1
|
Bloomer H, Dame HB, Parker SR, Oudin MJ. Neuronal mimicry in tumors: lessons from neuroscience to tackle cancer. Cancer Metastasis Rev 2025; 44:31. [PMID: 39934425 DOI: 10.1007/s10555-025-10249-3] [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] [Academic Contribution Register] [Received: 10/21/2024] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Cellular plasticity and the ability to avoid terminal differentiation are hallmarks of cancer. Here, we review the evidence that tumor cells themselves can take on properties of neurons of the central nervous system, which can regulate tumor growth and metastasis. We discuss recent evidence that axon guidance molecules and regulators of electrical activity and synaptic transmission, such as ion channels and neurotransmitters, can drive the oncogenic and invasive properties of tumor cells from a range of cancers. We also review how FDA-approved treatments for neurological disorders are being tested in pre-clinical models and clinical trials for repurposing as anti-cancer agents, offering the potential for new therapies for cancer patients that can be accessed more quickly.
Collapse
Affiliation(s)
- Hanan Bloomer
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Haley B Dame
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Savannah R Parker
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
2
|
Vasylyev DV, Liu CJ, Waxman SG. Sodium channels in non-excitable cells: powerful actions and therapeutic targets beyond Hodgkin and Huxley. Trends Cell Biol 2024:S0962-8924(24)00251-4. [PMID: 39743470 DOI: 10.1016/j.tcb.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/03/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Voltage-gated sodium channels (VGSCs) are best known for their role in the generation and propagation of action potentials in neurons, muscle cells, and cardiac myocytes, which have traditionally been labeled as 'excitable'. However, emerging evidence challenges this traditional perspective. It is now clear that VGSCs are also expressed in a broad spectrum of cells outside the neuromuscular realm, where they regulate diverse cellular functions. In this review, we summarize current knowledge on the expression, regulation, and function of VGSCs in non-neuromuscular cells, highlighting their contributions to physiological processes and pathological conditions. Dynamic expression patterns of VGSCs in different cell types, involvement of VGSCs in cellular functions, such as phagocytosis, motility, and cytokine release, and their potential as therapeutic targets for diseases that include inflammatory disorders, osteoarthritis (OA), and cancer, are discussed. This new understanding of VGSCs and their effects on cells outside the neuromuscular realm opens new avenues for research and therapeutic interventions.
Collapse
Affiliation(s)
- Dmytro V Vasylyev
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience & Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA; Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.
| |
Collapse
|
3
|
Djamgoz MBA. Electrical excitability of cancer cells-CELEX model updated. Cancer Metastasis Rev 2024; 43:1579-1591. [PMID: 38976181 PMCID: PMC11554705 DOI: 10.1007/s10555-024-10195-6] [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] [Academic Contribution Register] [Received: 04/27/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
The normal functioning of every cell in the body depends on its bioelectric properties and many diseases are caused by genetic and/or epigenetic dysregulation of the underlying ion channels. Metastasis, the main cause of death from cancer, is a complex multi-stage process in which cells break away from a primary tumour, invade the surrounding tissues, enter the circulation by encountering a blood vessel and spread around the body, ultimately lodging in distant organs and reproliferating to form secondary tumours leading to devastating organ failure. Such cellular behaviours are well known to involve ion channels. The CELEX model offers a novel insight to metastasis where it is the electrical excitation of the cancer cells that is responsible for their aggressive and invasive behaviour. In turn, the hyperexcitability is underpinned by concomitant upregulation of functional voltage-gated sodium channels and downregulation of voltage-gated potassium channels. Here, we update the in vitro and in vivo evidence in favour of the CELEX model for carcinomas. The results are unequivocal for the sodium channel. The potassium channel arm is also broadly supported by existing evidence although these data are complicated by the impact of the channels on the membrane potential and consequent secondary effects. Finally, consistent with the CELEX model, we show (i) that carcinomas are indeed electrically excitable and capable of generating action potentials and (ii) that combination of a sodium channel inhibitor and a potassium channel opener can produce a strong, additive anti-invasive effect. We discuss the possible clinical implications of the CELEX model in managing cancer.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
4
|
Liu H, Weng J, Huang CLH, Jackson AP. Voltage-gated sodium channels in cancers. Biomark Res 2024; 12:70. [PMID: 39060933 PMCID: PMC11282680 DOI: 10.1186/s40364-024-00620-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Voltage-gated sodium channels (VGSCs) initiate action potentials in electrically excitable cells and tissues. Surprisingly, some VGSC genes are aberrantly expressed in a variety of cancers, derived from "non-excitable" tissues that do not generate classic action potentials, showing potential as a promising pharmacological target for cancer. Most of the previous review articles on this topic are limited in scope, and largely unable to provide researchers with a comprehensive understanding of the role of VGSC in cancers. Here, we review the expression patterns of all nine VGSC α-subunit genes (SCN1A-11A) and their four regulatory β-subunit genes (SCN1B-4B). We reviewed data from the Cancer Genome Atlas (TCGA) database, complemented by an extensive search of the published papers. We summarized and reviewed previous independent studies and analyzed the VGSC genes in the TCGA database regarding the potential impact of VGSC on cancers. A comparison between evidence gathered from independent studies and data review was performed to scrutinize potential biases in prior research and provide insights into future research directions. The review supports the view that VGSCs play an important role in diagnostics as well as therapeutics of some cancer types, such as breast, colon, prostate, and lung cancer. This paper provides an overview of the current knowledge on voltage-gated sodium channels in cancer, as well as potential avenues for further research. While further research is required to fully understand the role of VGSCs in cancer, the potential of VGSCs for clinical diagnosis and treatment is promising.
Collapse
Affiliation(s)
- Hengrui Liu
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| | - Jieling Weng
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Christopher L-H Huang
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Antony P Jackson
- Department of Biochemistry, Hopkins Building, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, UK.
| |
Collapse
|
5
|
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] [Academic Contribution 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
|
6
|
Rauch DA, Ramos PV, Khanfar M, Harding J, Joseph A, Griffith O, Griffith M, Ratner L. Single-Cell Transcriptomic Analysis of Kaposi Sarcoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592010. [PMID: 38746135 PMCID: PMC11092626 DOI: 10.1101/2024.05.01.592010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 05/16/2024]
Abstract
Kaposi Sarcoma (KS) is a complex tumor caused by KS-associated herpesvirus 8 (KSHV). Histological analysis reveals a mixture of "spindle cells", vascular-like spaces, extravasated erythrocytes, and immune cells. In order to elucidate the infected and uninfected cell types in KS tumors, we examined skin and blood samples from twelve subjects by single cell RNA sequence analyses. Two populations of KSHV-infected cells were identified, one of which represented a proliferative fraction of lymphatic endothelial cells, and the second represented an angiogenic population of vascular endothelial tip cells. Both infected clusters contained cells expressing lytic and latent KSHV genes. Novel cellular biomarkers were identified in the KSHV infected cells, including the sodium channel SCN9A. The number of KSHV positive tumor cells was found to be in the 6% range in HIV-associated KS, correlated inversely with tumor-infiltrating immune cells, and was reduced in biopsies from HIV-negative individuals. T-cell receptor clones were expanded in KS tumors and blood, although in differing magnitudes. Changes in cellular composition in KS tumors were identified in subjects treated with antiretroviral therapy alone, or immunotherapy. These studies demonstrate the feasibility of single cell analyses to identify prognostic and predictive biomarkers. Author Summary Kaposi sarcoma (KS) is a malignancy caused by the KS-associated herpesvirus (KSHV) that causes skin lesions, and may also be found in lymph nodes, lungs, gastrointestinal tract, and other organs in immunosuppressed individuals more commonly than immunocompetent subjects. The current study examined gene expression in single cells from the tumor and blood of these subjects, and identified the characteristics of the complex mixtures of cells in the tumor. This method also identified differences in KSHV gene expression in different cell types and associated cellular genes expressed in KSHV infected cells. In addition, changes in the cellular composition could be elucidated with therapeutic interventions.
Collapse
|
7
|
Xiao P, Li C, Liu Y, Gao Y, Liang X, Liu C, Yang W. The role of metal ions in the occurrence, progression, drug resistance, and biological characteristics of gastric cancer. Front Pharmacol 2024; 15:1333543. [PMID: 38370477 PMCID: PMC10869614 DOI: 10.3389/fphar.2024.1333543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/05/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Metal ions exert pivotal functions within the human body, encompassing essential roles in upholding cell structure, gene expression regulation, and catalytic enzyme activity. Additionally, they significantly influence various pathways implicated in divergent mechanisms of cell death. Among the prevailing malignant tumors of the digestive tract worldwide, gastric cancer stands prominent, exhibiting persistent high mortality rates. A compelling body of evidence reveals conspicuous ion irregularities in tumor tissues, encompassing gastric cancer. Notably, metal ions have been observed to elicit distinct contributions to the progression, drug resistance, and biological attributes of gastric cancer. This review consolidates pertinent literature on the involvement of metal ions in the etiology and advancement of gastric cancer. Particular attention is directed towards metal ions, namely, Na, K, Mg, Ca, Fe, Cu, Zn, and Mn, elucidating their roles in the initiation and progression of gastric cancer, cellular demise processes, drug resistance phenomena, and therapeutic approaches.
Collapse
Affiliation(s)
- Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Gao
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaojing Liang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
8
|
Bahcheli AT, Min HK, Bayati M, Zhao H, Fortuna A, Dong W, Dzneladze I, Chan J, Chen X, Guevara-Hoyer K, Dirks PB, Huang X, Reimand J. Pan-cancer ion transport signature reveals functional regulators of glioblastoma aggression. EMBO J 2024; 43:196-224. [PMID: 38177502 PMCID: PMC10897389 DOI: 10.1038/s44318-023-00016-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] [Academic Contribution Register] [Received: 05/05/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Ion channels, transporters, and other ion-flux controlling proteins, collectively comprising the "ion permeome", are common drug targets, however, their roles in cancer remain understudied. Our integrative pan-cancer transcriptome analysis shows that genes encoding the ion permeome are significantly more often highly expressed in specific subsets of cancer samples, compared to pan-transcriptome expectations. To enable target selection, we identified 410 survival-associated IP genes in 33 cancer types using a machine-learning approach. Notably, GJB2 and SCN9A show prominent expression in neoplastic cells and are associated with poor prognosis in glioblastoma, the most common and aggressive brain cancer. GJB2 or SCN9A knockdown in patient-derived glioblastoma cells induces transcriptome-wide changes involving neuron projection and proliferation pathways, impairs cell viability and tumor sphere formation in vitro, perturbs tunneling nanotube dynamics, and extends the survival of glioblastoma-bearing mice. Thus, aberrant activation of genes encoding ion transport proteins appears as a pan-cancer feature defining tumor heterogeneity, which can be exploited for mechanistic insights and therapy development.
