1
|
den Hollander LS, Zweemer AJM, Béquignon OJM, Hammerl DM, Bleijs BTM, Veenhuizen M, Lantsheer WJF, Chau B, van Westen GJP, IJzerman AP, Heitman LH. CC chemokine receptor 2 is allosterically modulated by sodium ions and amiloride derivatives through a distinct sodium ion binding site. Biochem Pharmacol 2024; 229:116464. [PMID: 39111604 DOI: 10.1016/j.bcp.2024.116464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024]
Abstract
CC chemokine receptor 2 and CCL2 are highly involved in cancer growth and metastasis, and immune escape. Raised sodium ion concentrations in solid tumours have also been correlated to metastasis and immune modulation. Sodium ions can modulate class A G protein-coupled receptors through the sodium ion binding site characterized by a highly conserved aspartic acid residue (D2.50), also present in CCR2. Hence, we further explored this binding site in CCR2 by radioligand binding studies and mutagenesis. Modulation of three distinctly binding radioligands by sodium ions and amiloride derivates was investigated. Sodium ions were observed to be relatively weak modulators of antagonist binding, but substantially increased 125I-CCL2 dissociation from CCR2. 6-Substituted Hexamethylene Amiloride (HMA) modulated all tested radioligands. Induced-fit docking of HMA in the presumed sodium ion binding site of CCR2 confirmed its binding site. Finally, investigation of (cancer-associated) mutations in the sodium ion binding site showed a markedly decreased expression compared to wild type. Only two mutants, G123A3.35 and G127K3.39, were able to be bound by [3H]INCB3344 and [3H]CCR2-RA-[R]. Thus, mutagenesis showed that the sodium ion binding site residues, which are distinct from other class A GPCRs and related to chemokine receptor evolution, are crucial for receptor integrity. Moreover, the tested mutations appeared to have no effect on modulation observed by HMA or a minor effect on sodium chloride modulation on the tested radioligands. All in all, these results invite further exploration of the CCR2 sodium ion binding site in (cancer) biology, and potentially as a third druggable binding site.
Collapse
Affiliation(s)
- Lisa S den Hollander
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Annelien J M Zweemer
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Olivier J M Béquignon
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Dora M Hammerl
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Bente T M Bleijs
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Margo Veenhuizen
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Wernard J F Lantsheer
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Bobby Chau
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Gerard J P van Westen
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Adriaan P IJzerman
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands
| | - Laura H Heitman
- Leiden Academic Centre for Drug Research, Division of Drug Discovery and Safety, Leiden, the Netherlands; Oncode Institute, the Netherlands.
| |
Collapse
|
2
|
Wang J, Liao Y, Deng M, Wu X, Wang X, Li J. 24-Hour Urinary Sodium Excretion Is Associated With Increased Risk of Pancreatic Cancer: A Prospective Cohort Study. Clin Transl Gastroenterol 2024; 15:e1. [PMID: 38976319 PMCID: PMC11500774 DOI: 10.14309/ctg.0000000000000741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024] Open
Abstract
INTRODUCTION This study builds on previous research and its limitations, which indicate the need for further investigation in prospective cohorts. Our aim was to explore the association between estimated 24-hour urinary sodium excretion (indicative of daily sodium consumption) and the occurrence of pancreatic cancer in the UK Biobank's large prospective cohort. METHODS Using the INTERSALT equation, the study computed estimated 24-hour urinary sodium excretion by analyzing the baseline spot urine sodium measurements of 434,372 individuals enrolled in the UK Biobank. Pancreatic cancer cases were identified through UK cancer registries. Adjusted Cox proportional hazards models were used to evaluate hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between estimated 24-hour urinary sodium excretion and the risk of pancreatic cancer. RESULTS Over a median follow-up period of 13.8 years, 1,765 cases of pancreatic cancer were detected. The multivariable adjusted Cox model showed that each 1-gram rise in estimated 24-hour urinary sodium excretion corresponded to a 1.12 HR for incident pancreatic cancer (95% CI: 1.03, 1.22). The estimated HR for 24-hour urinary sodium excretion in binary form was 1.23 (95% CI: 1.05, 1.44). Compared with the lowest group, the group with the highest estimated 24-hour urinary sodium excretion exhibited an HR of 1.38 (95% CI: 1.21, 1.58). DISCUSSION These results propose an association between elevated sodium consumption and a heightened risk of pancreatic cancer. Further validation and exploration of potential mechanisms are warranted.
Collapse
Affiliation(s)
- Jiayi Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yangjie Liao
- Department of Gastroenterology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde, China
| | - Minzi Deng
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xing Wu
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jingbo Li
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Scirgolea C, Sottile R, De Luca M, Susana A, Carnevale S, Puccio S, Ferrari V, Lise V, Contarini G, Scarpa A, Scamardella E, Feno S, Camisaschi C, De Simone G, Basso G, Giuliano D, Mazza EMC, Gattinoni L, Roychoudhuri R, Voulaz E, Di Mitri D, Simonelli M, Losurdo A, Pozzi D, Tsui C, Kallies A, Timo S, Martano G, Barberis E, Manfredi M, Rescigno M, Jaillon S, Lugli E. NaCl enhances CD8 + T cell effector functions in cancer immunotherapy. Nat Immunol 2024; 25:1845-1857. [PMID: 39198631 DOI: 10.1038/s41590-024-01923-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/10/2024] [Indexed: 09/01/2024]
Abstract
CD8+ T cells control tumors but inevitably become dysfunctional in the tumor microenvironment. Here, we show that sodium chloride (NaCl) counteracts T cell dysfunction to promote cancer regression. NaCl supplementation during CD8+ T cell culture induced effector differentiation, IFN-γ production and cytotoxicity while maintaining the gene networks responsible for stem-like plasticity. Accordingly, adoptive transfer of tumor-specific T cells resulted in superior anti-tumor immunity in a humanized mouse model. In mice, a high-salt diet reduced the growth of experimental tumors in a CD8+ T cell-dependent manner by inhibiting terminal differentiation and enhancing the effector potency of CD8+ T cells. Mechanistically, NaCl enhanced glutamine consumption, which was critical for transcriptional, epigenetic and functional reprogramming. In humans, CD8+ T cells undergoing antigen recognition in tumors and predicting favorable responses to checkpoint blockade immunotherapy resembled those induced by NaCl. Thus, NaCl metabolism is a regulator of CD8+ T cell effector function, with potential implications for cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Simone Puccio
- IRCCS Humanitas Research Hospital, Milan, Italy
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, Milan, Italy
| | | | | | | | | | | | - Simona Feno
- IRCCS Humanitas Research Hospital, Milan, Italy
| | | | | | | | | | | | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
- University of Regensburg, Regensburg, Germany
- Center for Immunomedicine in Transplantation and Oncology (CITO), University Hospital Regensburg, Regensburg, Germany
| | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Cambridge, UK
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Emanuele Voulaz
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Division of Thoracic, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Matteo Simonelli
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | | | - Davide Pozzi
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Carlson Tsui
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity and Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria, Australia
| | - Sara Timo
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Martano
- IRCCS Humanitas Research Hospital, Milan, Italy
- Institute of Neuroscience, National Research Council of Italy (CNR) c/o Humanitas Mirasole S.p.A, Milan, Italy
| | - Elettra Barberis
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Maria Rescigno
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Sebastien Jaillon
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Enrico Lugli
- IRCCS Humanitas Research Hospital, Milan, Italy.
| |
Collapse
|
4
|
Kim JS, Kehrl JH. Inhibition of WNK Kinases in NK Cells Disrupts Cellular Osmoregulation and Control of Tumor Metastasis. J Innate Immun 2024; 16:451-469. [PMID: 39265537 PMCID: PMC11521464 DOI: 10.1159/000540744] [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: 11/22/2023] [Accepted: 08/01/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION The serine/threonine with-no-lysine (WNK) kinase family function in blood pressure control, electrolyte homeostasis, and cellular osmoregulation. These kinases and their downstream effectors are considered promising therapeutic targets in hypertension and stroke. However, the role of WNK kinases in immune cells remains poorly understood. METHODS Using the small-molecule WNK kinase inhibitors WNK463 and WNK-IN-11, we investigated how WNK kinase inhibition affects natural killer (NK) cell physiology. RESULTS WNK kinase inhibition with WNK463 or WNK-IN-11 significantly decreased IL-2-activated NK cell volume, motility, and cytolytic activity. Treatment of NK cells with these inhibitors induced autophagy by activating AMPK and inhibiting mTOR signaling. Moreover, WNK kinase inhibition increased phosphorylation of Akt and c-Myc by misaligning activity of activating kinases and inhibitory phosphatases. Treatment of tumor-bearing mice with WNK463 impaired tumor metastasis control by adoptively transferred NK cells. CONCLUSION The catalytic activity of WNK kinases has a critical role of multiple aspects of NK cell physiology and their pharmacologic inhibition negatively impacts NK cell function.
Collapse
Affiliation(s)
- Ji Sung Kim
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Giammello F, Biella C, Priori EC, Filippo MADS, Leone R, D'Ambrosio F, Paterno' M, Cassioli G, Minetti A, Macchi F, Spalletti C, Morella I, Ruberti C, Tremonti B, Barbieri F, Lombardi G, Brambilla R, Florio T, Galli R, Rossi P, Brandalise F. Modulating voltage-gated sodium channels to enhance differentiation and sensitize glioblastoma cells to chemotherapy. Cell Commun Signal 2024; 22:434. [PMID: 39251990 PMCID: PMC11382371 DOI: 10.1186/s12964-024-01819-z] [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: 04/05/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) stands as the most prevalent and aggressive form of adult gliomas. Despite the implementation of intensive therapeutic approaches involving surgery, radiation, and chemotherapy, Glioblastoma Stem Cells contribute to tumor recurrence and poor prognosis. The induction of Glioblastoma Stem Cells differentiation by manipulating the transcriptional machinery has emerged as a promising strategy for GBM treatment. Here, we explored an innovative approach by investigating the role of the depolarized resting membrane potential (RMP) observed in patient-derived GBM sphereforming cell (GSCs), which allows them to maintain a stemness profile when they reside in the G0 phase of the cell cycle. METHODS We conducted molecular biology and electrophysiological experiments, both in vitro and in vivo, to examine the functional expression of the voltage-gated sodium channel (Nav) in GSCs, particularly focusing on its cell cycle-dependent functional expression. Nav activity was pharmacologically manipulated, and its effects on GSCs behavior were assessed by live imaging cell cycle analysis, self-renewal assays, and chemosensitivity assays. Mechanistic insights into the role of Nav in regulating GBM stemness were investigated through pathway analysis in vitro and through tumor proliferation assay in vivo. RESULTS We demonstrated that Nav is functionally expressed by GSCs mainly during the G0 phase of the cell cycle, suggesting its pivotal role in modulating the RMP. The pharmacological blockade of Nav made GBM cells more susceptible to temozolomide (TMZ), a standard drug for this type of tumor, by inducing cell cycle re-entry from G0 phase to G1/S transition. Additionally, inhibition of Nav substantially influenced the self-renewal and multipotency features of GSCs, concomitantly enhancing their degree of differentiation. Finally, our data suggested that Nav positively regulates GBM stemness by depolarizing the RMP and suppressing the ERK signaling pathway. Of note, in vivo proliferation assessment confirmed the increased susceptibility to TMZ following pharmacological blockade of Nav. CONCLUSIONS This insight positions Nav as a promising prognostic biomarker and therapeutic target for GBM patients, particularly in conjunction with temozolomide treatment.
