| | | | | | | | | | - Marilena Lanzino
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| | - Catia Morelli
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| |
Collapse
|