Collapse
Affiliation(s)
- Alexander T Bahcheli
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Hyun-Kee Min
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Masroor Bayati
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Hongyu Zhao
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Neurosurgery and Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Alexander Fortuna
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Weifan Dong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Irakli Dzneladze
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Jade Chan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xin Chen
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kissy Guevara-Hoyer
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Cancer Immunomonitoring and Immuno-Mediated Pathologies Support Unit, Department of Clinical Immunology, Institute of Laboratory Medicine (IML) and Biomedical Research Foundation (IdiSCC), San Carlos Clinical Hospital, Madrid, Spain
| | - Peter B Dirks
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xi Huang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada.
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Gomez F, Fisk B, McMichael JF, Mosior M, Foltz JA, Skidmore ZL, Duncavage EJ, Miller CA, Abel H, Li YS, Russler-Germain DA, Krysiak K, Watkins MP, Ramirez CA, Schmidt A, Martins Rodrigues F, Trani L, Khanna A, Wagner JA, Fulton RS, Fronick CC, O'Laughlin MD, Schappe T, Cashen AF, Mehta-Shah N, Kahl BS, Walker J, Bartlett NL, Griffith M, Fehniger TA, Griffith OL. Ultra-Deep Sequencing Reveals the Mutational Landscape of Classical Hodgkin Lymphoma. CANCER RESEARCH COMMUNICATIONS 2023; 3:2312-2330. [PMID: 37910143 PMCID: PMC10648575 DOI: 10.1158/2767-9764.crc-23-0140] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/30/2023] [Revised: 07/27/2023] [Accepted: 10/24/2023] [Indexed: 11/03/2023]
Abstract
The malignant Hodgkin and Reed Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) are scarce in affected lymph nodes, creating a challenge to detect driver somatic mutations. As an alternative to cell purification techniques, we hypothesized that ultra-deep exome sequencing would allow genomic study of HRS cells, thereby streamlining analysis and avoiding technical pitfalls. To test this, 31 cHL tumor/normal pairs were exome sequenced to approximately 1,000× median depth of coverage. An orthogonal error-corrected sequencing approach verified >95% of the discovered mutations. We identified mutations in genes novel to cHL including: CDH5 and PCDH7, novel stop gain mutations in IL4R, and a novel pattern of recurrent mutations in pathways regulating Hippo signaling. As a further application of our exome sequencing, we attempted to identify expressed somatic single-nucleotide variants (SNV) in single-nuclei RNA sequencing (snRNA-seq) data generated from a patient in our cohort. Our snRNA analysis identified a clear cluster of cells containing a somatic SNV identified in our deep exome data. This cluster has differentially expressed genes that are consistent with genes known to be dysregulated in HRS cells (e.g., PIM1 and PIM3). The cluster also contains cells with an expanded B-cell clonotype further supporting a malignant phenotype. This study provides proof-of-principle that ultra-deep exome sequencing can be utilized to identify recurrent mutations in HRS cells and demonstrates the feasibility of snRNA-seq in the context of cHL. These studies provide the foundation for the further analysis of genomic variants in large cohorts of patients with cHL. SIGNIFICANCE Our data demonstrate the utility of ultra-deep exome sequencing in uncovering somatic variants in Hodgkin lymphoma, creating new opportunities to define the genes that are recurrently mutated in this disease. We also show for the first time the successful application of snRNA-seq in Hodgkin lymphoma and describe the expression profile of a putative cluster of HRS cells in a single patient.
Collapse
Affiliation(s)
- Felicia Gomez
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | - Bryan Fisk
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Joshua F. McMichael
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Matthew Mosior
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Jennifer A. Foltz
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Zachary L. Skidmore
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Eric J. Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Christopher A. Miller
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Haley Abel
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Yi-Shan Li
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - David A. Russler-Germain
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Kilannin Krysiak
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Marcus P. Watkins
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Cody A. Ramirez
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Alina Schmidt
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Fernanda Martins Rodrigues
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Lee Trani
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Ajay Khanna
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Julia A. Wagner
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Robert S. Fulton
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Catrina C. Fronick
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Michelle D. O'Laughlin
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Timothy Schappe
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Amanda F. Cashen
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Neha Mehta-Shah
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Brad S. Kahl
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Jason Walker
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Nancy L. Bartlett
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
| | - Malachi Griffith
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| | - Todd A. Fehniger
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | - Obi L. Griffith
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St Louis, Missouri
- McDonnell Genome Institute, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
10
|
Bian Y, Tuo J, He L, Li W, Li S, Chu H, Zhao Y. Voltage-gated sodium channels in cancer and their specific inhibitors. Pathol Res Pract 2023; 251:154909. [PMID: 37939447 DOI: 10.1016/j.prp.2023.154909] [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] [Academic Contribution Register] [Received: 02/19/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
Voltage-gated sodium channels (VGSCs) participate in generating and spreading action potentials in electrically excited cells such as neurons and muscle fibers. Abnormal expression of VGSCs has been observed in various types of tumors, while they are either not expressed or expressed at a low level in the matching normal tissue. Hence, this abnormal expression suggests that VGSCs confer some advantage or viability on tumor cells, making them a valuable indicator for identifying tumor cells. In addition, overexpression of VGSCs increased the ability of cancer cells to metastasize and invade, as well as correlated with the metastatic behavior of different cancers. Therefore, blocking VGSCs presents a new strategy for the treatment of cancers. A portion of this review summarizes the structure and function of VGSCs and also describes the correlation between VGSCs and cancers. Most importantly, we provide an overview of current research on various subtype-selective VGSC inhibitors and updates on ongoing clinical studies.
Collapse
Affiliation(s)
- Yuan Bian
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Jiale Tuo
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Liangpeng He
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Wenwen Li
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China
| | - Shangxiao Li
- School of Medical Devices, Shenyang Pharmaceutical University, Benxi, Liaoning 117004, PR China
| | - Huiying Chu
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yongshan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, PR China.
| |
Collapse
|
11
|
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] [Grants] [Track Full Text] [Subscribe] [Academic Contribution 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
|
12
|
Banderali U, Moreno M, Martina M. The elusive Na v1.7: From pain to cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:47-69. [PMID: 38007269 DOI: 10.1016/bs.ctm.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/27/2023]
Abstract
Voltage-gated sodium channels (Nav) are protein complexes that play fundamental roles in the transmission of signals in the nervous system, at the neuromuscular junction and in the heart. They are mainly present in excitable cells where they are responsible for triggering action potentials. Dysfunctions in Nav ion conduction give rise to a wide range of conditions, including neurological disorders, hypertension, arrhythmia, pain and cancer. Nav family 1 is composed of nine members, named numerically from 1 to 9. A Nax family also exists and is involved in body-fluid homeostasis. Of particular interest is Nav1.7 which is highly expressed in the sensory neurons of the dorsal root ganglions, where it is involved in the propagation of pain sensation. Gain-of-function mutations in Nav1.7 cause pathologies associated with increased pain sensitivity, while loss-of-function mutations cause reduced sensitivity to pain. The last decade has seen considerable effort in developing highly specific Nav1.7 blockers as pain medications, nonetheless, sufficient efficacy has yet to be achieved. Evidence is now conclusively showing that Navs are also present in many types of cancer cells, where they are involved in cell migration and invasiveness. Nav1.7 is anomalously expressed in endometrial, ovarian and lung cancers. Nav1.7 is also involved in Chemotherapy Induced Peripheral Neuropathy (CIPN). We propose that the knowledge and tools developed to study the role of Nav1.7 in pain can be exploited to develop novel cancer therapies. In this chapter, we illustrate the various aspects of Nav1.7 function in pain, cancer and CIPN, and outline therapeutic approaches.
Collapse
Affiliation(s)
- Umberto Banderali
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada.
| | - Maria Moreno
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| | - Marzia Martina
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal road, Ottawa, ON, Canada
| |
Collapse
|
13
|
Wang B, Zhao L, Yang C, Lin Y, Wang S, Ye Y, Luo J, Shen Z. IDH1 K224 acetylation promotes colorectal cancer via miR-9-5p/NHE1 axis-mediated regulation of acidic microenvironment. iScience 2023; 26:107206. [PMID: 37456829 PMCID: PMC10339209 DOI: 10.1016/j.isci.2023.107206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/25/2022] [Revised: 04/10/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
The acidic microenvironment is considered an important factor in colorectal cancer (CRC) that contributes to malignant transformation. However, the underlying mechanism remains unclear. In a previous study, we confirmed that IDH1 K224 deacetylation promotes enzymatic activity and the production of α-KG. Here, we further investigate the effect of IDH1 hyperacetylation on the CRC acidic microenvironment. We demonstrate that increased α-KG affects hydroxylation of Ago2 and mediates miR-9-5p targeting NHE1 protein. Knockdown of NHE1 dramatically attenuates CRC cell proliferation and migration by restricting transport of intracellular H+ out of cells. Furthermore, we show that miR-9-5p is the microRNA with the most significant difference in the alteration of IDH1 K224 acetylation and can downregulate NHE1 mRNA. Our data also indicate that hydroxylation stabilizes Ago2, which in turn promotes miR-9-5p activity. Taken together, our results reveal a novel mechanism through which IDH1 deacetylation regulates the cellular acidic microenvironment and inhibits CRC metastasis.