Collapse
Affiliation(s)
- Francesca Giammello
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
- PhD Program in Genetics, Molecular and Cellular Biology, University of Pavia, Pavia, Italy
| | - Chiara Biella
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Roberta Leone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | | - Martina Paterno'
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Giulia Cassioli
- Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Antea Minetti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Francesca Macchi
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Cristina Spalletti
- CNR Neuroscience Institute of Pisa, Via Giuseppe Moruzzi, 1, Pisa (PI), 56124, Italy
| | - Ilaria Morella
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Cristina Ruberti
- Advanced Technology Platform, Department of Biosciences, University of Milan, Milan, 20133, Italy
| | - Beatrice Tremonti
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Federica Barbieri
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
| | - Giuseppe Lombardi
- Department of Oncology 1, Oncology, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, Padua, 35128, Italy
| | - Riccardo Brambilla
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | - Tullio Florio
- Pharmacology Unit, Department of Internal Medicine, University of Genova, Genova, 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, 16132, Italy
| | - Rossella Galli
- IRCCS San Raffaele Hospital, Via Olgettina 58, Milan, 20132, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, 27100, Italy
| | | |
Collapse
|
6
|
Shrivastava A, Kumar A, Aggarwal LM, Pradhan S, Choudhary S, Ashish A, Kashyap K, Mishra S. Evolution of Bioelectric Membrane Potentials: Implications in Cancer Pathogenesis and Therapeutic Strategies. J Membr Biol 2024:10.1007/s00232-024-00323-2. [PMID: 39183198 DOI: 10.1007/s00232-024-00323-2] [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: 05/30/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Electrophysiology typically deals with the electrical properties of excitable cells like neurons and muscles. However, all other cells (non-excitable) also possess bioelectric membrane potentials for intracellular and extracellular communications. These membrane potentials are generated by different ions present in fluids available in and outside the cell, playing a vital role in communication and coordination between the cell and its organelles. Bioelectric membrane potential variations disturb cellular ionic homeostasis and are characteristic of many diseases, including cancers. A rapidly increasing interest has emerged in sorting out the electrophysiology of cancer cells. Compared to healthy cells, the distinct electrical properties exhibited by cancer cells offer a unique way of understanding cancer development, migration, and progression. Decoding the altered bioelectric signals influenced by fluctuating electric fields benefits understanding cancer more closely. While cancer research has predominantly focussed on genetic and molecular traits, the delicate area of electrophysiological characteristics has increasingly gained prominence. This review explores the historical exploration of electrophysiology in the context of cancer cells, shedding light on how alterations in bioelectric membrane potentials, mediated by ion channels and gap junctions, contribute to the pathophysiology of cancer.
Collapse
Affiliation(s)
- Anju Shrivastava
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India.
| | - Amit Kumar
- Department of Anatomy, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Lalit Mohan Aggarwal
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Satyajit Pradhan
- Radiation Oncology, Mahamana Pandit Madhan Mohan Malaviya Cancer Centre, Varanasi, India
| | - Sunil Choudhary
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Keshav Kashyap
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Shivani Mishra
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
7
|
Leslie TK, Tripp A, James AD, Fraser SP, Nelson M, Sajjaboontawee N, Capatina AL, Toss M, Fadhil W, Salvage SC, Garcia MA, Beykou M, Rakha E, Speirs V, Bakal C, Poulogiannis G, Djamgoz MBA, Jackson AP, Matthews HR, Huang CLH, Holding AN, Chawla S, Brackenbury WJ. A novel Na v1.5-dependent feedback mechanism driving glycolytic acidification in breast cancer metastasis. Oncogene 2024; 43:2578-2594. [PMID: 39048659 PMCID: PMC11329375 DOI: 10.1038/s41388-024-03098-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: 11/20/2023] [Revised: 06/29/2024] [Accepted: 07/04/2024] [Indexed: 07/27/2024]
Abstract
Solid tumours have abnormally high intracellular [Na+]. The activity of various Na+ channels may underlie this Na+ accumulation. Voltage-gated Na+ channels (VGSCs) have been shown to be functionally active in cancer cell lines, where they promote invasion. However, the mechanisms involved, and clinical relevance, are incompletely understood. Here, we show that protein expression of the Nav1.5 VGSC subtype strongly correlates with increased metastasis and shortened cancer-specific survival in breast cancer patients. In addition, VGSCs are functionally active in patient-derived breast tumour cells, cell lines, and cancer-associated fibroblasts. Knockdown of Nav1.5 in a mouse model of breast cancer suppresses expression of invasion-regulating genes. Nav1.5 activity increases ATP demand and glycolysis in breast cancer cells, likely by upregulating activity of the Na+/K+ ATPase, thus promoting H+ production and extracellular acidification. The pH of murine xenograft tumours is lower at the periphery than in the core, in regions of higher proliferation and lower apoptosis. In turn, acidic extracellular pH elevates persistent Na+ influx through Nav1.5 into breast cancer cells. Together, these findings show positive feedback between extracellular acidification and the movement of Na+ into cancer cells which can facilitate invasion. These results highlight the clinical significance of Nav1.5 activity as a potentiator of breast cancer metastasis and provide further evidence supporting the use of VGSC inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Theresa K Leslie
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Aurelien Tripp
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | - Andrew D James
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Scott P Fraser
- Department of Life Sciences, Imperial College London, London, UK
| | - Michaela Nelson
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Nattanan Sajjaboontawee
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Alina L Capatina
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Michael Toss
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Wakkas Fadhil
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham, UK
| | | | - Mar Arias Garcia
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | - Melina Beykou
- Division of Cancer Biology, Institute of Cancer Research, London, UK
- Department of Electrical and Electronic Engineering, Imperial College London, London, UK
| | - Emad Rakha
- Department of Pathology, School of Medicine, University of Nottingham, Nottingham, UK
| | - Valerie Speirs
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Chris Bakal
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | | | - Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, London, UK
- Biotechnology Research Centre, Cyprus International University, Haspolat, TRNC, Mersin, Turkey
| | - Antony P Jackson
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Hugh R Matthews
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Christopher L-H Huang
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Andrew N Holding
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - Sangeeta Chawla
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | - William J Brackenbury
- Department of Biology, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
8
|
Pukkanasut P, Jaskula-Sztul R, Gomora JC, Velu SE. Therapeutic targeting of voltage-gated sodium channel Na V1.7 for cancer metastasis. Front Pharmacol 2024; 15:1416705. [PMID: 39045054 PMCID: PMC11263763 DOI: 10.3389/fphar.2024.1416705] [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: 04/12/2024] [Accepted: 06/12/2024] [Indexed: 07/25/2024] Open
Abstract
This review focuses on the expression and function of voltage-gated sodium channel subtype NaV1.7 in various cancers and explores its impact on the metastasis driving cell functions such as proliferation, migration, and invasiveness. An overview of its structural characteristics, drug binding sites, inhibitors and their likely mechanisms of action are presented. Despite the lack of clarity on the precise mechanism by which NaV1.7 contributes to cancer progression and metastasis; many studies have suggested a connection between NaV1.7 and proteins involved in multiple signaling pathways such as PKA and EGF/EGFR-ERK1/2. Moreover, the functional activity of NaV1.7 appears to elevate the expression levels of MACC1 and NHE-1, which are controlled by p38 MAPK activity, HGF/c-MET signaling and c-Jun activity. This cascade potentially enhances the secretion of extracellular matrix proteases, such as MMPs which play critical roles in cell migration and invasion activities. Furthermore, the NaV1.7 activity may indirectly upregulate Rho GTPases Rac activity, which is critical for cytoskeleton reorganization, cell adhesion, and actin polymerization. The relationship between NaV1.7 and cancer progression has prompted researchers to investigate the therapeutic potential of targeting NaV1.7 using inhibitors. The positive outcome of such studies resulted in the discovery of several inhibitors with the ability to reduce cancer cell migration, invasion, and tumor growth underscoring the significance of NaV1.7 as a promising pharmacological target for attenuating cancer cell proliferation and metastasis. The research findings summarized in this review suggest that the regulation of NaV1.7 expression and function by small molecules and/or by genetic engineering is a viable approach to discover novel therapeutics for the prevention and treatment of metastasis of cancers with elevated NaV1.7 expression.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
9
|
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
|
10
|
Pio-Lopez L, Levin M. Aging as a loss of morphostatic information: A developmental bioelectricity perspective. Ageing Res Rev 2024; 97:102310. [PMID: 38636560 DOI: 10.1016/j.arr.2024.102310] [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: 11/05/2023] [Revised: 02/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Maintaining order at the tissue level is crucial throughout the lifespan, as failure can lead to cancer and an accumulation of molecular and cellular disorders. Perhaps, the most consistent and pervasive result of these failures is aging, which is characterized by the progressive loss of function and decline in the ability to maintain anatomical homeostasis and reproduce. This leads to organ malfunction, diseases, and ultimately death. The traditional understanding of aging is that it is caused by the accumulation of molecular and cellular damage. In this article, we propose a complementary view of aging from the perspective of endogenous bioelectricity which has not yet been integrated into aging research. We propose a view of aging as a morphostasis defect, a loss of biophysical prepattern information, encoding anatomical setpoints used for dynamic tissue and organ homeostasis. We hypothesize that this is specifically driven by abrogation of the endogenous bioelectric signaling that normally harnesses individual cell behaviors toward the creation and upkeep of complex multicellular structures in vivo. Herein, we first describe bioelectricity as the physiological software of life, and then identify and discuss the links between bioelectricity and life extension strategies and age-related diseases. We develop a bridge between aging and regeneration via bioelectric signaling that suggests a research program for healthful longevity via morphoceuticals. Finally, we discuss the broader implications of the homologies between development, aging, cancer and regeneration and how morphoceuticals can be developed for aging.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [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: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
12
|
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
|
13
|
Djamgoz MBA. Ranolazine: a potential anti-metastatic drug targeting voltage-gated sodium channels. Br J Cancer 2024; 130:1415-1419. [PMID: 38424164 PMCID: PMC11058819 DOI: 10.1038/s41416-024-02622-w] [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: 11/15/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Multi-faceted evidence from a range of cancers suggests strongly that de novo expression of voltage-gated sodium channels (VGSCs) plays a significant role in driving cancer cell invasiveness. Under hypoxic conditions, common to growing tumours, VGSCs develop a persistent current (INaP) which can be blocked selectively by ranolazine. METHODS Several different carcinomas were examined. We used data from a range of experimental approaches relating to cellular invasiveness and metastasis. These were supplemented by survival data mined from cancer patients. RESULTS In vitro, ranolazine inhibited invasiveness of cancer cells especially under hypoxia. In vivo, ranolazine suppressed the metastatic abilities of breast and prostate cancers and melanoma. These data were supported by a major retrospective epidemiological study on breast, colon and prostate cancer patients. This showed that risk of dying from cancer was reduced by ca.60% among those taking ranolazine, even if this started 4 years after the diagnosis. Ranolazine was also shown to reduce the adverse effects of chemotherapy on heart and brain. Furthermore, its anti-cancer effectiveness could be boosted by co-administration with other drugs. CONCLUSIONS Ranolazine, alone or in combination with appropriate therapies, could be reformulated as a safe anti-metastatic drug offering many potential advantages over current systemic treatment modalities.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
- Biotechnology Research Centre, Cyprus International University, Haspolat, Nicosia, TRNC, Mersin, 10, Türkiye.
| |
Collapse
|
14
|
Winnand P, Boernsen KO, Ooms M, Heitzer M, Lammert M, Eschweiler J, Hölzle F, Modabber A. The role of potassium in depth profiling of the tumor border in bone-invasive oral cancer using laser-induced breakdown spectroscopy (LIBS): a pilot study. J Cancer Res Clin Oncol 2023; 149:16635-16645. [PMID: 37716922 PMCID: PMC10645631 DOI: 10.1007/s00432-023-05411-9] [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: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023]
Abstract
PURPOSE Microscopic tumor spread beyond the macroscopically visible tumor mass in bone represents a major risk in surgical oncology, where the spatial complexity of bony resection margins cannot be countered with rapid bone analysis techniques. Laser-induced breakdown spectroscopy (LIBS) has recently been introduced as a promising option for rapid bone analysis. The present study aimed to use LIBS-based depth profiling based on electrolyte disturbance tracking to evaluate the detection of microscopic tumor spread in bone. METHODS After en bloc resection, the tumor-infiltrated mandible section of a patient's segmental mandibulectomy specimen was natively investigated using LIBS. Spectral and electrolytic depth profiles were analyzed across 30 laser shots per laser spot position in healthy bone and at the tumor border. For the histological validation of the lasered positions, the mandibular section was marked with a thin separating disc. RESULTS Solid calcium (Ca) from hydroxyapatite and soluble Ca from dissolved Ca can be reliably differentiated using LIBS and reflect the natural heterogeneity of healthy bone. Increased potassium (K) emission values in otherwise typically healthy bone spectra are the first spectral signs of tumorous bone invasion. LIBS-based depth profiles at the tumor border region can be used to track tumor-associated changes within the bone with shot accuracy based on the distribution of K. CONCLUSION Depth profiling using LIBS might enable the detection of microscopic tumor spread in bone. In the future, direct electrolyte tracking using LIBS should be applied to other intraoperative challenges in surgical oncology to advance rapid bone analysis by spectroscopic-optical techniques.
Collapse
Affiliation(s)
- Philipp Winnand
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany.
| | - K Olaf Boernsen
- Advanced Osteotomy Tools AG, Wallstrasse 6, 4051, Basel, Switzerland
| | - Mark Ooms
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Marius Heitzer
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Matthias Lammert
- Institute of Pathology, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Jörg Eschweiler
- Department of Orthopaedics, Trauma and Reconstructive Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Frank Hölzle
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Ali Modabber
- Department of Oral and Maxillofacial Surgery, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| |
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
|
Yang Q, Chen D. Na + Binding and Transport: Insights from Light-Driven Na +-Pumping Rhodopsin. Molecules 2023; 28:7135. [PMID: 37894614 PMCID: PMC10608830 DOI: 10.3390/molecules28207135] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Na+ plays a vital role in numerous physiological processes across humans and animals, necessitating a comprehensive understanding of Na+ transmembrane transport. Among the various Na+ pumps and channels, light-driven Na+-pumping rhodopsin (NaR) has emerged as a noteworthy model in this field. This review offers a concise overview of the structural and functional studies conducted on NaR, encompassing ground/intermediate-state structures and photocycle kinetics. The primary focus lies in addressing key inquiries: (1) unraveling the translocation pathway of Na+; (2) examining the role of structural changes within the photocycle, particularly in the O state, in facilitating Na+ transport; and (3) investigating the timing of Na+ uptake/release. By delving into these unresolved issues and existing debates, this review aims to shed light on the future direction of Na+ pump research.
Collapse
Affiliation(s)
- Qifan Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Deliang Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
17
|
Folz J, Wasserman JH, Jo J, Wang X, Kopelman R. Photoacoustic Chemical Imaging Sodium Nano-Sensor Utilizing a Solvatochromic Dye Transducer for In Vivo Application. BIOSENSORS 2023; 13:923. [PMID: 37887116 PMCID: PMC10605089 DOI: 10.3390/bios13100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
Sodium has many vital and diverse roles in the human body, including maintaining the cellular pH, generating action potential, and regulating osmotic pressure. In cancer, sodium dysregulation has been correlated with tumor growth, metastasis, and immune cell inhibition. However, most in vivo sodium measurements are performed via Na23 NMR, which is handicapped by slow acquisition times, a low spatial resolution (in mm), and low signal-to-noise ratios. We present here a plasticizer-free, ionophore-based sodium-sensing nanoparticle that utilizes a solvatochromic dye transducer to circumvent the pH cross-sensitivity of most previously reported sodium nano-sensors. We demonstrate that this nano-sensor is non-toxic, boasts a 200 μM detection limit, and is over 1000 times more selective for sodium than potassium. Further, the in vitro photoacoustic calibration curve presented demonstrates the potential of this nano-sensor for performing the in vivo chemical imaging of sodium over the entire physiologically relevant concentration range.
Collapse
Affiliation(s)
- Jeff Folz
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA;
| | | | - Janggun Jo
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (J.J.); (X.W.)
| | - Xueding Wang
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; (J.J.); (X.W.)
| | - Raoul Kopelman
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA;
| |
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
|
Ghosh S, Yang R, Duraki D, Zhu J, Kim JE, Jabeen M, Mao C, Dai X, Livezey MR, Boudreau MW, Park BH, Nelson ER, Hergenrother PJ, Shapiro DJ. Plasma Membrane Channel TRPM4 Mediates Immunogenic Therapy-Induced Necrosis. Cancer Res 2023; 83:3115-3130. [PMID: 37522838 PMCID: PMC10635591 DOI: 10.1158/0008-5472.can-23-0157] [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: 01/13/2023] [Revised: 05/15/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Several emerging therapies kill cancer cells primarily by inducing necrosis. As necrosis activates immune cells, potentially, uncovering the molecular drivers of anticancer therapy-induced necrosis could reveal approaches for enhancing immunotherapy efficacy. To identify necrosis-associated genes, we performed a genome-wide CRISPR-Cas9 screen with negative selection against necrosis-inducing preclinical agents BHPI and conducted follow-on experiments with ErSO. The screen identified transient receptor potential melastatin member 4 (TRPM4), a calcium-activated, ATP-inhibited, sodium-selective plasma membrane channel. Cancer cells selected for resistance to BHPI and ErSO exhibited robust TRPM4 downregulation, and TRPM4 reexpression restored sensitivity to ErSO. Notably, TRPM4 knockout (TKO) abolished ErSO-induced regression of breast tumors in mice. Supporting a broad role for TRPM4 in necrosis, knockout of TRPM4 reversed cell death induced by four additional diverse necrosis-inducing cancer therapies. ErSO induced anticipatory unfolded protein response (a-UPR) hyperactivation, long-term necrotic cell death, and release of damage-associated molecular patterns that activated macrophages and increased monocyte migration, all of which was abolished by TKO. Furthermore, loss of TRPM4 suppressed the ErSO-induced increase in cell volume and depletion of ATP. These data suggest that ErSO triggers initial activation of the a-UPR but that it is TRPM4-mediated sodium influx and cell swelling, resulting in osmotic stress, which sustains and propagates lethal a-UPR hyperactivation. Thus, TRPM4 plays a pivotal role in sustaining lethal a-UPR hyperactivation that mediates the anticancer activity of diverse necrosis-inducing therapies. SIGNIFICANCE A genome-wide CRISPR screen reveals a pivotal role for TRPM4 in cell death and immune activation following treatment with diverse necrosis-inducing anticancer therapies, which could facilitate development of necrosis-based cancer immunotherapies.
Collapse
Affiliation(s)
- Santanu Ghosh
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Rachel Yang
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Darjan Duraki
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Junyao Zhu
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ji Eun Kim
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Musarrat Jabeen
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Chengjian Mao
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xinyi Dai
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Mara R. Livezey
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Chemistry and Biochemistry, University of Detroit Mercy, Detroit, MI 48221, USA (present address)
| | - Matthew W. Boudreau
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215 (present address)
| | - Ben H. Park
- Vanderbilt University College of Medicine, Nashville, TN, 37232, USA
| | - Erik R. Nelson
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Paul J. Hergenrother
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - David J. Shapiro
- Departments of Biochemistry, Molecular and Integrative Physiology and Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
20
|
Hrvat A, Schmidt M, Wagner B, Zwanziger D, Kimmig R, Volbracht L, Brandau S, Mallmann-Gottschalk N. Electrolyte imbalance causes suppression of NK and T cell effector function in malignant ascites. J Exp Clin Cancer Res 2023; 42:235. [PMID: 37684704 PMCID: PMC10485936 DOI: 10.1186/s13046-023-02798-8] [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: 04/18/2023] [Accepted: 08/13/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Malignant ascites commonly occurs in advanced or recurrent stages of epithelial ovarian cancer during peritoneal carcinomatosis and is correlated with poor prognosis. Due to its complex composition of cellular and acellular components malignant ascites creates a unique tumor microenvironment, which mediates immunosuppression and promotes progression of disease. However, the immunosuppressive mechanisms remain poorly understood. METHODS In the present study, we explored the antitumor activity of healthy donor NK and T cells directed against ovarian cancer cells in presence of malignant ascites derived from patients with advanced or recurrent peritoneal carcinomatosis. A wide range of methods was used to study the effect of ascites on NK and T cells (FACS, ELISA, EliSpot, qPCR, Live-cell and confocal microscopy, Western blot and electrolyte flux assays). The ascites components were assessed using quantitative analysis (nephelometry, potentiometry and clinical chemistry) and separation methods (dialysis, ultracentrifugal filtration and lipid depletion). RESULTS Ascites rapidly inhibited NK cell degranulation, tumor lysis, cytokine secretion and calcium signaling. Similarly, target independent NK and T cell activation was impaired in ascites environment. We identified imbalanced electrolytes in ascites as crucial factors causing extensive immunosuppression of NK and T cells. Specifically, high sodium, low chloride and low potassium content significantly suppressed NK-mediated cytotoxicity. Electrolyte imbalance led to changes in transcription and protein expression of electrolyte channels and impaired NK and T cell activation. Selected inhibitors of sodium electrolyte channels restored intracellular calcium flux, conjugation, degranulation and transcript expression of signaling molecules. The levels of ascites-mediated immunosuppression and sodium/chloride/potassium imbalance correlated with poor patient outcome and selected molecular alterations were confirmed in immune cells from ovarian cancer patients. CONCLUSION Our data suggest a novel electrolyte-based mechanism of immunosuppression in malignant ascites of patients with peritoneal carcinomatosis. We show for the first time that the immunosuppression of NK cytotoxicity in coculture assays is correlated to patient poor survival. Therapeutic application of sodium channel inhibitors may provide new means for restoring immune cell activity in ascites or similar electrolyte imbalanced environments.
Collapse
Affiliation(s)
- Antonio Hrvat
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Mathias Schmidt
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Bernd Wagner
- Department of Clinical Chemistry, University Hospital Essen, 45147, Essen, Germany
| | - Denise Zwanziger
- Department of Clinical Chemistry, University Hospital Essen, 45147, Essen, Germany
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, 45147, Essen, Germany
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, 45147, Essen, Germany
| | - Lothar Volbracht
- Department of Clinical Chemistry, University Hospital Essen, 45147, Essen, Germany
| | - Sven Brandau
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
- partner site Essen-Düsseldorf, German Cancer Consortium (DKTK), 45147, Essen, Germany.
| | - Nina Mallmann-Gottschalk
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
- Department of Gynecology and Obstetrics, University Hospital of Cologne, 50931, Cologne, Germany
| |
Collapse
|
21
|
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
|
22
|
Clemente N, Baroni S, Fiorilla S, Tasso F, Reano S, Borsotti C, Ruggiero MR, Alchera E, Corrazzari M, Walker G, Follenzi A, Crich SG, Carini R. Boosting intracellular sodium selectively kills hepatocarcinoma cells and induces hepatocellular carcinoma tumor shrinkage in mice. Commun Biol 2023; 6:574. [PMID: 37248274 DOI: 10.1038/s42003-023-04946-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/16/2023] [Indexed: 05/31/2023] Open
Abstract
Pharmacological treatments for advanced hepatocellular carcinoma (HCC) have a partial efficacy. Augmented Na+ content and water retention are observed in human cancers and offer unexplored targets for anticancer therapies. Na+ levels are evaluated upon treatments with the antibiotic cation ionophore Monensin by fluorimetry, ICP-MS, 23Na-MRI, NMR relaxometry, confocal or time-lapse analysis related to energy production, water fluxes and cell death, employing both murine and human HCC cell lines, primary murine hepatocytes, or HCC allografts in NSG mice. Na+ levels of HCC cells and tissue are 8-10 times higher than that of healthy hepatocytes and livers. Monensin further increases Na+ levels in HCC cells and in HCC allografts but not in primary hepatocytes and in normal hepatic and extrahepatic tissue. The Na+ increase is associated with energy depletion, mitochondrial Na+ load and inhibition of O2 consumption. The Na+ increase causes an enhancement of the intracellular water lifetime and death of HCC cells, and a regression and necrosis of allograft tumors, without affecting the proliferating activity of either HCCs or healthy tissues. These observations indicate that HCC cells are, unlike healthy cells, energetically incapable of compensating and surviving a pharmacologically induced Na+ load, highlighting Na+ homeostasis as druggable target for HCC therapy.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Simona Baroni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126, Torino, Italy
| | - Simone Fiorilla
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Francesco Tasso
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Simone Reano
- Department of Department of Translational Medicine, Unit of Muscle Biology, Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Chiara Borsotti
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Maria Rosaria Ruggiero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126, Torino, Italy
| | - Elisa Alchera
- Division of Experimental Oncology/Unit of Urology, URI, IRCCS, Ospedale San Raffaele, Milan, Italy
| | - Marco Corrazzari
- Department of Health Science and Interdisciplinary Research Center of Autoimmune Disease (IRCAD), Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Gillian Walker
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Antonia Follenzi
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy
| | - Simonetta Geninatti Crich
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126, Torino, Italy.
| | - Rita Carini
- Department of Health Science Università del Piemonte Orientale, Via Solaroli, 17, 28100, Novara, Italy.
| |
Collapse
|
23
|
Leslie TK, Brackenbury WJ. Sodium channels and the ionic microenvironment of breast tumours. J Physiol 2023; 601:1543-1553. [PMID: 36183245 PMCID: PMC10953337 DOI: 10.1113/jp282306] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022] Open
Abstract
Cancers of epithelial origin such as breast, prostate, cervical, gastric, colon and lung cancer account for a large proportion of deaths worldwide. Better treatment of metastasis, the main cause of cancer deaths, is therefore urgently required. Several of these tumours have been shown to have an abnormally high concentration of Na+ ([Na+ ]) and emerging evidence points to this accumulation being due to elevated intracellular [Na+ ]. This poses intriguing questions about the cellular mechanisms underlying Na+ dysregulation in cancer, and its pathophysiological significance. Elevated intracellular [Na+ ] may be due to alterations in activity of the Na+ /K+ -ATPase, and/or increased influx via Na+ channels and Na+ -linked transporters. Maintenance of the electrochemical Na+ gradient across the plasma membrane is vital to power many cellular processes that are highly active in cancer cells, including glucose and glutamine import. Na+ channels are also upregulated in cancer cells, which in turn promotes tumour growth and metastasis. For example, ENaC and ASICs are overexpressed in cancers, increasing invasion and proliferation. In addition, voltage-gated Na+ channels are also upregulated in a range of tumour types, where they promote metastatic cell behaviours via various mechanisms, including membrane potential depolarisation and altered pH regulation. Together, recent findings relating to elevated Na+ in the tumour microenvironment and how this may be regulated by several classes of Na+ channels provide a link between altered Na+ handling and poor clinical outcome. There are new opportunities to leverage this altered Na+ microenvironment for therapeutic benefit, as exemplified by several ongoing clinical trials.