Collapse
Affiliation(s)
- Bo Wang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| | - Long Zhao
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| | - Changjiang Yang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| | - Yilin Lin
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| | - Shan Wang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| | - Jianyuan Luo
- Department of Medical Genetics, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, No.11 Xizhimen South Street, Beijing 100044, P.R. China
| |
Collapse
|
14
|
Sanchez-Sandoval AL, Hernández-Plata E, Gomora JC. Voltage-gated sodium channels: from roles and mechanisms in the metastatic cell behavior to clinical potential as therapeutic targets. Front Pharmacol 2023; 14:1206136. [PMID: 37456756 PMCID: PMC10348687 DOI: 10.3389/fphar.2023.1206136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
During the second half of the last century, the prevalent knowledge recognized the voltage-gated sodium channels (VGSCs) as the proteins responsible for the generation and propagation of action potentials in excitable cells. However, over the last 25 years, new non-canonical roles of VGSCs in cancer hallmarks have been uncovered. Their dysregulated expression and activity have been associated with aggressive features and cancer progression towards metastatic stages, suggesting the potential use of VGSCs as cancer markers and prognostic factors. Recent work has elicited essential information about the signalling pathways modulated by these channels: coupling membrane activity to transcriptional regulation pathways, intracellular and extracellular pH regulation, invadopodia maturation, and proteolytic activity. In a promising scenario, the inhibition of VGSCs with FDA-approved drugs as well as with new synthetic compounds, reduces cancer cell invasion in vitro and cancer progression in vivo. The purpose of this review is to present an update regarding recent advances and ongoing efforts to have a better understanding of molecular and cellular mechanisms on the involvement of both pore-forming α and auxiliary β subunits of VGSCs in the metastatic processes, with the aim at proposing VGSCs as new oncological markers and targets for anticancer treatments.
Collapse
Affiliation(s)
- Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Medicina Genómica, Hospital General de México “Dr Eduardo Liceaga”, Mexico City, Mexico
| | - Everardo Hernández-Plata
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
15
|
Pukkanasut P, Whitt J, Guenter R, Lynch SE, Gallegos C, Rosendo-Pineda MJ, Gomora JC, Chen H, Lin D, Sorace A, Jaskula-Sztul R, Velu SE. Voltage-Gated Sodium Channel Na V1.7 Inhibitors with Potent Anticancer Activities in Medullary Thyroid Cancer Cells. Cancers (Basel) 2023; 15:2806. [PMID: 37345144 PMCID: PMC10216335 DOI: 10.3390/cancers15102806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/13/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Our results from quantitative RT-PCR, Western blotting, immunohistochemistry, and the tissue microarray of medullary thyroid cancer (MTC) cell lines and patient specimens confirm that VGSC subtype NaV1.7 is uniquely expressed in aggressive MTC and not expressed in normal thyroid cells and tissues. We establish the druggability of NaV1.7 in MTC by identifying a novel inhibitor (SV188) and investigate its mode of binding and ability to inhibit INa current in NaV1.7. The whole-cell patch-clamp studies of the SV188 in the NaV1.7 channels expressed in HEK-293 cells show that SV188 inhibited the INa current in NaV1.7 with an IC50 value of 3.6 µM by a voltage- and use-dependent blockade mechanism, and the maximum inhibitory effect is observed when the channel is open. SV188 inhibited the viability of MTC cell lines, MZ-CRC-1 and TT, with IC50 values of 8.47 μM and 9.32 μM, respectively, and significantly inhibited the invasion of MZ-CRC-1 cells by 35% and 52% at 3 μM and 6 μM, respectively. In contrast, SV188 had no effect on the invasion of TT cells derived from primary tumor, which have lower basal expression of NaV1.7. In addition, SV188 at 3 μM significantly inhibited the migration of MZ-CRC-1 and TT cells by 27% and 57%, respectively.
Collapse
Affiliation(s)
- Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Jason Whitt
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
| | - Rachael Guenter
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
| | - Shannon E. Lynch
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
| | - Carlos Gallegos
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Margarita Jacaranda Rosendo-Pineda
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.J.R.-P.); (J.C.G.)
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (M.J.R.-P.); (J.C.G.)
| | - Herbert Chen
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Diana Lin
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Anna Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (C.G.)
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Renata Jaskula-Sztul
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; (J.W.); (R.G.); (H.C.)
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
16
|
Shi H, Duan J, Chen Z, Huang M, Han W, Kong R, Guan X, Qi Z, Zheng S, Lu M. A prognostic gene signature for gastric cancer and the immune infiltration-associated mechanism underlying the signature gene, PLG. Clin Transl Oncol 2023; 25:995-1010. [PMID: 36376702 DOI: 10.1007/s12094-022-03003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Globally, gastric cancer (GC) is a common and lethal solid malignant tumor. Identifying the molecular signature and its functions can provide mechanistic insights into GC development and new methods for targeted therapy. METHODS Differentially expressed genes (DEGs) and prognostic genes (from univariate Cox regression analysis) were overlapped to obtain prognostic DEGs. Subsequently, molecular modules and the functions of these prognostic DEGs were identified by Metascape and Gene Ontology (GO)/Kyoto Encyclopedia of Genes and Genomes (KEGG)/Gene Set Enrichment Analysis (GSEA) enrichment analyses, respectively. Protein-protein interaction (PPI) networks of up- and down-regulated prognostic DEGs in GC were analyzed using the MCC algorithm of the Cytohubba plug-in in Cytoscape. The prognostic gene signature was defined on hub genes of the PPI networks by least absolute shrinkage and selection operator (LASSO)-Cox regression analysis. Furthermore, the expressional level of PLG in our clinical GC samples was validated by quantitative PCR (qPCR), western blotting, and immunohistochemistry (IHC). Subsequently, the PLG expression-correlation analysis was performed to assess the role of PLG in GC progression. Immune infiltration analysis was performed by single-sample gene set enrichment analysis (ssGSEA) to assess the inhibitory effect of PLG on immune infiltration. RESULTS Firstly, Up- and down-regulated prognostic DEGs and hub genes in protein-protein interaction (PPI) networks in GC were identified. A prognostic five-gene signature (i.e., PLG, SPARC, FGB, SERPINE1, and KLHL41) was identified. Among the five genes, the relationship between plasminogen (PLG) and GC remains largely unclear. Moreover, the functions of PLG-correlated genes in GC, like 'fibrinolysis', 'hemostasis', 'ion channel complex', and 'transporter complex' were identified. In addition, PLG expression correlated negatively with the infiltration of almost all immune cell types. Interestingly, the expression of PLG was significantly and highly correlated with that of CD160, an immune checkpoint inhibitor. CONCLUSION Our findings defined a new five-gene signature for predicting GC prognosis, but more validation is required to assess the effects and mechanism of the five genes, especially PLG, for the development of new GC therapies.
Collapse
Affiliation(s)
- Hui Shi
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, No.81, Mei Shan Road, Hefei, 230032, Anhui, China
| | - Jiangling Duan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, No.81, Mei Shan Road, Hefei, 230032, Anhui, China
| | - Zhangming Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengqi Huang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenxiu Han
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Rui Kong
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, No.81, Mei Shan Road, Hefei, 230032, Anhui, China
| | - Xiuyin Guan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, No.81, Mei Shan Road, Hefei, 230032, Anhui, China
| | - Zhen Qi
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, No.81, Mei Shan Road, Hefei, 230032, Anhui, China
| | - Shuang Zheng
- Department of Rheumatology, The First Affiliated Hospital of Anhui Medical University, No.218, Ji Xi Road, Hefei, 230032, Anhui, China.
| | - Ming Lu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, No.81, Mei Shan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
17
|
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] [Academic Contribution 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
|
18
|
Di Gregorio E, Israel S, Staelens M, Tankel G, Shankar K, Tuszyński JA. The distinguishing electrical properties of cancer cells. Phys Life Rev 2022; 43:139-188. [PMID: 36265200 DOI: 10.1016/j.plrev.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
In recent decades, medical research has been primarily focused on the inherited aspect of cancers, despite the reality that only 5-10% of tumours discovered are derived from genetic causes. Cancer is a broad term, and therefore it is inaccurate to address it as a purely genetic disease. Understanding cancer cells' behaviour is the first step in countering them. Behind the scenes, there is a complicated network of environmental factors, DNA errors, metabolic shifts, and electrostatic alterations that build over time and lead to the illness's development. This latter aspect has been analyzed in previous studies, but how the different electrical changes integrate and affect each other is rarely examined. Every cell in the human body possesses electrical properties that are essential for proper behaviour both within and outside of the cell itself. It is not yet clear whether these changes correlate with cell mutation in cancer cells, or only with their subsequent development. Either way, these aspects merit further investigation, especially with regards to their causes and consequences. Trying to block changes at various levels of occurrence or assisting in their prevention could be the key to stopping cells from becoming cancerous. Therefore, a comprehensive understanding of the current knowledge regarding the electrical landscape of cells is much needed. We review four essential electrical characteristics of cells, providing a deep understanding of the electrostatic changes in cancer cells compared to their normal counterparts. In particular, we provide an overview of intracellular and extracellular pH modifications, differences in ionic concentrations in the cytoplasm, transmembrane potential variations, and changes within mitochondria. New therapies targeting or exploiting the electrical properties of cells are developed and tested every year, such as pH-dependent carriers and tumour-treating fields. A brief section regarding the state-of-the-art of these therapies can be found at the end of this review. Finally, we highlight how these alterations integrate and potentially yield indications of cells' malignancy or metastatic index.
Collapse
Affiliation(s)
- Elisabetta Di Gregorio
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Simone Israel
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Autem Therapeutics, 35 South Main Street, Hanover, 03755, NH, USA
| | - Michael Staelens
- Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada
| | - Gabriella Tankel
- Department of Mathematics & Statistics, McMaster University, 1280 Main Street West, Hamilton, L8S 4K1, ON, Canada
| | - Karthik Shankar
- Department of Electrical & Computer Engineering, University of Alberta, 9211 116 Street NW, Edmonton, T6G 1H9, AB, Canada
| | - Jack A Tuszyński
- Dipartimento di Ingegneria Meccanica e Aerospaziale (DIMEAS), Politecnico di Torino, Corso Duca degli Abruzzi, 24, Torino, 10129, TO, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Drive NW, Edmonton, T6G 2E1, AB, Canada; Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton, T6G 1Z2, AB, Canada.
| |
Collapse
|
19
|
Zhang X, Luo Y, Cen Y, Qiu X, Li J, Jie M, Yang S, Qin S. MACC1 promotes pancreatic cancer metastasis by interacting with the EMT regulator SNAI1. Cell Death Dis 2022; 13:923. [PMID: 36333284 PMCID: PMC9636131 DOI: 10.1038/s41419-022-05285-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/23/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Metastasis is the dominant cause of cancer-related mortality. Metastasis-associated with colon cancer protein 1 (MACC1) has been proven to play a critical role in cancer metastasis. However, the prometastatic role of MACC1 in regulating the pancreatic cancer (PC) metastatic phenotype remains elusive. Here, we report that MACC1 is highly expressed in The Cancer Genome Atlas (TCGA) and tissue microarray (TMA) and identified as a good indicator for poor prognosis. Overexpression or knockdown of MACC1 in PC cells correspondingly promoted or inhibited pancreatic cancer cell migration and invasion in a MET proto-oncogene receptor tyrosine kinase (MET)-independent manner. Notably, knockdown of MACC1 in PC cells markedly decreased the liver metastatic lesions in a liver metastasis model. Mechanistically, MACC1 binds to the epithelial-mesenchymal transition (EMT) regulator snail family transcriptional repressor 1 (SNAI1) to drive EMT via upregulating the transcriptional activity of SNAI1, leading to the transactivation of fibronectin 1 (FN1) and the trans-repression of cadherin 1 (CDH1). Collectively, our results unveil a new mechanism by which MACC1 drives pancreatic cancer cell metastasis and suggest that the MACC1-SNAI1 complex-mediated mesenchymal transition may be a therapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Xianglian Zhang
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Ya Luo
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Yu Cen
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Xin Qiu
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| | - Jing Li
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Mengmeng Jie
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Shiming Yang
- grid.417298.10000 0004 1762 4928Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037 China
| | - Shanyu Qin
- grid.412594.f0000 0004 1757 2961Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021 China
| |
Collapse
|
20
|
Pellegrino M, Ricci E, Ceraldi R, Nigro A, Bonofiglio D, Lanzino M, Morelli C. From HDAC to Voltage-Gated Ion Channels: What's Next? The Long Road of Antiepileptic Drugs Repositioning in Cancer. Cancers (Basel) 2022; 14:cancers14184401. [PMID: 36139561 PMCID: PMC9497059 DOI: 10.3390/cancers14184401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Although in the last decades the clinical outcome of cancer patients considerably improved, the major drawbacks still associated with chemotherapy are the unwanted side effects and the development of drug resistance. Therefore, a continuous effort in trying to discover new tumor markers, possibly of diagnostic, prognostic and therapeutic value, is being made. This review is aimed at highlighting the anti-tumor activity that several antiepileptic drugs (AEDs) exert in breast, prostate and other types of cancers, mainly focusing on their ability to block the voltage-gated Na+ and Ca++ channels, as well as to inhibit the activity of histone deacetylases (HDACs), all well-documented tumor markers and/or molecular targets. The existence of additional AEDs molecular targets is highly suspected. Therefore, the repurposing of already available drugs as adjuvants in cancer treatment would have several advantages, such as reductions in dose-related toxicity CVs will be sent in a separate mail to the indicated address of combined treatments, lower production costs, and faster approval for clinical use. Abstract Cancer is a major health burden worldwide. Although the plethora of molecular targets identified in the last decades and the deriving developed treatments, which significantly improved patients’ outcome, the occurrence of resistance to therapies remains the major cause of relapse and mortality. Thus, efforts in identifying new markers to be exploited as molecular targets in cancer therapy are needed. This review will first give a glance on the diagnostic and therapeutic significance of histone deacetylase (HDAC) and voltage gated ion channels (VGICs) in cancer. Nevertheless, HDAC and VGICs have also been reported as molecular targets through which antiepileptic drugs (AEDs) seem to exert their anticancer activity. This should be claimed as a great advantage. Indeed, due to the slowness of drug approval procedures, the attempt to turn to off-label use of already approved medicines would be highly preferable. Therefore, an updated and accurate overview of both preclinical and clinical data of commonly prescribed AEDs (mainly valproic acid, lamotrigine, carbamazepine, phenytoin and gabapentin) in breast, prostate, brain and other cancers will follow. Finally, a glance at the emerging attempt to administer AEDs by means of opportunely designed drug delivery systems (DDSs), so to limit toxicity and improve bioavailability, is also given.
Collapse
Affiliation(s)
| | | | | | | | | | - Marilena Lanzino
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| | - Catia Morelli
- Correspondence: (M.L.); (C.M.); Tel.: +39-0984-496206 (M.L.); +39-0984-496211 (C.M.)
| |
Collapse
|
21
|
Song SJ, Liu X, Ji Q, Sun DZ, Xiu LJ, Xu JY, Yue XQ. Ziyin Huatan Recipe, a Chinese herbal compound, inhibits migration and invasion of gastric cancer by upregulating RUNX3 expression. JOURNAL OF INTEGRATIVE MEDICINE 2022; 20:355-364. [PMID: 35249836 DOI: 10.1016/j.joim.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/01/2021] [Accepted: 10/30/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVES Ziyin Huatan Recipe (ZYHT), a traditional Chinese medicine comprised of Lilii Bulbus, Pinelliae Rhizoma, and Hedyotis Diffusa, has shown promise in treating gastric cancer (GC). However, its potential mechanism has not yet been clearly addressed. This study aimed to predict targets and molecular mechanisms of ZYHT in treating GC by network pharmacology analysis and to explore the role of ZYHT in GC both in vitro and in vivo. METHODS Targets and molecular mechanisms of ZYHT were predicted via network pharmacology analysis. The effects of ZYHT on the expression of metastasis-associated targets were further validated by Western blot and quantitative real-time polymerase chain reaction. To explore the specific molecular mechanisms of the effects of ZYHT on migration and invasion, the runt-related transcription factor 3 (RUNX3) gene was knocked out by clustered regularly interspaced short palindromic repeats/Cas9, and lentiviral vectors were transfected into SGC-7901 cells. Then lung metastasis model of GC in nude mice was established to explore the anti-metastasis effect of ZYHT. Western blot and immunohistochemistry were used to explore the impact of ZYHT on the expression of metastasis-related proteins with or without RUNX3 gene. RESULTS The network pharmacology analysis showed that ZYHT might inhibit focal adhesion, migration, invasion and metastasis of GC. ZYHT inhibited the proliferation, migration and invasion of GC cells in vitro via regulating the expression of metastasis-associated targets. Knocking out RUNX3 almost completely reversed the cell phenotypes (migration and invasion) and protein expression levels elicited by ZYHT. In vivo studies showed that ZYHT inhibited the metastasis of GC cells to the lung and prolonged the survival time of the nude mice. Knocking out RUNX3 partly reversed the metastasis of GC cells to the lung and the protein expression levels elicited by ZYHT. CONCLUSION ZYHT can effectively inhibit the invasion and migration of GC in vitro and in vivo, and its molecular mechanism may relate to the upregulation of RUNX3 expression.
Collapse
Affiliation(s)
- Shang-Jin Song
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, China; Strategic Support Force Xingcheng Special Duty Sanatorium, Xingcheng 125100, Liaoning Province, China
| | - Xuan Liu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Qing Ji
- Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Da-Zhi Sun
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Li-Juan Xiu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jing-Yu Xu
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xiao-Qiang Yue
- Department of Traditional Chinese Medicine, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
22
|
Nav1.6 promotes the progression of human follicular thyroid carcinoma cells via JAK-STAT signaling pathway. Pathol Res Pract 2022; 236:153984. [PMID: 35753135 DOI: 10.1016/j.prp.2022.153984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/12/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022]
Abstract
Follicular thyroid carcinoma (FTC) is one of the most common malignant tumors of the endocrine system. Recent studies have shown that voltage-gated sodium channels (VGSCs) affect the proliferation, migration, and invasion of tumor cells. However, the expression and functions of VGSCs, and the molecular pathways activated by VGSCs in FTC cells remain unclear. Our studies revealed that the expression of Nav1.6, encoded by SCN8A, was the predominantly upregulated subtype of VGSCs in FTC tissues. Knockdown of Nav1.6 significantly inhibited the proliferation, epithelial-mesenchymal transition and invasiveness of FTC cells. Using gene set enrichment analysis and Kyoto Encyclopedia of Genes and Genomics, SCN8A was predicted to be related to the JAK-STAT signaling pathway. Hence, we targeted the JAK-STAT pathway and demonstrated that Nav1.6 enhanced FTC cell proliferation, epithelial-mesenchymal transition, and invasion by phosphorylating JAK2 to activate STAT3. Furthermore, downregulating the expression of Nav1.6 improve the susceptibility of FTC cells to ubenimex in vitro. These results suggest Nav1.6 accelerates FTC progression through JAK/STAT signaling and may be a potential target for FTC therapy.
Collapse
|
23
|
Wang Y, Han J, Zhou H, Ai S, Wan D. A Prognosis Marker Dynein Cytoplasmic 1 Heavy Chain 1 Correlates with EMT and Immune Signature in Liver Hepatocellular Carcinoma by Bioinformatics and Experimental Analysis. DISEASE MARKERS 2022; 2022:6304859. [PMID: 35601740 PMCID: PMC9117040 DOI: 10.1155/2022/6304859] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/08/2022] [Revised: 04/17/2022] [Accepted: 04/25/2022] [Indexed: 12/19/2022]
Abstract
Background Liver hepatocellular carcinoma (LIHC) has had a continuous increase in incidence and mortality rates over the last 40 years. Dynein Cytoplasmic 1 Heavy Chain 1 (DYNC1H1) is a protein coding gene which encodes the cytoplasmic dynein heavy chain family. This is the first investigation into the expression of DYNC1H1 and its mechanisms of action in LIHC patients. Methods Based on the DYNC1H1 expression data from the TCGA database, we performed the DYNC1H1 expression, clinicopathological data, gene enrichment, and immune infiltration analysis. TIMER and CIBERSORT were used to assess immune responses of DYNC1H1 in LIHC. GEPIA, K-M survival analysis, and immunohistochemical staining pictures from the THPA were used to validate the results. In order to evaluate the diagnostic value of DYNC1H1, GEO datasets were analyzed by using ROC analysis. And quantitative real-time polymerase chain reaction was also carried out to evaluate the expression of DYNC1H1. Results DYNC1H1 expression levels were associated with T classification, pathologic stage, histologic grade, and serum AFP levels. DYNC1H1 is an independent factor for a poor prognosis in patients with LIHC. Further study showed that high expression of DYNC1H1 was enriched in epithelial-mesenchymal transition (EMT) and the TGF β signaling pathway by GSEA analysis enrichment, indicating that DYNC1H1 might play a key role in the progression of CRC through EMT and immune response, which also had been validated by the experimental assays. Conclusions DYNC1H1 will provide a novel and important perspective for the mechanisms of LIHC by regulating EMT. This gene will be able to act as an efficacious tool for the early diagnosis and effective intervention of LIHC.