Collapse
Affiliation(s)
- Theresa K. Leslie
- Department of BiologyUniversity of YorkHeslingtonYorkUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkUK
| | - William J. Brackenbury
- Department of BiologyUniversity of YorkHeslingtonYorkUK
- York Biomedical Research InstituteUniversity of YorkHeslingtonYorkUK
| |
Collapse
|
24
|
Sun J, Tong Y, Jia Y, Jia X, Wang H, Chen Y, Wu J, Jin W, Ma Z, Cao K, Li X, Chen Z, Yang G. Effects of extremely low frequency electromagnetic fields on the tumor cell inhibition and the possible mechanism. Sci Rep 2023; 13:6989. [PMID: 37117238 PMCID: PMC10147919 DOI: 10.1038/s41598-023-34144-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/25/2023] [Indexed: 04/30/2023] Open
Abstract
Low-frequency magnetic fields exert a significant inhibitory effect on tumor growth and have been developed as a therapeutic modality. However, the effect of a low-frequency magnetic field on the interaction between cells is still poorly understood. This study aimed to preliminarily evaluate the direct effect of magnetic field ditectely on cultured cells and indirect effect mediated by cell-environment (conditioned medium). 293 T cells, Hepg2 cells, A549 cells have been cultured at 37 ± 0.18 °C in presence of an extremely low-frequency magnetic field of 20 Hz, 5-mT. The adherent tumor cells were more sensitive to magnetic field inhibition in the original environment (conditioned medium) with adherence inhibition rate for Hepg2 and A549 estimated at 18% and 30% respectively. The inhibition effect was suppressed when the suspended cells separated or clump density at a low density. The nontumor cell lines showed no inhibitory effect on exposure to a low-frequency magnetic field. The intracellular ion fluorescence (IIF) showed that the magnetic field significantly altered the membrane potential, indicating hyperpolarization of the adherent cells (ΔIIF 293 T cells: - 25%, ΔIIF Hepg2 cells: - 20% and ΔIIF A549 cells: - 13%) and depolarization of the suspended cells (ΔIIF Raji cells: + 9%). In addition, the conditioned media collected after magnetic field exposure acted on unexposed tumor cells and caused inhibition. Our findings might provide a basis for the mechanism of magnetic field interaction between cells and cell environment in the future.
Collapse
Affiliation(s)
- Jie Sun
- International Research Center for Biological Sciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- National Aquatic Animal Pathogen Collection Center, Shanghai Ocean University, Shanghai, 201306, China
- Aquatic Animal Genetics and Breeding Center, Shanghai Ocean University, Shanghai, 201306, China
| | - Yingying Tong
- International Research Center for Biological Sciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- National Aquatic Animal Pathogen Collection Center, Shanghai Ocean University, Shanghai, 201306, China
- Aquatic Animal Genetics and Breeding Center, Shanghai Ocean University, Shanghai, 201306, China
| | - Yu Jia
- International Research Center for Biological Sciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- National Aquatic Animal Pathogen Collection Center, Shanghai Ocean University, Shanghai, 201306, China
- Aquatic Animal Genetics and Breeding Center, Shanghai Ocean University, Shanghai, 201306, China
| | - Xu Jia
- International Research Center for Biological Sciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- National Aquatic Animal Pathogen Collection Center, Shanghai Ocean University, Shanghai, 201306, China
- Aquatic Animal Genetics and Breeding Center, Shanghai Ocean University, Shanghai, 201306, China
| | - Hua Wang
- Shanghai Telebio Biomedical Co., Ltd, Shanghai, China
| | - Yang Chen
- International Research Center for Biological Sciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
- National Aquatic Animal Pathogen Collection Center, Shanghai Ocean University, Shanghai, 201306, China
- Aquatic Animal Genetics and Breeding Center, Shanghai Ocean University, Shanghai, 201306, China
| | - Jiamin Wu
- Zhejiang Huayi Health Industry Development Co., Ltd, Hangzhou, China
| | - Weiyang Jin
- Zhejiang Huayi Health Industry Development Co., Ltd, Hangzhou, China
| | - Zheng Ma
- Huisi Anpu Medical System Co., Ltd, Qinhuangdao, China
| | - Kai Cao
- Huisi Anpu Medical System Co., Ltd, Qinhuangdao, China
| | - Xiangdong Li
- Huisi Anpu Medical System Co., Ltd, Qinhuangdao, China
| | - Zhonglin Chen
- Huisi Anpu Medical System Co., Ltd, Qinhuangdao, China
| | - Guanghua Yang
- International Research Center for Biological Sciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.
- National Aquatic Animal Pathogen Collection Center, Shanghai Ocean University, Shanghai, 201306, China.
- Aquatic Animal Genetics and Breeding Center, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
25
|
Ridley B, Morsillo F, Zaaraoui W, Nonino F. Variability by region and method in human brain sodium concentrations estimated by 23Na magnetic resonance imaging: a meta-analysis. Sci Rep 2023; 13:3222. [PMID: 36828873 PMCID: PMC9957999 DOI: 10.1038/s41598-023-30363-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/21/2023] [Indexed: 02/26/2023] Open
Abstract
Sodium imaging (23Na-MRI) is of interest in neurological conditions given potential sensitivity to the physiological and metabolic status of tissues. Benchmarks have so far been restricted to parenchyma or grey/white matter (GM/WM). We investigate (1) the availability of evidence, (2) regional pooled estimates and (3) variability attributable to region/methodology. MEDLINE literature search for tissue sodium concentration (TSC) measured in specified 'healthy' brain regions returned 127 reports, plus 278 retrieved from bibliographies. 28 studies met inclusion criteria, including 400 individuals. Reporting variability led to nested data structure, so we used multilevel meta-analysis and a random effects model to pool effect sizes. The pooled mean from 141 TSC estimates was 40.51 mM (95% CI 37.59-43.44; p < 0.001, I2Total=99.4%). Tissue as a moderator was significant (F214 = 65.34, p-val < .01). Six sub-regional pooled means with requisite statistical power were derived. We were unable to consider most methodological and demographic factors sought because of non-reporting, but each factor included beyond tissue improved model fit. Significant residual heterogeneity remained. The current estimates provide an empirical point of departure for better understanding in 23Na-MRI. Improving on current estimates supports: (1) larger, more representative data collection/sharing, including (2) regional data, and (3) agreement on full reporting standards.
Collapse
Affiliation(s)
- Ben Ridley
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy.
- Ben Ridley, Epidemiologia e Statistica, IRCCS Istituto Delle Scienze Neurologiche di Bologna, Padiglione G, Via Altura, 3, 40139, Bologna, Italy.
| | - Filomena Morsillo
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Wafaa Zaaraoui
- Aix Marseille Univ, CNRS, CRMBM, Marseille, France
- APHM, Hôpital de La Timone, CEMEREM, Marseille, France
| | - Francesco Nonino
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
26
|
Bala N, Yu L, Liu LP, Shelton L, Xu Y, Ghayee HK, Alli AA. Metabolic Characterization and Glyceraldehyde-3-Phosphate Dehydrogenase-Dependent Regulation of Epithelial Sodium Channels in hPheo1 Wild-type and SDHB Knockdown Cells. Endocrinology 2023; 164:7034155. [PMID: 36763043 DOI: 10.1210/endocr/bqad026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023]
Abstract
Pheochromocytomas (PCC) and paragangliomas (PGL) are rare neuroendocrine tumors with limited curative treatment options outside of surgical resection. Patients with mutations in succinate dehydrogenase subunit B (SDHB) are at an increased risk of malignant and aggressive disease. As cation channels are associated with tumorigenesis, we studied the expression and activity of cation channels from the Degenerin superfamily in a progenitor cell line derived from a human PCC. hPheo1 wild-type (WT) and SDHB knockdown (KD) cells were studied to investigate whether epithelial sodium channels (ENaC) and acid-sensing ion channels (ASIC) are regulated by the activity of glyceraldehyde-3-phosphate dehydrogenase (GAPDH). First, we performed targeted metabolomic studies and quantified changes in glycolysis pathway intermediates and citric acid cycle intermediates using hPheo1 WT cells and SDHB KD cells. Next, we performed protein biochemistry and electrophysiology studies to characterize the protein expression and activity, respectively, of these ion channels. Our western blot experiments show both ENaC alpha and ASIC1/2 are expressed in both hPheo1 WT and SDHB KD cells, with lower levels of a cleaved 60 kDa form of ENaC in SDHB KD cells. Single-channel patch clamp studies corroborate these results and further indicate channel activity is decreased in SDHB KD cells. Additional experiments showed a more significant decreased membrane potential in SDHB KD cells, which were sensitive to amiloride compared to WT cells. We provide evidence for the differential expression and activity of ENaC and ASIC hybrid channels in hPheo1 WT and SDHB KD cells, providing an important area of investigation in understanding SDHB-related disease.
Collapse
Affiliation(s)
- Niharika Bala
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, USA
| | - Ling Yu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, USA
| | - Lauren P Liu
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Yiling Xu
- Department of Medicine, Division of Endocrinology & Metabolism, University of Florida College of Medicine, Gainesville, FL, USA
| | - Hans K Ghayee
- Department of Medicine, Division of Endocrinology & Metabolism, University of Florida College of Medicine, Gainesville, FL, USA
- Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Abdel A Alli
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, USA
- Department of Medicine Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
27
|
Fairhurst C, Martin F, Watt I, Bland M, Doran T, Brackenbury WJ. Sodium channel-inhibiting drugs and cancer-specific survival: a population-based study of electronic primary care data. BMJ Open 2023; 13:e064376. [PMID: 36737094 PMCID: PMC9900071 DOI: 10.1136/bmjopen-2022-064376] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 01/19/2023] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Antiepileptic and antiarrhythmic drugs inhibit voltage-gated sodium (Na+) channels (VGSCs), and preclinical studies show that these medications reduce tumour growth, invasion and metastasis. We investigated the association between VGSC inhibitor use and survival in patients with breast, bowel and prostate cancer. DESIGN Retrospective cohort study. SETTING Individual electronic primary healthcare records extracted from the Clinical Practice Research Datalink. PARTICIPANTS Records for 132 996 patients with a diagnosis of breast, bowel or prostate cancer. OUTCOME MEASURES Adjusted Cox proportional hazards regression was used to analyse cancer-specific survival associated with exposure to VGSC inhibitors. Exposure to non-VGSC-inhibiting antiepileptic medication and other non-VGSC blockers were also considered. Drug exposure was treated as a time-varying covariate to account for immortal time bias. RESULTS During 1 002 225 person-years of follow-up, there were 42 037 cancer-specific deaths. 53 724 (40.4%) patients with cancer had at least one prescription for a VGSC inhibitor of interest. Increased risk of cancer-specific mortality was associated with exposure to this group of drugs (HR 1.59, 95% CI 1.56 to 1.63, p<0.001). This applied to VGSC-inhibiting tricyclic antidepressants (HR 1.61, 95% CI 1.50 to 1.65, p<0.001), local anaesthetics (HR 1.49, 95% CI 1.43 to 1.55, p<0.001) and anticonvulsants (HR 1.40, 95% CI 1.34 to 1.48, p<0.001) and persisted in sensitivity analyses. In contrast, exposure to VGSC-inhibiting class 1c and 1d antiarrhythmics was associated with significantly improved cancer-specific survival (HR 0.75, 95% CI 0.64 to 0.88, p<0.001 and HR 0.54, 95% CI 0.33 to 0.88, p=0.01, respectively). CONCLUSIONS Association between VGSC inhibitor use and mortality in patients with cancer varies according to indication. Exposure to VGSC-inhibiting antiarrhythmics, but not anticonvulsants, supports findings from preclinical data, with improved survival. However, additional confounding factors may underlie these associations, highlighting the need for further study.
Collapse
Affiliation(s)
| | - Fabiola Martin
- University of Queensland, Brisbane, Queensland, Australia
| | - Ian Watt
- Health Sciences, University of York, York, UK
| | | | - Tim Doran
- Health Sciences, University of York, York, UK
| | | |
Collapse
|
28
|
Shen F, Fang Y, Wu Y, Zhou M, Shen J, Fan X. Metal ions and nanometallic materials in antitumor immunity: Function, application, and perspective. J Nanobiotechnology 2023; 21:20. [PMID: 36658649 PMCID: PMC9850565 DOI: 10.1186/s12951-023-01771-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
The slightest change in the extra/intracellular concentration of metal ions results in amplified effects by signaling cascades that regulate both cell fate within the tumor microenvironment and immune status, which influences the network of antitumor immunity through various pathways. Based on the fact that metal ions influence the fate of cancer cells and participate in both innate and adaptive immunity, they are widely applied in antitumor therapy as immune modulators. Moreover, nanomedicine possesses the advantage of precise delivery and responsive release, which can perfectly remedy the drawbacks of metal ions, such as low target selectivity and systematic toxicity, thus providing an ideal platform for metal ion application in cancer treatment. Emerging evidence has shown that immunotherapy applied with nanometallic materials may significantly enhance therapeutic efficacy. Here, we focus on the physiopathology of metal ions in tumorigenesis and discuss several breakthroughs regarding the use of nanometallic materials in antitumor immunotherapeutics. These findings demonstrate the prominence of metal ion-based nanomedicine in cancer therapy and prophylaxis, providing many new ideas for basic immunity research and clinical application. Consequently, we provide innovative insights into the comprehensive understanding of the application of metal ions combined with nanomedicine in cancer immunotherapy in the past few years.
Collapse
Affiliation(s)
- Feiyang Shen
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025 China
| | - Yan Fang
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025 China
| | - Yijia Wu
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025 China
| | - Min Zhou
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025 China
| | - Jianfeng Shen
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Xianqun Fan
- grid.16821.3c0000 0004 0368 8293Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200025 China ,grid.16821.3c0000 0004 0368 8293Institute of Translational Medicine, National Facility for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240 China
| |
Collapse
|
29
|
Mitochondria directly sense osmotic stress to trigger rapid metabolic remodeling via regulation of pyruvate dehydrogenase phosphorylation. J Biol Chem 2022; 299:102837. [PMID: 36581206 PMCID: PMC9879793 DOI: 10.1016/j.jbc.2022.102837] [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: 07/20/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022] Open
Abstract
A high-salt diet significantly impacts various diseases, ilncluding cancer and immune diseases. Recent studies suggest that the high-salt/hyperosmotic environment in the body may alter the chronic properties of cancer and immune cells in the disease context. However, little is known about the acute metabolic changes in hyperosmotic stress. Here, we found that hyperosmotic stress for a few minutes induces Warburg-like metabolic remodeling in HeLa and Raw264.7 cells and suppresses fatty acid oxidation. Regarding Warburg-like remodeling, we determined that the pyruvate dehydrogenase phosphorylation status was altered bidirectionally (high in hyperosmolarity and low in hypoosmolarity) to osmotic stress in isolated mitochondria, suggesting that mitochondria themselves have an acute osmosensing mechanism. Additionally, we demonstrate that Warburg-like remodeling is required for HeLa cells to maintain ATP levels and survive under hyperosmotic conditions. Collectively, our findings suggest that cells exhibit acute metabolic remodeling under osmotic stress via the regulation of pyruvate dehydrogenase phosphorylation by direct osmosensing within mitochondria.
Collapse
|
30
|
Lopez-Charcas O, Poisson L, Benouna O, Lemoine R, Chadet S, Pétereau A, Lahlou W, Guyétant S, Ouaissi M, Pukkanasut P, Dutta S, Velu SE, Besson P, Moussata D, Roger S. Voltage-Gated Sodium Channel Na V1.5 Controls NHE-1-Dependent Invasive Properties in Colon Cancer Cells. Cancers (Basel) 2022; 15:cancers15010046. [PMID: 36612049 PMCID: PMC9817685 DOI: 10.3390/cancers15010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of death worldwide, with 0.9 million deaths per year. The metastatic stage of the disease is identified in about 20% of cases at the first diagnosis and is associated with low patient-survival rates. Voltage-gated sodium channels (NaV) are abnormally overexpressed in several carcinomas including CRC and are strongly associated with the metastatic behavior of cancer cells. Acidification of the extracellular space by Na+/H+ exchangers (NHE) contributes to extracellular matrix degradation and cell invasiveness. In this study, we assessed the expression levels of pore-forming α-subunits of NaV channels and NHE exchangers in tumor and adjacent non-malignant tissues from colorectal cancer patients, CRC cell lines and primary tumor cells. In all cases, SCN5A (gene encoding for NaV1.5) was overexpressed and positively correlated with cancer stage and poor survival prognosis for patients. In addition, we identified an anatomical differential expression of SCN5A and SLC9A1 (gene encoding for NHE-1) being particularly relevant for tumors that originated on the sigmoid colon epithelium. The functional activity of NaV1.5 channels was characterized in CRC cell lines and the primary cells of colon tumors obtained using tumor explant methodologies. Furthermore, we assessed the performance of two new small-molecule NaV1.5 inhibitors on the reduction of sodium currents, as well as showed that silencing SCN5A and SLC9A1 substantially reduced the 2D invasive capabilities of cancer cells. Thus, our findings show that both NaV1.5 and NHE-1 represent two promising targetable membrane proteins against the metastatic progression of CRC.
Collapse
Affiliation(s)
- Osbaldo Lopez-Charcas
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
- Correspondence: (O.L.-C.); (S.R.); Tel.: +33-2-47-36-61-30 (S.R.)
| | - Lucile Poisson
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
| | - Oumnia Benouna
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
| | - Roxane Lemoine
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
| | - Stéphanie Chadet
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
| | - Adrien Pétereau
- Service D’anatomie et de Cytologie Pathologiques, Hôpital Trousseau, CHU de Tours, 37170 Tours, France
| | - Widad Lahlou
- Service D’hépato-Gastroentérologie et de Cancérologie Digestive, Hôpital Trousseau, CHU de Tours, 37170 Tours, France
| | - Serge Guyétant
- Service D’anatomie et de Cytologie Pathologiques, Hôpital Trousseau, CHU de Tours, 37170 Tours, France
| | - Mehdi Ouaissi
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
- Service de Chirurgie Viscérale et Oncologique, Hôpital Trousseau, CHU de Tours, 37170 Tours, France
| | - Piyasuda Pukkanasut
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294-1240, USA
| | - Shilpa Dutta
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294-1240, USA
| | - Sadanandan E. Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294-1240, USA
| | - Pierre Besson
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
| | - Driffa Moussata
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
- Service D’hépato-Gastroentérologie et de Cancérologie Digestive, Hôpital Trousseau, CHU de Tours, 37170 Tours, France
| | - Sébastien Roger
- EA4245, Transplantation, Immunologie et Inflammation, Faculté de Médecine, Université de Tours, 37032 Tours, France
- Correspondence: (O.L.-C.); (S.R.); Tel.: +33-2-47-36-61-30 (S.R.)
| |
Collapse
|
31
|
Diržiuvienė R, Šlekienė L, Palubinskienė J, Balnytė I, Lasienė K, Stakišaitis D, Valančiūtė A. Tumors derived from lung cancer cells respond differently to treatment with sodium valproate (a HDAC inhibitor) in a chicken embryo chorioallantoic membrane model. Histol Histopathol 2022; 37:1201-1212. [PMID: 35703146 DOI: 10.14670/hh-18-482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lung cancer is the most frequent cause of cancer death. Some human lung malignant tumors have a combined small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC) histology, with tumor cell phenotype changing during tumor progression. Valproic acid is used as an anti-seizure medication to treat migraine, and bipolar mood disorders. Recently, its efficacy as an adjuvant therapy was shown in cancer due to its histone deacetylase (HDAC) inhibitory property. HDACs are upregulated in lung tumors, and HDAC inhibitors, including valproic acid, inhibit endothelial cell proliferation in vitro and in vivo and have antiproliferative and antimigratory properties. We tested valproic acid for possible antiangiogenic and antimigratory effects on experimental lung tumors grafted onto the chicken embryo chorioallantoic membrane (CAM). Tumors were formed from two NSCLC cell lines and a single SCLC cell line. To investigate tumor and CAM interactions, in vivo biomicroscopy, visualization of blood vessels with injected fluorescent dextran, histological, immunohistochemical and histomorphometric methods were applied. Our results showed that a sodium valproate (NaVP) treatment-induced a dose-dependent decrease of experimental tumor invasion into the CAM mesenchyme and a reduction in angiogenesis. Both the invasion and the angiogenic response were dependent on the type of cell line used: invasion and angiogenesis of tumors derived from A549 and NCI-H146 cell lines responded to increasing doses of NaVP from 4 to 8 mM, whereas Sk_Lu_1 cells response were antimigratory and antiangiogenic when NaVP was used up to 6 mM. When 8mM NaVP was used, stimulated invasion and angiogenesis in tumors from Sk_Lu_1 cells were observed.
Collapse
Affiliation(s)
- Raminta Diržiuvienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Lina Šlekienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Jolita Palubinskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kristina Lasienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Laboratory of Molecular Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
32
|
Edenfield S, Sims AM, Porretta C, Gould HJ, Paul D. Effect of Cell Cycle on Cell Surface Expression of Voltage-Gated Sodium Channels and Na +,K +-ATPase. Cells 2022; 11:cells11203240. [PMID: 36291108 PMCID: PMC9600173 DOI: 10.3390/cells11203240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/25/2022] Open
Abstract
Voltage-gated sodium channels (VGSCs) are the target for many therapies. Variation in membrane potential occurs throughout the cell cycle, yet little attention has been devoted to the role of VGSCs and Na+,K+-ATPases. We hypothesized that in addition to doubling DNA and cell membrane in anticipation of cell division, there should be a doubling of VGSCs and Na+,K+-ATPase compared to non-dividing cells. We tested this hypothesis in eight immortalized cell lines by correlating immunocytofluorescent labeling of VGSCs or Na+,K+-ATPase with propidium iodide or DAPI fluorescence using flow cytometry and imaging. Cell surface expression of VGSCs during phases S through M was double that seen during phases G0–G1. By contrast, Na+,K+-ATPase expression increased only 1.5-fold. The increases were independent of baseline expression of channels or pumps. The variation in VGSC and Na+,K+-ATPase expression has implications for both our understanding of sodium’s role in controlling the cell cycle and variability of treatments targeted at these components of the Na+ handling system.
Collapse
Affiliation(s)
- Samantha Edenfield
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Abigail M. Sims
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Constance Porretta
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Harry J. Gould
- Department of Neurology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Anesthesiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Correspondence: (H.J.G.III); (D.P.)
| | - Dennis Paul
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Neurology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Anesthesiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Dental and Craniofacial Biology Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70119, USA
- Correspondence: (H.J.G.III); (D.P.)
| |
Collapse
|
33
|
Cancer as a Channelopathy—Appreciation of Complimentary Pathways Provides a Different Perspective for Developing Treatments. Cancers (Basel) 2022; 14:cancers14194627. [PMID: 36230549 PMCID: PMC9562872 DOI: 10.3390/cancers14194627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/17/2022] [Accepted: 09/21/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary While improvements in technology have improved our ability to treat many forms of cancer when diagnosed at an early stage of the disease, the ability to improve survival and quality of life for patients with late stage disease has been limited, largely due to the ability of cancer cells to evade destruction when treatments block preferred paths for survival. Here, we review the role that ions and ion channels play in normal cell function, the development of disease and their role in the life and death of a cell. It is hoped that viewing cancer from the perspective of altered ion channel expression and ion balance may provide a novel approach for developing more effective treatments for this devastating disease. Abstract Life depends upon the ability of cells to evaluate and adapt to a constantly changing environment and to maintain internal stability to allow essential biochemical reactions to occur. Ions and ion channels play a crucial role in this process and are essential for survival. Alterations in the expression of the transmembrane proteins responsible for maintaining ion balance that occur as a result of mutations in the genetic code or in response to iatrogenically induced changes in the extracellular environment is a characteristic feature of oncogenesis and identifies cancer as one of a constellation of diseases known as channelopathies. The classification of cancer as a channelopathy provides a different perspective for viewing the disease. Potentially, it may expand opportunities for developing novel ways to affect or reverse the deleterious changes that underlie establishing and sustaining disease and developing tolerance to therapeutic attempts at treatment. The role of ions and ion channels and their interactions in the cell’s ability to maintain ionic balance, homeostasis, and survival are reviewed and possible approaches that mitigate gain or loss of ion channel function to contribute to new or enhance existing cancer therapies are discussed.