Collapse
Affiliation(s)
- Yanhong Wang
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Shanghai 200065, China
| | - Jiyu Han
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Shanghai 200065, China
| | - Haichao Zhou
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Shanghai 200065, China
| | - Songtao Ai
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Daqian Wan
- Department of Orthopedics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Shanghai 200065, China
| |
Collapse
|
24
|
Yan Y, He W, Chen Y, Li Q, Pan J, Yuan Y, Zeng W, Chen D, Xing W. Comprehensive Analysis to Identify the Encoded Gens of Sodium Channels as a Prognostic Biomarker in Hepatocellular Carcinoma. Front Genet 2022; 12:802067. [PMID: 35126466 PMCID: PMC8815461 DOI: 10.3389/fgene.2021.802067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/07/2021] [Accepted: 12/30/2021] [Indexed: 12/24/2022] Open
Abstract
The SCN family as the encoded gens of sodium channels has been proven to participate in development of cancers including hepatocellular carcinoma (HCC), but the prognostic value of the SCN family is unclear. The results of the UALCAN database had showed that SCN2A/4A/5A/8A mRNA were highly expressed in tumour tissues, while SCN1A/7A/11A mRNA were expressed at low levels (p < 0.05), furthermore, the expression of SCN4A and SCN7A had the similar levels in microarray analysis result. The pan-tumour analysis showed that SCN7A expression was stably lower in tumours than SCN4A expression by TIMER. Both SCN4A and SCN7A were related to tumour grade, nodal metastatic status, histological subtype, patient race, individual cancer stages and TP53 mutation status to varying degrees. The Kaplan–Meier plotter demonstrated that high SCN4A mRNA expression was correlated with better overall survival (OS), disease-specific survival (DSS) and progression-free survival (PFS) and that high expression of SCN7A mRNA was associated with better OS; however, in Asians, higher SCN4A was correlated with better OS and DSS, and higher SCN7A was well correlated with better OS, recurrence-free survival (RFS), DSS and PFS. Analysis of data from cBioPortal showed that mutation of SCN7A was related to RFS and PFS. The protein expression of SCN4A and SCN7A had been detected by Immunohistochemistry. Univariate survival analysis revealed that high SCN7A protein expression was significantly linked to better OS (p = 0.001) and RFS (p = 0.003). Moreover, SCN7A displayed as an independent prognostic factor by multivariate analysis. In addition, a lower methylation level indicated a poor outcome. Pathway and functional enrichment analysis predicted a relationship between SCN7A and the PI3K pathway. In conclusion, there are significant and stable changes in SCN4A and SCN7A expression in HCC. SCN7A expression has better prognostic value and might participate in HCC progression.
Collapse
Affiliation(s)
- Yan Yan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Anesthesiology, Huizhou Municipal Central Hospital, Huizhou, China
| | - Wen He
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yonghua Chen
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qiang Li
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jiahao Pan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yunfei Yuan
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Weian Zeng, ; Dongtai Chen, ; Wei Xing,
| | - Dongtai Chen
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Weian Zeng, ; Dongtai Chen, ; Wei Xing,
| | - Wei Xing
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Weian Zeng, ; Dongtai Chen, ; Wei Xing,
| |
Collapse
|
25
|
Cure MC, Cure E. Prolonged NHE Activation may be both Cause and Outcome of Cytokine Release Syndrome in COVID-19. Curr Pharm Des 2022; 28:1815-1822. [PMID: 35838211 DOI: 10.2174/1381612828666220713121741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/03/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023]
Abstract
The release of cytokines and chemokines such as IL-1β, IL-2, IL-6, IL-7, IL-10, TNF-α, IFN-γ, CCL2, CCL3, and CXCL10 is increased in critically ill patients with COVID-19. Excessive cytokine release during COVID-19 is related to increased morbidity and mortality. Several mechanisms are put forward for cytokine release syndrome during COVID-19. Here we have mentioned novel pathways. SARS-CoV-2 increases angiotensin II levels by rendering ACE2 nonfunctional. Angiotensin II causes cytokine release via AT1 and AT2 receptors. Moreover, angiotensin II potently stimulates the Na+/H+ exchanger (NHE). It is a pump found in the membranes of many cells that pumps Na+ inward and H+ outward. NHE has nine isoforms. NHE1 is the most common isoform found in endothelial cells and many cells. NHE is involved in keeping the intracellular pH within physiological limits. When the intracellular pH is acidic, NHE is activated, bringing the intracellular pH to physiological levels, ending its activity. Sustained NHE activity is highly pathological and causes many problems. Prolonged NHE activation in COVID-19 may cause a decrease in intracellular pH through H+ ion accumulation in the extracellular area and subsequent redox reactions. The activation reduces the intracellular K+ concentration and leads to Na+ and Ca2+ overload. Increased ROS can cause intense cytokine release by stimulating NF-κB and NLRP3 inflammasomes. Cytokines also cause overstimulation of NHE. As the intracellular pH decreases, SARS-CoV-2 rapidly infects new cells, increasing the viral load. This vicious circle increases morbidity and mortality in patients with COVID-19. On the other hand, SARS-CoV-2 interaction with NHE3 in intestinal tissue is different from other tissues. SARS-CoV-2 can trigger CRS via NHE3 inhibition by disrupting the intestinal microbiota. This review aimed to help develop new treatment models against SARS-CoV-2- induced CRS by revealing the possible effects of SARS-CoV-2 on the NHE.
Collapse
Affiliation(s)
| | - Erkan Cure
- Department of Internal Medicine, Bagcilar Medilife Hospital, Istanbul, Turkey
| |
Collapse
|
26
|
Na +/H +-Exchanger Family as Novel Prognostic Biomarkers in Colorectal Cancer. JOURNAL OF ONCOLOGY 2021; 2021:3241351. [PMID: 34759967 PMCID: PMC8575632 DOI: 10.1155/2021/3241351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 08/28/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022]
Abstract
Background The acidic characteristics of the tumor microenvironment (TME) are attributed to cancer cells' needs of metabolism which produce a large amount of H+. In order not to affect its own life activities, it needs to release H+ into the intercellular space through an efficient Na+/H+ exchanger. On account of the intestine whose physiological function is highly dependent on intestinal pH value, NHE family members may play a critical role in the occurrence and development of colorectal cancer (CRC). Methods TCGA, GEPIA2, ONCOMINE, UALCAN, STRING, TIMER, Cytoscape, TargetScan, ENCORI, LncBase v.2, DNMIVD, HPA, and CellMinerTM databases were used in our study. Results The mRNA expressions of SLC9A1, SLC9A2, SLC9A3, and SLC9A9 were evidently lower in COAD than in normal samples; however, the mRNA expressions of SLC9A5, SLC9A8, and SLC9B2 were higher. Besides, mRNA expressions of NHE family were extremely associated with clinicopathological features, tumor immune microenvironment and stemness score, DNA methylation, and patient prognosis in COAD. Moreover, we conjectured that NHE family may play a role through MAPK or ErbB signaling pathway according to the results of GO/KEGG enrichment analysis. At last, we found that NHE family members were key factors of various kinds of cancers. Conclusion Our study indicated that NHE family represented new diagnostic and therapeutic targets for CRC, which could have important significance for the clinical treatment of CRC.
Collapse
|
27
|
Murtadha AH, Azahar IIM, Sharudin NA, Has ATC, Mokhtar NF. Influence of nNav1.5 on MHC class I expression in breast cancer. J Biosci 2021. [DOI: 10.1007/s12038-021-00196-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
|
28
|
Lopez-Charcas O, Pukkanasut P, Velu SE, Brackenbury WJ, Hales TG, Besson P, Gomora JC, Roger S. Pharmacological and nutritional targeting of voltage-gated sodium channels in the treatment of cancers. iScience 2021; 24:102270. [PMID: 33817575 PMCID: PMC8010468 DOI: 10.1016/j.isci.2021.102270] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023] Open
Abstract
Voltage-gated sodium (NaV) channels, initially characterized in excitable cells, have been shown to be aberrantly expressed in non-excitable cancer tissues and cells from epithelial origins such as in breast, lung, prostate, colon, and cervix, whereas they are not expressed in cognate non-cancer tissues. Their activity was demonstrated to promote aggressive and invasive potencies of cancer cells, both in vitro and in vivo, whereas their deregulated expression in cancer tissues has been associated with metastatic progression and cancer-related death. This review proposes NaV channels as pharmacological targets for anticancer treatments providing opportunities for repurposing existing NaV-inhibitors or developing new pharmacological and nutritional interventions.