Collapse
|
34
|
Shegay PV, Zabolotneva AA, Shatova OP, Shestopalov AV, Kaprin AD. Evolutionary View on Lactate-Dependent Mechanisms of Maintaining Cancer Cell Stemness and Reprimitivization. Cancers (Basel) 2022; 14:cancers14194552. [PMID: 36230479 PMCID: PMC9559641 DOI: 10.3390/cancers14194552] [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: 08/23/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
The role of lactic acid (lactate) in cell metabolism has been significantly revised in recent decades. Initially, lactic acid was attributed to the role of a toxic end-product of metabolism, with its accumulation in the cell and extracellular space leading to acidosis, muscle pain, and other adverse effects. However, it has now become obvious that lactate is not only a universal fuel molecule and the main substrate for gluconeogenesis but also one of the most ancient metabolites, with a signaling function that has a wide range of regulatory activity. The Warburg effect, described 100 years ago (the intensification of glycolysis associated with high lactate production), which is characteristic of many malignant tumors, confirms the key role of lactate not only in physiological conditions but also in pathologies. The study of lactate’s role in the malignant transformation becomes more relevant in the light of the “atavistic theory of carcinogenesis,” which suggests that tumor cells return to a more primitive hereditary phenotype during microevolution. In this review, we attempt to summarize the accumulated knowledge about the functions of lactate in cell metabolism and its role in the process of carcinogenesis and to consider the possible evolutionary significance of the Warburg effect.
Collapse
Affiliation(s)
- Petr V. Shegay
- Federal State Budget Institution National Medical Research Radiology Center of the Ministry of Healthcare of the Russian Federation, 2nd Botkinsky pas., 3, 125284 Moscow, Russia
| | - Anastasia A. Zabolotneva
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, st. Ostrovityanova, 1, 117997 Moscow, Russia
- Correspondence:
| | - Olga P. Shatova
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, st. Ostrovityanova, 1, 117997 Moscow, Russia
- Faculty of Medicine, RUDN University, st. Miklukho-Maklaya, 6, 117198 Moscow, Russia
| | - Aleksandr V. Shestopalov
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Pirogov Russian National Research Medical University, st. Ostrovityanova, 1, 117997 Moscow, Russia
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Ministry of Health of the Russian Federation, st. Samora Mashela, 1, 117997 Moscow, Russia
| | - Andrei D. Kaprin
- Federal State Budget Institution National Medical Research Radiology Center of the Ministry of Healthcare of the Russian Federation, 2nd Botkinsky pas., 3, 125284 Moscow, Russia
- Faculty of Medicine, RUDN University, st. Miklukho-Maklaya, 6, 117198 Moscow, Russia
| |
Collapse
|
35
|
Inhibition of Colon Cancer Recurrence via Exogenous TRAIL Delivery Using Gel-like Coacervate Microdroplets. Gels 2022; 8:gels8070427. [PMID: 35877512 PMCID: PMC9319433 DOI: 10.3390/gels8070427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 02/04/2023] Open
Abstract
Colon cancer (CC) belongs to the three major malignancies with a high recurrence rate. Therefore, a novel drug delivery system that can prevent CC recurrence while minimizing side effects is needed. Tumor-necrosis-factor-related apoptosis-inducing ligand (TRAIL) has recently been spotlighted as a protein drug that can induce apoptosis of cancer cells specifically. However, its short in vivo half-life is still a challenge to overcome. Hence, in this study, a gel-like mPEGylated coacervate (mPEG-Coa) delivery platform was developed through electrostatic interaction of mPEG-poly(ethylene arginylaspartate diglyceride) (mPEG-PEAD) and heparin for effective protection of cargo TRAIL, subsequently preserving its bioactivity. mPEG-Coa could protect cargo TRAIL against protease. Sustained release was observed for a long-term (14 days). In addition, recurrence of HCT-116 cells was suppressed when cells were treated with TRAIL-loaded mPEG-Coa for 7 days through long-term continuous supply of active TRAIL, whereas re-proliferation occurred in the bolus TRAIL-treated group. Taken together, these results suggest that our gel-like mPEG-Coa could be utilized as a functional delivery platform to suppress CC recurrence by exogenously supplying TRAIL for a long time with a single administration.
Collapse
|
36
|
Rodrigues T, Piccirillo S, Magi S, Preziuso A, Dos Santos Ramos V, Serfilippi T, Orciani M, Maciel Palacio Alvarez M, Luis Dos Santos Tersariol I, Amoroso S, Lariccia V. Control of Ca 2+ and metabolic homeostasis by the Na +/Ca 2+ exchangers (NCXs) in health and disease. Biochem Pharmacol 2022; 203:115163. [PMID: 35803319 DOI: 10.1016/j.bcp.2022.115163] [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: 04/01/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022]
Abstract
Spatial and temporal control of calcium (Ca2+) levels is essential for the background rhythms and responses of living cells to environmental stimuli. Whatever other regulators a given cellular activity may have, localized and wider scale Ca2+ events (sparks, transients, and waves) are hierarchical determinants of fundamental processes such as cell contraction, excitability, growth, metabolism and survival. Different cell types express specific channels, pumps and exchangers to efficiently generate and adapt Ca2+ patterns to cell requirements. The Na+/Ca2+ exchangers (NCXs) in particular contribute to Ca2+ homeostasis by buffering intracellular Ca2+ loads according to the electrochemical gradients of substrate ions - i.e., Ca2+ and sodium (Na+) - and under a dynamic control of redundant regulatory processes. An interesting feature of NCX emerges from the strict relationship that connects transporter activity with cell metabolism: on the one hand NCX operates under constant control of ATP-dependent regulatory processes, on the other hand the ion fluxes generated through NCX provide mechanistic support for the Na+-driven uptake of glutamate and Ca2+ influx to fuel mitochondrial respiration. Proof of concept evidence highlights therapeutic potential of preserving a timed and balanced NCX activity in a growing rate of diseases (including excitability, neurodegenerative, and proliferative disorders) because of an improved ability of stressed cells to safely maintain ion gradients and mitochondrial bioenergetics. Here, we will summarize and review recent works that have focused on the pathophysiological roles of NCXs in balancing the two-way relationship between Ca2+ signals and metabolism.
Collapse
Affiliation(s)
- Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, SP, Brazil.
| | - Silvia Piccirillo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Alessandra Preziuso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vyctória Dos Santos Ramos
- Interdisciplinary Center for Biochemistry Investigation (CIIB), University of Mogi das Cruzes (UMC), Mogi das Cruzes, SP, Brazil
| | - Tiziano Serfilippi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Histology, University "Politecnica delle Marche", Ancona, Italy.
| | - Marcela Maciel Palacio Alvarez
- Department of Biochemistry, São Paulo School of Medicine, Federal University of São Paulo (Unifesp) São Paulo, SP, Brazil
| | | | - Salvatore Amoroso
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche", Ancona, Italy.
| |
Collapse
|
37
|
James AD, Leslie TK, Kaggie JD, Wiggins L, Patten L, Murphy O'Duinn J, Langer S, Labarthe MC, Riemer F, Baxter G, McLean MA, Gilbert FJ, Kennerley AJ, Brackenbury WJ. Sodium accumulation in breast cancer predicts malignancy and treatment response. Br J Cancer 2022; 127:337-349. [PMID: 35462561 PMCID: PMC9296657 DOI: 10.1038/s41416-022-01802-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 03/10/2022] [Accepted: 03/22/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer remains a leading cause of death in women and novel imaging biomarkers are urgently required. Here, we demonstrate the diagnostic and treatment-monitoring potential of non-invasive sodium (23Na) MRI in preclinical models of breast cancer. METHODS Female Rag2-/- Il2rg-/- and Balb/c mice bearing orthotopic breast tumours (MDA-MB-231, EMT6 and 4T1) underwent MRI as part of a randomised, controlled, interventional study. Tumour biology was probed using ex vivo fluorescence microscopy and electrophysiology. RESULTS 23Na MRI revealed elevated sodium concentration ([Na+]) in tumours vs non-tumour regions. Complementary proton-based diffusion-weighted imaging (DWI) linked elevated tumour [Na+] to increased cellularity. Combining 23Na MRI and DWI measurements enabled superior classification accuracy of tumour vs non-tumour regions compared with either parameter alone. Ex vivo assessment of isolated tumour slices confirmed elevated intracellular [Na+] ([Na+]i); extracellular [Na+] ([Na+]e) remained unchanged. Treatment with specific inward Na+ conductance inhibitors (cariporide, eslicarbazepine acetate) did not affect tumour [Na+]. Nonetheless, effective treatment with docetaxel reduced tumour [Na+], whereas DWI measures were unchanged. CONCLUSIONS Orthotopic breast cancer models exhibit elevated tumour [Na+] that is driven by aberrantly elevated [Na+]i. Moreover, 23Na MRI enhances the diagnostic capability of DWI and represents a novel, non-invasive biomarker of treatment response with superior sensitivity compared to DWI alone.
Collapse
Affiliation(s)
- Andrew D James
- Department of Biology, University of York, York, UK
- York Biomedical Research Institute, University of York, York, UK
| | | | - Joshua D Kaggie
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | | | - Lewis Patten
- Department of Mathematics, University of York, York, UK
| | | | - Swen Langer
- Bioscience Technology Facility, Department of Biology, University of York, York, UK
| | | | - Frank Riemer
- Mohn Medical Imaging and Visualization Centre, Haukeland University Hospital Bergen, Bergen, Norway
| | - Gabrielle Baxter
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Mary A McLean
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Fiona J Gilbert
- Department of Radiology & NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Aneurin J Kennerley
- York Biomedical Research Institute, University of York, York, UK
- Department of Chemistry, University of York, York, UK
| | - William J Brackenbury
- Department of Biology, University of York, York, UK.
- York Biomedical Research Institute, University of York, York, UK.
| |
Collapse
|
38
|
Li X, Alu A, Wei Y, Wei X, Luo M. The modulatory effect of high salt on immune cells and related diseases. Cell Prolif 2022; 55:e13250. [PMID: 35747936 PMCID: PMC9436908 DOI: 10.1111/cpr.13250] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The adverse effect of excessive salt intake has been recognized in decades. Researchers have mainly focused on the association between salt intake and hypertension. However, studies in recent years have proposed the existence of extra-renal sodium storage and provided insight into the immunomodulatory function of sodium. OBJECTIVES In this review, we discuss the modulatory effects of high salt on various innate and adaptive immune cells and immune-regulated diseases. METHODS We identified papers through electronic searches of PubMed database from inception to March 2022. RESULTS An increasing body of evidence has demonstrated that high salt can modulate the differentiation, activation and function of multiple immune cells. Furthermore, a high-salt diet can increase tissue sodium concentrations and influence the immune responses in microenvironments, thereby affecting the development of immune-regulated diseases, including hypertension, multiple sclerosis, cancer and infections. These findings provide a novel mechanism for the pathology of certain diseases and indicate that salt might serve as a target or potential therapeutic agent in different disease contexts. CONCLUSION High salt has a profound impact on the differentiation, activation and function of multiple immune cells. Additionally, an HSD can modulate the development of various immune-regulated diseases.