Collapse
Affiliation(s)
- Osbaldo Lopez-Charcas
- Université de Tours, EA4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Piyasuda Pukkanasut
- Department of Chemistry, The University of Alabama at Birmingham, CHEM 280. 901, 14th Street S, Birmingham, AL 35294, USA
| | - Sadanandan E. Velu
- Department of Chemistry, The University of Alabama at Birmingham, CHEM 280. 901, 14th Street S, Birmingham, AL 35294, USA
| | - William J. Brackenbury
- Department of Biology, York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Tim G. Hales
- Institute of Academic Anaesthesia, Division of Systems Medicine, School of Medicine, the University of Dundee, DD1 9SY, Dundee, UK
| | - Pierre Besson
- Université de Tours, EA4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
| | - Juan Carlos Gomora
- Instituto de Fisiología Celular, Circuito Exterior s/n Ciudad Universitaria, Universidad Nacional Autónoma de México, Mexico City, 04510 México
| | - Sébastien Roger
- Université de Tours, EA4245 Transplantation, Immunologie, Inflammation, Faculté de Médecine de Tours, 10 Boulevard Tonnellé, 37032 Tours, France
- Institut Universitaire de France, 75005 Paris, France
| |
Collapse
|
29
|
Hou K, Liu J, Du J, Mi S, Ma S, Ba Y, Ji H, Li B, Hu S. Dihydroartemisinin prompts amplification of photodynamic therapy-induced reactive oxygen species to exhaust Na/H exchanger 1-mediated glioma cells invasion and migration. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 219:112192. [PMID: 34000476 DOI: 10.1016/j.jphotobiol.2021.112192] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/16/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022]
Abstract
Photodynamic therapy (PDT) is a promising glioma therapy; however, its efficacy is compromised due to the PDT-induced reactive oxygen species (ROS) production being limited by the local hypoxic tumor microenvironment. Furthermore, Hypoxia activates sodium/hydrogen exchanger 1 (NHE1), an essential component for tumor progression and metastasis, enables glioma cells (GC) to escape PDT-mediated phototoxicity via increased H+ extrusion. However, interactions between NHE1 expression with ROS level involving response of GC remain unclear. Dihydroartemisinin (DHA), a ROS generator, has extensive anti-tumor effects. This study aimed to explore whether PDT along with DHA could amplify the total ROS levels and diminish GC invasion and migration by inhibiting NHE1 expression. Proliferation and invasion of U251 and LN229 cells were evaluated under different treatments using cell counting Kit-8 (CCK-8), transwell, and wound healing assays. ROS levels were measured using fluorescence probes and flow cytometry. NHE1 levels were detected by immunofluorescence and western blotting. Co-treatment effects and molecular events were further confirmed in a bilateral tumor-bearing nude mouse model. PDT with synergistic DHA significantly increased the total abundance of ROS to further suppress the invasion and migration of GC by reducing NHE1 levels in vitro. Using a bilateral glioma xenograft mouse model with primary and recurrent gliomas, we found that PDT markedly suppressed primary tumor growth, while PDT in synergy with DHA also suppressed recurrent tumors, and improved overall survival by regulating the ROS-NHE1 axis. No evident side effects were observed. Our results suggest that PDT with DHA can amplify the total ROS levels to weaken GC invasion and migration by suppressing NHE1 expression in vitro and in vivo, thus abolishing the resistance of GC to PDT. The synergistic therapy of PDT and DHA therefore represents a more efficient and safe strategy for comprehensive glioma treatment.
Collapse
Affiliation(s)
- Kuiyuan Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jie Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Jianyang Du
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Shan Mi
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Shuai Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yixu Ba
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Hang Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Bo Li
- Department of Neurosurgery, The First People's Hospital of Taizhou, Taizhou 318020, China
| | - Shaoshan Hu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
30
|
Systematic Identification of MACC1-Driven Metabolic Networks in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13050978. [PMID: 33652667 PMCID: PMC7956336 DOI: 10.3390/cancers13050978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/11/2020] [Revised: 02/16/2021] [Accepted: 02/21/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary We aimed at the systematic identification of MACC1-driven metabolic networks in colorectal cancer. By this systematic analysis, our studies revealed new insights into MACC1-caused metabolomics phenotypes: (i) MACC1 fosters metastasis by rewiring glucose and glutamine metabolism, (ii) MACC1 increases glucose use by enhanced surface GLUT1; (iii) MACC1 increases glutamine and pyruvate use by enhanced uptake, and (iv) MACC1 reduces glutamine flux but has minor effects on pyruvate flux. Therefore, MACC1 is an important regulator of cancer metabolism. Abstract MACC1 is a prognostic and predictive metastasis biomarker for more than 20 solid cancer entities. However, its role in cancer metabolism is not sufficiently explored. Here, we report on how MACC1 impacts the use of glucose, glutamine, lactate, pyruvate and fatty acids and show the comprehensive analysis of MACC1-driven metabolic networks. We analyzed concentration-dependent changes in nutrient use, nutrient depletion, metabolic tracing employing 13C-labeled substrates, and in vivo studies. We found that MACC1 permits numerous effects on cancer metabolism. Most of those effects increased nutrient uptake. Furthermore, MACC1 alters metabolic pathways by affecting metabolite production or turnover from metabolic substrates. MACC1 supports use of glucose, glutamine and pyruvate via their increased depletion or altered distribution within metabolic pathways. In summary, we demonstrate that MACC1 is an important regulator of metabolism in cancer cells.
Collapse
|
31
|
Sodium ion channels as potential therapeutic targets for cancer metastasis. Drug Discov Today 2021; 26:1136-1147. [PMID: 33545383 DOI: 10.1016/j.drudis.2021.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2020] [Revised: 07/31/2020] [Accepted: 01/27/2021] [Indexed: 12/28/2022]
Abstract
Is it possible to develop drugs for the treatment of a specific type of metastatic cancer by targeting sodium ion channels?
Collapse
|
32
|
Voltage-gated sodium channel Na v1.5 promotes tumor progression and enhances chemosensitivity to 5-fluorouracil in colorectal cancer. Cancer Lett 2020; 500:119-131. [PMID: 33338532 DOI: 10.1016/j.canlet.2020.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/14/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022]
Abstract
Nav1.5, encoded by SCN5A, has been associated with metastasis in colorectal cancer (CRC). Here, we investigated the mechanism by which Nav1.5 regulates tumor progression and whether Nav1.5 influences chemosensitivity to 5-fluorouracil (5-FU) in CRCs. CRC cases were evaluated for Nav1.5 expression. Elevated Nav1.5 expression was associated with poor prognosis in CRCs, whereas stage II/III patients with upregulated SCN5A expression could have better survival after receiving 5-FU-based adjuvant chemotherapy. In CRC cells, SCN5A knockdown reduced the proliferation, migration and invasion. According to RNA sequencing, SCN5A knockdown inhibited both the cell cycle and epithelial-mesenchymal transition. In addition, Nav1.5 stabilized the KRas-calmodulin complex to modulate Ras signaling, promoting Ca2+ influx through the Na+-Ca2+ exchanger and Ca2+ release-activated calcium channel. Meanwhile, SCN5A knockdown increased the 50% inhibitory concentration to 5-FU by upregulating 5-FU-stimulated apoptosis in CRCs. In conclusion, Nav1.5 could progress to proliferation and metastasis through Ca2+/calmodulin-dependent Ras signaling in CRC, and it could also enhance 5-FU-stimulated apoptosis. Clinically, patients with stage II/III CRCs with elevated SCN5A expression demonstrated poor prognosis, yet those patients could benefit more from 5-FU-based chemotherapy than patients with lower SCN5A expression.
Collapse
|
33
|
Koltai T. Targeting the pH Paradigm at the Bedside: A Practical Approach. Int J Mol Sci 2020; 21:E9221. [PMID: 33287221 PMCID: PMC7730959 DOI: 10.3390/ijms21239221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/12/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023] Open
Abstract
The inversion of the pH gradient in malignant tumors, known as the pH paradigm, is increasingly becoming accepted by the scientific community as a hallmark of cancer. Accumulated evidence shows that this is not simply a metabolic consequence of a dysregulated behavior, but rather an essential process in the physiopathology of accelerated proliferation and invasion. From the over-simplification of increased lactate production as the cause of the paradigm, as initially proposed, basic science researchers have arrived at highly complex and far-reaching knowledge, that substantially modified that initial belief. These new developments show that the paradigm entails a different regulation of membrane transporters, electrolyte exchangers, cellular and membrane enzymes, water trafficking, specialized membrane structures, transcription factors, and metabolic changes that go far beyond fermentative glycolysis. This complex world of dysregulations is still shuttered behind the walls of experimental laboratories and has not yet reached bedside medicine. However, there are many known pharmaceuticals and nutraceuticals that are capable of targeting the pH paradigm. Most of these products are well known, have low toxicity, and are also inexpensive. They need to be repurposed, and this would entail shorter clinical studies and enormous cost savings if we compare them with the time and expense required for the development of a new molecule. Will targeting the pH paradigm solve the "cancer problem"? Absolutely not. However, reversing the pH inversion would strongly enhance standard treatments, rendering them more efficient, and in some cases permitting lower doses of toxic drugs. This article's goal is to describe how to reverse the pH gradient inversion with existing drugs and nutraceuticals that can easily be used in bedside medicine, without adding toxicity to established treatments. It also aims at increasing awareness among practicing physicians that targeting the pH paradigm would be able to improve the results of standard therapies. Some clinical cases will be presented as well, showing how the pH gradient inversion can be treated at the bedside in a simple manner with repurposed drugs.
Collapse
Affiliation(s)
- Tomas Koltai
- Centro de Diagnostico y Tratamiento de la Obra Social del Personal de la Alimentacion, Talar de Pacheco, Buenos Aires 1617, Argentina
| |
Collapse
|
34
|
Zhang L, Wang Z, Xiao J, Chen H, Zhang Z, Li H, Wang Y, Piao H, Li F, Zhang L, Zhang J. Sodium to globulin ratio as a prognostic factor for patients with advanced gastric cancer. J Cancer 2020; 11:7320-7328. [PMID: 33193896 PMCID: PMC7646180 DOI: 10.7150/jca.47314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/22/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Electrolyte disturbance and systemic inflammation contributes to poor prognosis of cancer patients. Levels of serum sodium and globulin can reflect electrolyte homeostasis and inflammatory state, respectively, therefore have potential as prognostic factors for cancer patients. In this study, we hypothesized that sodium to globulin ratio (SGR) could have superior accuracy in predicting cancer patient survival, than sodium and globulin alone. We therefore sought to investigate its efficacy in prognosis of patients with advanced gastric cancer (GC) receiving first-line chemotherapy. Methods: A total of 265 patients, with advanced GC, were recruited in this retrospective study from January 2014 to January 2019. We first determined SGR cut-off values using the receiver operating characteristic (ROC) analysis, then analyzed the relationship between pretreatment SGR and clinicopathological features and the effect of chemotherapy. Finally, we evaluated progression-free survival (PFS) and overall survival (OS) rates of the entire and subgroup populations using univariate and multivariate logistic regressions. Results: SGR recorded a cut-off value of 5.54, and had a significantly higher area under the curve (AUC) value (0.619, p = 0.001) than fibrinogen (0.575, p = 0.034) and albumin (0.610, p = 0.002) alone. Organ metastasis, and peritoneal invasion ratios, as well as neutrophil and CA72-4 levels varied significantly between the low-SGR (SGR≤ 5.54) and high SGR (SGR> 5.54) groups (all p < 0.05). Specifically, patients in the low-SGR group exhibited significantly lower disease control rates (83.4%) than those in the high-SGR group (97.2%) (p < 0.001). Results from multivariate analysis indicated that high-SGR was an independent risk factor for PFS (Hazard ratio [HR]: 0.539, p < 0.001) and OS (HR: 0.574, p < 0.001). Moreover, patients in the low-SGR group exhibited significantly worse PFS (134 vs. 221 days, p < 0.001) and OS (311 vs. 420 days, p < 0.001) than those in the high-SGR group. Furthermore, subgroup analysis revealed that SGR was still a powerful prognostic indicator in GC patients with good prognosis or normal biochemical indexes, including no peritoneal infiltration, normal neutrophil counts, and normal serum sodium and globulin levels (all p < 0.001). Conclusions: Overall, our findings indicate that SGR is a novel and promising prognostic factor for GC patients. It has superior accuracy, to sodium and globulin alone, hence it is a powerful tool for evaluating effects of treatment, PFS, and OS in patients with advanced GC, who receive first-line chemotherapy.
Collapse
Affiliation(s)
- Liqun Zhang
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China.,Department of Medical Oncology, Shenyang Fifth People Hospital, Tiexi District, Shenyang 110020, Liaoning Province, China
| | - Zhuo Wang
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China.,Department of Medical Oncology, Liaohua Hospital, Hongwei District, Liaoyang 111003, Liaoning Province, China
| | - Jiawen Xiao
- Department of Medical Oncology, Shenyang Fifth People Hospital, Tiexi District, Shenyang 110020, Liaoning Province, China
| | - Hao Chen
- Department of Medical Oncology, Liaohua Hospital, Hongwei District, Liaoyang 111003, Liaoning Province, China
| | - Zhiyan Zhang
- Department of Medical Oncology, Shenyang Fifth People Hospital, Tiexi District, Shenyang 110020, Liaoning Province, China
| | - Haijing Li
- Department of Medical Oncology, Shenyang Fifth People Hospital, Tiexi District, Shenyang 110020, Liaoning Province, China
| | - Yuanhe Wang
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| | - Haiyan Piao
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| | - Fang Li
- Department of Hepatobiliary Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| | - Lisha Zhang
- Department of Gastrointestinal Surgery, The Second Hospital Affiliated to Harbin Medical University, No. 246, Xuefu Road, Nangang District, Harbin 150086, Heilongjiang Province, China
| | - Jingdong Zhang
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang 110042, Liaoning Province, China
| |
Collapse
|
35
|
Wang P, Wadsworth PA, Dvorak NM, Singh AK, Chen H, Liu Z, Zhou R, Holthauzen LMF, Zhou J, Laezza F. Design, Synthesis, and Pharmacological Evaluation of Analogues Derived from the PLEV Tetrapeptide as Protein-Protein Interaction Modulators of Voltage-Gated Sodium Channel 1.6. J Med Chem 2020; 63:11522-11547. [PMID: 33054193 DOI: 10.1021/acs.jmedchem.0c00531] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022]
Abstract
The voltage-gated Na+ (Nav) channel is the molecular determinant of excitability. Disruption of protein-protein interactions (PPIs) between Nav1.6 and fibroblast growth factor 14 (FGF14) leads to impaired excitability of neurons in clinically relevant brain areas associated with channelopathies. Here, we designed, synthesized, and pharmacologically characterized new peptidomimetics based on a PLEV tetrapeptide scaffold derived from the FGF14:Nav1.6 PPI interface. Addition of an N-terminal 1-adamantanecarbonyl pharmacophore significantly improved peptidomimetic inhibitory potency. Surface plasmon resonance studies revealed that while this moiety was sufficient to confer binding to FGF14, altering the C-terminal moiety from methoxy (21a) to π bond-containing (23a and 23b) or cycloalkane substituents (23e) abrogated the binding to Nav1.6. Whole-cell patch-clamp electrophysiology subsequently revealed that 21a had functionally relevant interactions with both the C-terminal tail of Nav1.6 and FGF14. Collectively, these findings support that 21a (PW0564) may serve as a promising lead to develop target-selective neurotherapeutics by modulating protein-channel interactions.
Collapse
|
36
|
Cardoso FC. Multi-targeting sodium and calcium channels using venom peptides for the treatment of complex ion channels-related diseases. Biochem Pharmacol 2020; 181:114107. [PMID: 32579958 DOI: 10.1016/j.bcp.2020.114107] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/08/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Venom peptides are amongst the most exquisite group of bioactive molecules able to alter the normal physiology of organisms. These bioactive peptides penetrate tissues and blood vessels to encounter a number of receptors and ion channels to which they bind with high affinity and execute modulatory activities. Arachnid is the most diverse class of venomous animals often rich in peptides modulating voltage-gated sodium (NaV), calcium (CaV), and potassium (KV) channels. Spider venoms, in particular, contain potent and selective peptides targeting these channels, with a few displaying interesting multi-target properties for NaV and CaV channels underlying disease mechanisms such as in neuropathic pain, motor neuron disease and cancer. The elucidation of the pharmacology and structure-function properties of these venom peptides are invaluable for the development of effective drugs targeting NaV and CaV channels. This perspective discusses spider venom peptides displaying multi-target properties to modulate NaV and CaV channels in regard to their pharmacological features, structure-function relationships and potential to become the next generation of effective drugs to treat neurological disorders and other multi-ion channels related diseases.
Collapse
Affiliation(s)
- Fernanda C Cardoso
- Institute for Molecular Bioscience, The University of Queensland, 306 Carmody Rd., St Lucia, QLD AU 4072, Australia
| |
Collapse
|
37
|
Almasi S, El Hiani Y. Exploring the Therapeutic Potential of Membrane Transport Proteins: Focus on Cancer and Chemoresistance. Cancers (Basel) 2020; 12:cancers12061624. [PMID: 32575381 PMCID: PMC7353007 DOI: 10.3390/cancers12061624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Improving the therapeutic efficacy of conventional anticancer drugs represents the best hope for cancer treatment. However, the shortage of druggable targets and the increasing development of anticancer drug resistance remain significant problems. Recently, membrane transport proteins have emerged as novel therapeutic targets for cancer treatment. These proteins are essential for a plethora of cell functions ranging from cell homeostasis to clinical drug toxicity. Furthermore, their association with carcinogenesis and chemoresistance has opened new vistas for pharmacology-based cancer research. This review provides a comprehensive update of our current knowledge on the functional expression profile of membrane transport proteins in cancer and chemoresistant tumours that may form the basis for new cancer treatment strategies.
Collapse
Affiliation(s)
- Shekoufeh Almasi
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON KIH 8M5, Canada;
| | - Yassine El Hiani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
- Correspondence:
| |
Collapse
|
38
|
Díaz-García A, Varela D. Voltage-Gated K +/Na + Channels and Scorpion Venom Toxins in Cancer. Front Pharmacol 2020; 11:913. [PMID: 32655396 PMCID: PMC7325878 DOI: 10.3389/fphar.2020.00913] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/02/2020] [Accepted: 06/04/2020] [Indexed: 12/25/2022] Open
Abstract
Ion channels have recently been recognized as novel therapeutic targets in cancer research since they are overexpressed in different histological tissues, and their activity is linked to proliferation, tumor progression, angiogenesis, metastasis, and apoptosis. Voltage gated-potassium channels (VGKC) are involved in cell proliferation, cancer progression, cell cycle transition, and apoptosis. Moreover, voltage-dependent sodium channels (VGSC) contribute to decreases in extracellular pH, which, in turn, promotes cancer cell migration and invasion. Furthermore, VGSC and VGKC modulate voltage-sensitive Ca2+ channel activity by controlling the membrane potential and regulating Ca2+ influx, which functions as a second messenger in processes related to proliferation, invasion, migration, and metastasis. The subgroup of these types of channels that have shown a high oncogenic potential have become known as "oncochannels", and the evidence has highlighted them as key potential therapeutic targets. Scorpion venoms contain a high proportion of peptide toxins that act by modulating voltage-gated Na+/K+ channel activity. Increasing scientific data have pointed out that scorpion venoms and their toxins can affect the activity of oncochannels, thus showing their potential for anticancer therapy. In this review, we provide an update of the most relevant voltage-gated Na+\K+ ion channels as cellular targets and discuss the possibility of using scorpion venom and toxins for anticancer therapy.
Collapse
Affiliation(s)
- Alexis Díaz-García
- LifEscozul Chile SpA, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
| | - Diego Varela
- Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Universidad de Chile, Santiago, Chile
- Program of Physiology and Biophysics, Faculty of Medicine, Institute of Biomedical Sciences (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
39
|
ZiYinHuaTan Recipe Inhibits Cell Proliferation and Promotes Apoptosis in Gastric Cancer by Suppressing PI3K/AKT Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2018162. [PMID: 32382534 PMCID: PMC7193275 DOI: 10.1155/2020/2018162] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/26/2019] [Revised: 07/15/2019] [Accepted: 07/25/2019] [Indexed: 01/26/2023]
Abstract
In recent years, traditional Chinese medicine has played an important role in the treatment of gastric cancer in China. ZiYinHuaTan (ZYHT) recipe was developed for advanced gastric cancer and had shown its promising value in the clinic. In this study, we explore the effect of ZYHT on gastric cancer in vitro and in vivo. ZYHT can inhibit tumor growth and improve the general condition of mice in subcutaneous transplantation nude mice models of gastric cancer. And ZYHT can also inhibit cell proliferation and blocked the cells in G0/G1 to induce cell apoptosis in HGC27 and MGC803 cells. Then, network pharmacology analysis showed that ZYHT may exert antitumor effect mainly through PI3K/AKT signaling pathway. Furthermore, the expression of PI3K, p-Akt, CyclinD1, and Bcl-2 was detected in vitro and in vivo. The results showed that ZYHT could decrease the expression of PI3K, CyclinD1, and Bcl-2 both in vitro and in vivo. These results suggested that ZYHT could be used as a method for the treatment of developed gastric cancer.