Collapse
Affiliation(s)
- Xian Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Min Luo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Mazzitelli I, Bleichmar L, Melucci C, Gerber PP, Toscanini A, Cuestas ML, Diaz FE, Geffner J. High Salt Induces a Delayed Activation of Human Neutrophils. Front Immunol 2022; 13:831844. [PMID: 35720394 PMCID: PMC9204211 DOI: 10.3389/fimmu.2022.831844] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/11/2022] [Indexed: 01/18/2023] Open
Abstract
High salt (NaCl) concentrations are found in a number of tissues under physiological and pathological conditions. Here, we analyzed the effects induced by high salt on the function of human neutrophils. The culture of neutrophils in medium supplemented with high salt (50 mM NaCl) for short periods (30-120 min) inhibited the ability of conventional agonists to induce the production of IL-8 and the activation of respiratory burst. By contrast, exposure to high salt for longer periods (6-18 h) resulted in the activation of neutrophils revealed by the production of high levels of IL-8, the activation of the respiratory burst, and a marked synergistic effect on the production of TNF-α induced by LPS. Increasing osmolarity of the culture medium by the addition of sorbitol or mannitol (100 mM) was shown to be completely unable to stimulate neutrophil responses, suggesting that high sodium but not an increased osmolarity mediates the activation on neutrophils responses. A similar biphasic effect was observed when the function of monocytes was analyzed. Short term exposure to high salt suppressed IL-8 and TNF-α production induced by LPS while culture for longer periods triggered the production of IL-8 but not TNF-α in the absence of LPS stimulation. Contradictory results have been published regarding how high salt modulates neutrophil function. Our results suggest that the modulation of neutrophil function by high salt is strongly dependent on the exposure time.
Collapse
Affiliation(s)
- Ignacio Mazzitelli
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lucía Bleichmar
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Claudia Melucci
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pehuén Pereyra Gerber
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
| | - Agustina Toscanini
- Microbiología y Parasitología Médica
Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Luján Cuestas
- Microbiología y Parasitología Médica
Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernando Erra Diaz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Consejo Nacional de Investigaciones Cientìficas y Tecnològicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- *Correspondence: Jorge Geffner,
| |
Collapse
|
40
|
Anti-invasive effects of minoxidil on human breast cancer cells: combination with ranolazine. Clin Exp Metastasis 2022; 39:679-689. [PMID: 35643818 PMCID: PMC9338910 DOI: 10.1007/s10585-022-10166-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 04/11/2022] [Indexed: 12/11/2022]
Abstract
A plethora of ion channels have been shown to be involved systemically in the pathophysiology of cancer and ion channel blockers can produce anti-metastatic effects. However, although ion channels are known to frequently function in concerted action, little is known about possible combined effects of ion channel modulators on metastatic cell behaviour. Here, we investigated functional consequences of pharmacologically modulating ATP-gated potassium (KATP) channel and voltage-gated sodium channel (VGSC) activities individually and in combination. Two triple-negative human breast cancer cell lines were used: MDA-MB-231 and MDA-MB-468, the latter mainly for comparison. Most experiments were carried out on hypoxic cells. Electrophysiological effects were studied by whole-cell patch clamp recording. Minoxidil (a KATP channel opener) and ranolazine (a blocker of the VGSC persistent current) had no effect on cell viability and proliferation, alone or in combination. In contrast, invasion was significantly reduced in a dose-dependent manner by clinical concentrations of minoxidil and ranolazine. Combining the two drugs produced significant additive effects at concentrations as low as 0.625 μM ranolazine and 2.5 μM minoxidil. Electrophysiologically, acute application of minoxidil shifted VGSC steady-state inactivation to more hyperpolarised potentials and slowed recovery from inactivation, consistent with inhibition of VGSC activation. We concluded (i) that clinically relevant doses of minoxidil and ranolazine individually could inhibit cellular invasiveness dose dependently and (ii) that their combination was additionally effective. Accordingly, ranolazine, minoxidil and their combination may be repurposed as novel anti-metastatic agents.
Collapse
|
41
|
Abstract
Rapid fluctuations in the plasma membrane potential (Vm) provide the basis underlying the action potential waveform in electrically excitable cells; however, a growing body of literature shows that the Vm is also functionally instructive in nonexcitable cells, including cancer cells. Various ion channels play a key role in setting and fine tuning the Vm in cancer and stromal cells within the tumor microenvironment (TME), raising the possibility that the Vm could be targeted therapeutically using ion channel-modulating compounds. Emerging evidence points to the Vm as a viable therapeutic target, given its functional significance in regulating cell cycle progression, migration, invasion, immune infiltration, and pH regulation. Several compounds are now undergoing clinical trials and there is increasing interest in therapeutic manipulation of the Vm via application of pulsed electric fields. The purpose of this article is to update the reader on the significant recent and ongoing progress to elucidate the functional significance of Vm regulation in tumors, to highlight key remaining questions and the prospect of future therapeutic targeting. In particular, we focus on key developments in understanding the functional consequences of Vm alteration on tumor development via the activation of small GTPase (K-Ras and Rac1) signaling, as well as the impact of Vm changes within the heterogeneous TME on immune cell function and cancer progression.
Collapse
Affiliation(s)
- Ming Yang
- York Biomedical Research Institute, Department of Biology, University of York, Heslington, United Kingdom
| | - William J. Brackenbury
- York Biomedical Research Institute, Department of Biology, University of York, Heslington, United Kingdom
| |
Collapse
|
42
|
Shandell MA, Capatina AL, Lawrence SM, Brackenbury WJ, Lagos D. Inhibition of the Na +/K +-ATPase by cardiac glycosides suppresses expression of the IDO1 immune checkpoint in cancer cells by reducing STAT1 activation. J Biol Chem 2022; 298:101707. [PMID: 35150740 PMCID: PMC8902613 DOI: 10.1016/j.jbc.2022.101707] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/18/2022] Open
Abstract
Despite extensive basic and clinical research on immune checkpoint regulatory pathways, little is known about the effects of the ionic tumor microenvironment on immune checkpoint expression and function. Here we describe a mechanistic link between Na+/K+-ATPase (NKA) inhibition and activity of the immune checkpoint protein indoleamine-pyrrole 2',3'-dioxygenase 1 (IDO1). We found that IDO1 was necessary and sufficient for production of kynurenine, a downstream tryptophan metabolite, in cancer cells. We developed a spectrophotometric assay to screen a library of 31 model ion transport-targeting compounds for potential effects on IDO1 function in A549 lung and MDA-MB-231 breast cancer cells. This revealed that the cardiac glycosides ouabain and digoxin inhibited kynurenine production at concentrations that did not affect cell survival. NKA inhibition by ouabain and digoxin resulted in increased intracellular Na+ levels and downregulation of IDO1 mRNA and protein levels, which was consistent with the reduction in kynurenine levels. Knockdown of ATP1A1, the ɑ1 subunit of the NKA and target of cardiac glycosides, increased Na+ levels to a lesser extent than cardiac glycoside treatment and did not affect IDO1 expression. However, ATP1A1 knockdown significantly enhanced the effect of cardiac glycosides on IDO1 expression and kynurenine production. Mechanistically, we show that cardiac glycoside treatment resulted in curtailing the length of phosphorylation-mediated stabilization of STAT1, a transcriptional regulator of IDO1 expression, an effect enhanced by ATP1A1 knockdown. Our findings reveal cross talk between ionic modulation via cardiac glycosides and immune checkpoint protein expression in cancer cells with broad mechanistic and clinical implications.
Collapse
Affiliation(s)
- Mia A Shandell
- Department of Biology, University of York, York, United Kingdom; Hull York Medical School, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | - Alina L Capatina
- Department of Biology, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | | | - William J Brackenbury
- Department of Biology, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom
| | - Dimitris Lagos
- Hull York Medical School, University of York, York, United Kingdom; York Biomedical Research Institute, University of York, York, United Kingdom.
| |
Collapse
|
43
|
Tomiyasu M, Harada M. In vivo Human MR Spectroscopy Using a Clinical Scanner: Development, Applications, and Future Prospects. Magn Reson Med Sci 2022; 21:235-252. [PMID: 35173095 PMCID: PMC9199975 DOI: 10.2463/mrms.rev.2021-0085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
MR spectroscopy (MRS) is a unique and useful method for noninvasively evaluating biochemical metabolism in human organs and tissues, but its clinical dissemination has been slow and often limited to specialized institutions or hospitals with experts in MRS technology. The number of 3-T clinical MR scanners is now increasing, representing a major opportunity to promote the use of clinical MRS. In this review, we summarize the theoretical background and basic knowledge required to understand the results obtained with MRS and introduce the general consensus on the clinical utility of proton MRS in routine clinical practice. In addition, we present updates to the consensus guidelines on proton MRS published by the members of a working committee of the Japan Society of Magnetic Resonance in Medicine in 2013. Recent research into multinuclear MRS equipped in clinical MR scanners is explained with an eye toward future development. This article seeks to provide an overview of the current status of clinical MRS and to promote the understanding of when it can be useful. In the coming years, MRS-mediated biochemical evaluation is expected to become available for even routine clinical practice.
Collapse
Affiliation(s)
- Moyoko Tomiyasu
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology.,Department of Radiology, Kanagawa Children's Medical Center
| | - Masafumi Harada
- Department of Radiology and Radiation Oncology, Graduate School of Biomedical Sciences, Tokushima University
| |
Collapse
|
44
|
Damanskienė E, Balnytė I, Valančiūtė A, Alonso MM, Preikšaitis A, Stakišaitis D. The Different Temozolomide Effects on Tumorigenesis Mechanisms of Pediatric Glioblastoma PBT24 and SF8628 Cell Tumor in CAM Model and on Cells In Vitro. Int J Mol Sci 2022; 23:ijms23042001. [PMID: 35216113 PMCID: PMC8877228 DOI: 10.3390/ijms23042001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/04/2022] [Accepted: 02/06/2022] [Indexed: 02/05/2023] Open
Abstract
It is necessary to elucidate the individual effects of temozolomide (TMZ) on carcinogenesis and tumor resistance to chemotherapy mechanisms. The study aimed to investigate the TMZ 50 and 100 μM dose effect difference between PBT24 and SF8628 cell line high-grade pediatric glioblastoma (phGBM) xenografts in a chicken chorioallantoic membrane (CAM) model, on PCNA and EZH2 immunohistochemical expression in the tumor and on the expression of NKCC1, KCC2, E- and N-cadherin genes in TMZ-treated and control cell groups in vitro. TMZ at a 100 μg dose reduced the incidence of PBT24 xenograft invasion into the CAM, CAM thickening and the number of blood vessels in the CAM (p < 0.05), but did not affect the SF8628 tumor in the CAM model. The TMZ impact on PBT24 and SF8628 tumor PCNA expression was similarly significantly effective but did not alter EZH2 expression in the studied tumors. The TMZ at 50 μM caused significantly increased RNA expression of the NKCC1 gene in both studied cell types compared with controls (p < 0.05). The expression of the KCC2 gene was increased in PBT24 TMZ-treated cells (p < 0.05), and no TMZ effect was found in SF8628-treated cells. The study supports the suggestion that individual sensitivity to TMZ should be assessed when starting treatment.
Collapse
Affiliation(s)
- Eligija Damanskienė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
- Correspondence: (E.D.); (D.S.)
| | - Ingrida Balnytė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
| | - Angelija Valančiūtė
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
| | - Marta Maria Alonso
- Department of Pediatrics, Clínica Universidad de Navarra, University of Navarra, 31008 Pamplona, Spain;
| | - Aidanas Preikšaitis
- Centre of Neurosurgery, Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania;
| | - Donatas Stakišaitis
- Department of Histology and Embryology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania; (I.B.); (A.V.)
- Laboratory of Molecular Oncology, National Cancer Institute, 08660 Vilnius, Lithuania
- Correspondence: (E.D.); (D.S.)
| |
Collapse
|
45
|
Issitt T, Wiggins L, Veysey M, Sweeney S, Brackenbury W, Redeker K. Volatile compounds in human breath: critical review and meta-analysis. J Breath Res 2022; 16. [PMID: 35120340 DOI: 10.1088/1752-7163/ac5230] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/04/2022] [Indexed: 11/12/2022]
Abstract
Volatile compounds contained in human breath reflect the inner workings of the body. A large number of studies have been published that link individual components of breath to disease, but diagnostic applications remain limited, in part due to inconsistent and conflicting identification of breath biomarkers. New approaches are therefore required to identify effective biomarker targets. Here, volatile organic compounds have been identified in the literature from four metabolically and physiologically distinct diseases and grouped into chemical functional groups (e.g. - methylated hydrocarbons or aldehydes; based on known metabolic and enzymatic pathways) to support biomarker discovery and provide new insight on existing data. Using this functional grouping approach, principal component analysis doubled explanatory capacity from 19.1% to 38% relative to single individual compound approaches. Random forest and linear discriminant analysis reveal 93% classification accuracy for cancer. This review and meta-analysis provides insight for future research design by identifying volatile functional groups associated with disease. By incorporating our understanding of the complexities of the human body, along with accounting for variability in methodological and analytical approaches, this work demonstrates that a suite of targeted, functional volatile biomarkers, rather than individual biomarker compounds, will improve accuracy and success in diagnostic research and application.
Collapse
Affiliation(s)
- Theo Issitt
- Biology, University of York, University of York, York, York, YO10 5DD, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Laura Wiggins
- Biology, University of York, University of York, York, York, YO10 5DD, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Martin Veysey
- The University of Newcastle, School of Medicine & Public Health, Callaghan, New South Wales, 2308, AUSTRALIA
| | - Sean Sweeney
- Biology, University of York, University of York, York, York, YO10 5DD, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - William Brackenbury
- Biology, University of York, University of York, York, York, YO10 5DD, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Kelly Redeker
- Biology, University of York, Biology Dept. University of York, York, York, North Yorkshire, YO10 5DD, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| |
Collapse
|
46
|
Voloaca OM, Clench MR, Koellensperger G, Cole LM, Haywood-Small SL, Theiner S. Elemental Mapping of Human Malignant Mesothelioma Tissue Samples Using High-Speed LA–ICP–TOFMS Imaging. Anal Chem 2022; 94:2597-2606. [PMID: 35073065 PMCID: PMC8829826 DOI: 10.1021/acs.analchem.1c04857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
This
is the first report of the use of laser ablation–inductively
coupled plasma time-of-flight mass spectrometry (LA–ICP–TOFMS)
to analyze human malignant pleural mesothelioma (MPM) samples at the
cellular level. MPM is an aggressive, incurable cancer associated
with asbestos exposure, with a long latency and poor overall survival.
Following careful optimization of the laser fluence, the simultaneous
ablation of soft biological tissue and hard mineral fibers was possible,
allowing the spatial detection of elements such as Si, Mg, Ca, and
Fe, which are also present in the glass substrate. A low-dispersion
LA setup was employed, which provided the high spatial resolution
necessary to identify the asbestos fibers and fiber fragments in the
tissue and to characterize the metallome at the cellular level (a
pixel size of 2 μm), with a high speed (at 250 Hz). The multielement
LA–ICP–TOFMS imaging approach enabled (i) the detection
of asbestos fibers/mineral impurities within the MPM tissue samples
of patients, (ii) the visualization of the tissue structure with the
endogenous elemental pattern at high spatial resolution, and (iii)
obtaining insights into the metallome of MPM patients with different
pathologies in a single analysis run. Asbestos and other mineral fibers
were detected in the lung and pleura tissue of MPM patients, respectively,
based on their multielement pattern (Si, Mg, Ca, Fe, and Sr). Interestingly,
strontium was detected in asbestos fibers, suggesting a link between
this potential toxic element and MPM pathogenesis. Furthermore, monitoring
the metallome around the talc deposit regions (characterized by elevated
levels of Al, Mg, and Si) revealed significant tissue damage and inflammation
caused by talc pleurodesis. LA–ICP–TOFMS results correlated
to Perls’ Prussian blue and histological staining of the corresponding
serial sections. Ultimately, the ultra-high-speed and high-spatial-resolution
capabilities of this novel LA–ICP–TOFMS setup may become
an important clinical tool for simultaneous asbestos detection, metallome
monitoring, and biomarker identification.
Collapse
Affiliation(s)
- Oana M. Voloaca
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 1WB Sheffield, U.K
| | - Malcolm R. Clench
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 1WB Sheffield, U.K
| | - Gunda Koellensperger
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria
| | - Laura M. Cole
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 1WB Sheffield, U.K
| | - Sarah L. Haywood-Small
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, S1 1WB Sheffield, U.K
| | - Sarah Theiner
- Institute of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria
| |
Collapse
|
47
|
Hyder F, Khan MH. Dysregulated proton and sodium gradients highlight cancer invasion and proliferation. Transl Oncol 2021; 16:101310. [PMID: 34896853 PMCID: PMC8681017 DOI: 10.1016/j.tranon.2021.101310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/15/2021] [Accepted: 12/05/2021] [Indexed: 11/29/2022] Open
Abstract
The hallmarks of cancer all directly linked to proton and sodium imbalances, where reduced extracellular pH and salinity are both the resultant of oncogenic metabolic changes in the tumor microenvironment and are supported by upregulated cellular mechanisms like carbonic anhydrase 9 (CA IX) and sodium proton exchanger (NHE). The dysregulated proton and sodium gradients highlight cancer invasion and proliferation and can be imaged by merging of 23Na and 1H magnetic resonance techniques.
Acidity and salinity of the extracellular fluid, reflecting degrees of acid and sodium contents respectively, regulate essential cellular functions in health and disease, especially cancer. Tumor invasiveness is enhanced by the acidic extracellular milieu as a consequence of upregulated aerobic glycolysis. But recent discoveries also suggest that enhanced proliferative mitosis, which also hallmarks cancer, is impacted by interstitial salinity. Abnormal transmembrane proton/sodium gradients lead to pathophysiological alterations at the cellular level. These novel perspectives mandate pioneering imaging of extracellular acidity and salinity, preferably monitored simultaneously. By dissecting the interplay between dysregulated pH and electrolyte imbalance within the tumor habitat, these biomarkers hold promise for early cancer diagnosis and tracking therapies, from chemotherapy to immunotherapy.
Collapse
Affiliation(s)
- Fahmeed Hyder
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America; Department of Radiology & Biomedical Imaging, Yale University, New Haven, CT, United States of America; Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, United States of America; Quantitative Neuroimaging with Magnetic Resonance (QNMR) Research Program, Yale University, New Haven, CT, United States of America.
| | - Muhammad H Khan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America; Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, United States of America
| |
Collapse
|
48
|
Fuchs E, Messerer DAC, Karpel-Massler G, Fauler M, Zimmer T, Jungwirth B, Föhr KJ. Block of Voltage-Gated Sodium Channels as a Potential Novel Anti-cancer Mechanism of TIC10. Front Pharmacol 2021; 12:737637. [PMID: 34744721 PMCID: PMC8567104 DOI: 10.3389/fphar.2021.737637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/15/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Tumor therapeutics are aimed to affect tumor cells selectively while sparing healthy ones. For this purpose, a huge variety of different drugs are in use. Recently, also blockers of voltage-gated sodium channels (VGSCs) have been recognized to possess potentially beneficial effects in tumor therapy. As these channels are a frequent target of numerous drugs, we hypothesized that currently used tumor therapeutics might have the potential to block VGSCs in addition to their classical anti-cancer activity. In the present work, we have analyzed the imipridone TIC10, which belongs to a novel class of anti-cancer compounds, for its potency to interact with VGSCs. Methods: Electrophysiological experiments were performed by means of the patch-clamp technique using heterologously expressed human heart muscle sodium channels (hNav1.5), which are among the most common subtypes of VGSCs occurring in tumor cells. Results: TIC10 angular inhibited the hNav1.5 channel in a state- but not use-dependent manner. The affinity for the resting state was weak with an extrapolated Kr of about 600 μM. TIC10 most probably did not interact with fast inactivation. In protocols for slow inactivation, a half-maximal inhibition occurred around 2 µM. This observation was confirmed by kinetic studies indicating that the interaction occurred with a slow time constant. Furthermore, TIC10 also interacted with the open channel with an affinity of approximately 4 µM. The binding site for local anesthetics or a closely related site is suggested as a possible target as the affinity for the well-characterized F1760K mutant was reduced more than 20-fold compared to wild type. Among the analyzed derivatives, ONC212 was similarly effective as TIC10 angular, while TIC10 linear more selectively interacted with the different states. Conclusion: The inhibition of VGSCs at low micromolar concentrations might add to the anti-tumor properties of TIC10.
Collapse
Affiliation(s)
- Eva Fuchs
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | | | | | - Michael Fauler
- Institute of General Physiology, University of Ulm, Ulm, Germany
| | - Thomas Zimmer
- Institute of Physiology, University Hospital of Jena, Jena, Germany
| | - Bettina Jungwirth
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| | - Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
49
|
Hagiwara A, Bydder M, Oughourlian TC, Yao J, Salamon N, Jahan R, Villablanca JP, Enzmann DR, Ellingson BM. Sodium MR Neuroimaging. AJNR Am J Neuroradiol 2021; 42:1920-1926. [PMID: 34446457 PMCID: PMC8583254 DOI: 10.3174/ajnr.a7261] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022]
Abstract
Sodium MR imaging has the potential to complement routine proton MR imaging examinations with the goal of improving diagnosis, disease characterization, and clinical monitoring in neurologic diseases. In the past, the utility and exploration of sodium MR imaging as a valuable clinical tool have been limited due to the extremely low MR signal, but with recent improvements in imaging techniques and hardware, sodium MR imaging is on the verge of becoming clinically realistic for conditions that include brain tumors, ischemic stroke, and epilepsy. In this review, we briefly describe the fundamental physics of sodium MR imaging tailored to the neuroradiologist, focusing on the basics necessary to understand factors that play into making sodium MR imaging feasible for clinical settings and describing current controversies in the field. We will also discuss the current state of the field and the potential future clinical uses of sodium MR imaging in the diagnosis, phenotyping, and therapeutic monitoring in neurologic diseases.
Collapse
Affiliation(s)
- A Hagiwara
- From the UCLA Brain Tumor Imaging Laboratory (A.H., M.B., T.C.O., J.Y., B.M.E.), Center for Computer Vision and Imaging Biomarkers
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - M Bydder
- From the UCLA Brain Tumor Imaging Laboratory (A.H., M.B., T.C.O., J.Y., B.M.E.), Center for Computer Vision and Imaging Biomarkers
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - T C Oughourlian
- From the UCLA Brain Tumor Imaging Laboratory (A.H., M.B., T.C.O., J.Y., B.M.E.), Center for Computer Vision and Imaging Biomarkers
- Neuroscience Interdepartmental Program (T.C.O., B.M.E.)
| | - J Yao
- From the UCLA Brain Tumor Imaging Laboratory (A.H., M.B., T.C.O., J.Y., B.M.E.), Center for Computer Vision and Imaging Biomarkers
- Department of Bioengineering (J.Y., B.M.E.), Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, California
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - N Salamon
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - R Jahan
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - J P Villablanca
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - D R Enzmann
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
| | - B M Ellingson
- From the UCLA Brain Tumor Imaging Laboratory (A.H., M.B., T.C.O., J.Y., B.M.E.), Center for Computer Vision and Imaging Biomarkers
- Department of Bioengineering (J.Y., B.M.E.), Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, California
- Department of Radiological Sciences (A.H., M.B., J.Y., N.S., R.J., J.P.V., D.R.E., B.M.E.)
- Neuroscience Interdepartmental Program (T.C.O., B.M.E.)
- Department of Psychiatry and Biobehavioral Sciences (B.M.E.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
50
|
Wu Y, Li Z, Shi M, Yuan K, Meng HM, Qu L, Li Z. Programmable DNAzyme Computing for Specific In Vivo Imaging: Intracellular Stimulus-Unlocked Target Sensing and Signal Amplification. Anal Chem 2021; 93:12456-12463. [PMID: 34449199 DOI: 10.1021/acs.analchem.1c02699] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Molecular probe that enables in vivo imaging is the cornerstone of accurate disease diagnosis, prognostic estimation, and therapies. Although several nucleic acid-based probes have been reported for tumor detection, it is still a challenge to develop programmable methodology for accurately identifying tumors in vivo. Herein, a reconfigurable DNA hybridization-based reaction was constructed to assemble DNAzyme computing that contains an intracellular miRNA-unlocked entropy-driven catalysis module and an endogenous metal ion-responsive DNAzyme module for specific in vivo imaging. By reasonable design, the programmable DNAzyme computing can not only successfully distinguish tumor cells from normal cells but also enable tumor imaging in living mice. Due to its excellent operation with high specificity and sensitivity, this design may be broadly applied in the biological study and personalized medicine.
Collapse
Affiliation(s)
- Yanan Wu
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Zhijun Li
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Mingqing Shi
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Kun Yuan
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Hong-Min Meng
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Lingbo Qu
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| | - Zhaohui Li
- College of Chemistry, Green Catalysis Center, Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou Key Laboratory of Functional Nanomaterial and Medical Theranostic, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|