Collapse
|
40
|
The Interplay of Dysregulated pH and Electrolyte Imbalance in Cancer. Cancers (Basel) 2020; 12:cancers12040898. [PMID: 32272658 PMCID: PMC7226178 DOI: 10.3390/cancers12040898] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/02/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer cells and tissues have an aberrant regulation of hydrogen ion dynamics driven by a combination of poor vascular perfusion, regional hypoxia, and increased the flux of carbons through fermentative glycolysis. This leads to extracellular acidosis and intracellular alkalinization. Dysregulated pH dynamics influence cancer cell biology, from cell transformation and tumorigenesis to proliferation, local growth, invasion, and metastasis. Moreover, this dysregulated intracellular pH (pHi) drives a metabolic shift to increased aerobic glycolysis and reduced mitochondrial oxidative phosphorylation, referred to as the Warburg effect, or Warburg metabolism, which is a selective feature of cancer. This metabolic reprogramming confers a thermodynamic advantage on cancer cells and tissues by protecting them against oxidative stress, enhancing their resistance to hypoxia, and allowing a rapid conversion of nutrients into biomass to enable cell proliferation. Indeed, most cancers have increased glucose uptake and lactic acid production. Furthermore, cancer cells have very dysregulated electrolyte balances, and in the interaction of the pH dynamics with electrolyte, dynamics is less well known. In this review, we highlight the interconnected roles of dysregulated pH dynamics and electrolytes imbalance in cancer initiation, progression, adaptation, and in determining the programming and reprogramming of tumor cell metabolism.
Collapse
|
41
|
Poku LO, Phil M, Cheng Y, Wang K, Sun X. 23 Na-MRI as a Noninvasive Biomarker for Cancer Diagnosis and Prognosis. J Magn Reson Imaging 2020; 53:995-1014. [PMID: 32219933 PMCID: PMC7984266 DOI: 10.1002/jmri.27147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/13/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022] Open
Abstract
The influx of sodium (Na+) ions into a resting cell is regulated by Na+ channels and by Na+/H+ and Na+/Ca2+ exchangers, whereas Na+ ion efflux is mediated by the activity of Na+/K+‐ATPase to maintain a high transmembrane Na+ ion gradient. Dysfunction of this system leads to changes in the intracellular sodium concentration that promotes cancer metastasis by mediating invasion and migration. In addition, the accumulation of extracellular Na+ ions in cancer due to inflammation contributes to tumor immunogenicity. Thus, alterations in the Na+ ion concentration may potentially be used as a biomarker for malignant tumor diagnosis and prognosis. However, current limitations in detection technology and a complex tumor microenvironment present significant challenges for the in vivo assessment of Na+ concentration in tumor. 23Na‐magnetic resonance imaging (23Na‐MRI) offers a unique opportunity to study the effects of Na+ ion concentration changes in cancer. Although challenged by a low signal‐to‐noise ratio, the development of ultrahigh magnetic field scanners and specialized sodium acquisition sequences has significantly advanced 23Na‐MRI. 23Na‐MRI provides biochemical information that reflects cell viability, structural integrity, and energy metabolism, and has been shown to reveal rapid treatment response at the molecular level before morphological changes occur. Here we review the basis of 23Na‐MRI technology and discuss its potential as a direct noninvasive in vivo diagnostic and prognostic biomarker for cancer therapy, particularly in cancer immunotherapy. We propose that 23Na‐MRI is a promising method with a wide range of applications in the tumor immuno‐microenvironment research field and in cancer immunotherapy monitoring. Level of Evidence 2 Technical Efficacy Stage 2
Collapse
Affiliation(s)
| | - M Phil
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.,Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Yongna Cheng
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.,Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Kai Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.,Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, China.,Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin, China.,TOF-PET/CT/MR Center, The Fourth Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
42
|
Cao L, Yuan Z, Liu M, Stock C. (Patho-)Physiology of Na +/H + Exchangers (NHEs) in the Digestive System. Front Physiol 2020; 10:1566. [PMID: 32009977 PMCID: PMC6974801 DOI: 10.3389/fphys.2019.01566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Na+/H+ exchangers (NHEs) are expressed in virtually all human tissues and organs. Two major tasks of those NHE isoforms that are located in plasma membranes are cell volume control by Na+-uptake and cellular pH regulation by H+-extrusion. Several NHEs, particularly NHE 1–4 and 8, are involved in the pathogenesis of diseases of the digestive system such as inflammatory bowel disease (ulcerative colitis, Crohn’s disease) and gastric and colorectal tumorigenesis. In the present review, we describe the physiological purposes, possible malfunctions and pathophysiological effects of the different NHE isoforms along the alimentary canal from esophagus to colon, including pancreas, liver and gallbladder. Particular attention is paid to the functions of NHEs in injury repair and to the role of NHE1 in Barrett’s esophagus. The impact of NHEs on gut microbiota and intestinal mucosal integrity is also dealt with. As the hitherto existing findings are not always consistent, sometimes even controversial, they are compared and critically discussed.
Collapse
Affiliation(s)
- Li Cao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenglin Yuan
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei Liu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Christian Stock
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
43
|
Malacrida A, Rivara M, Di Domizio A, Cislaghi G, Miloso M, Zuliani V, Nicolini G. 3D proteome-wide scale screening and activity evaluation of a new ALKBH5 inhibitor in U87 glioblastoma cell line. Bioorg Med Chem 2019; 28:115300. [PMID: 31937477 DOI: 10.1016/j.bmc.2019.115300] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/24/2019] [Revised: 12/18/2019] [Accepted: 12/26/2019] [Indexed: 12/14/2022]
Abstract
The imidazobenzoxazin-5-thione MV1035, synthesized as a new sodium channel blocker, has been tested on tumoral cells that differ for origin and for expressed NaV pool (U87-MG, H460 and A549). In this paper we focus on the effect of MV1035 in reducing U87 glioblastoma cell line migration and invasiveness. Since the effect of this compound on U87-MG cells seemed not dependent on its sodium channel blocking capability, alternative off-target interaction for MV1035 have been identified using SPILLO-PBSS software. This software performs a structure-based in silico screening on a proteome-wide scale, that allows to identify off-target interactions. Among the top-ranked off-targets of MV1035, we focused on the RNA demethylase ALKBH5 enzyme, known for playing a key role in cancer. In order to prove the effect of MV1035 on ALKBH5 in vitro coincubation of MV1035 and ALKBH5 has been performed demonstrating a consequent increase of N6-methyladenosine (m6A) RNA. To further validate the pathway involving ALKBH5 inhibition by MV1035 in U87-MG reduced migration and invasiveness, we evaluated CD73 as possible downstream protein. CD73 is an extrinsic protein involved in the generation of adenosine and is overexpressed in several tumors including glioblastoma. We have demonstrated that treating U87-MG with MV1035, CD73 protein expression was reduced without altering CD73 transcription. Our results show that MV1035 is able to significantly reduce U87 cell line migration and invasiveness inhibiting ALKBH5, an RNA demethylase that can be considered an interesting target in fighting glioblastoma aggressiveness. Our data encourage to further investigate the MV1035 inhibitory effect on glioblastoma.
Collapse
Affiliation(s)
- Alessio Malacrida
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy
| | - Mirko Rivara
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, PR, Italy.
| | - Alessandro Di Domizio
- Department of Pharmacological and Biomolecular Sciences, University of Milano, via Balzaretti 9, 20133 Milano, Italy; SPILLOproject, via Stradivari 17, 20037 Paderno Dugnano, Milano, Italy(2)
| | - Giacomo Cislaghi
- SPILLOproject, via Stradivari 17, 20037 Paderno Dugnano, Milano, Italy(2)
| | - Mariarosaria Miloso
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy
| | - Valentina Zuliani
- Food and Drug Department, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, PR, Italy
| | - Gabriella Nicolini
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, via Cadore 48, 20900 Monza, MB, Italy
| |
Collapse
|
44
|
Gradek F, Lopez-Charcas O, Chadet S, Poisson L, Ouldamer L, Goupille C, Jourdan ML, Chevalier S, Moussata D, Besson P, Roger S. Sodium Channel Na v1.5 Controls Epithelial-to-Mesenchymal Transition and Invasiveness in Breast Cancer Cells Through its Regulation by the Salt-Inducible Kinase-1. Sci Rep 2019; 9:18652. [PMID: 31819138 PMCID: PMC6901527 DOI: 10.1038/s41598-019-55197-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/23/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Loss of epithelial polarity and gain in invasiveness by carcinoma cells are critical events in the aggressive progression of cancers and depend on phenotypic transition programs such as the epithelial-to-mesenchymal transition (EMT). Many studies have reported the aberrant expression of voltage-gated sodium channels (NaV) in carcinomas and specifically the NaV1.5 isoform, encoded by the SCN5A gene, in breast cancer. NaV1.5 activity, through an entry of sodium ions, in breast cancer cells is associated with increased invasiveness, but its participation to the EMT has to be clarified. In this study, we show that reducing the expression of NaV1.5 in highly aggressive human MDA-MB-231 breast cancer cells reverted the mesenchymal phenotype, reduced cancer cell invasiveness and the expression of the EMT-promoting transcription factor SNAI1. The heterologous expression of NaV1.5 in weakly invasive MCF-7 breast cancer cells induced their expression of both SNAI1 and ZEB1 and increased their invasive capacities. In MCF-7 cells the stimulation with the EMT-activator signal TGF-β1 increased the expression of SCN5A. Moreover, the reduction of the salt-inducible kinase 1 (SIK1) expression promoted NaV1.5-dependent invasiveness and expression of EMT-associated transcription factor SNAI1. Altogether, these results indicated a prominent role of SIK1 in regulating NaV1.5-dependent EMT and invasiveness.
Collapse
Affiliation(s)
- Frédéric Gradek
- EA4245 Transplantation, Immunologie, Inflammation; Université de Tours, Tours, France
| | - Osbaldo Lopez-Charcas
- EA4245 Transplantation, Immunologie, Inflammation; Université de Tours, Tours, France
| | - Stéphanie Chadet
- EA4245 Transplantation, Immunologie, Inflammation; Université de Tours, Tours, France
| | - Lucile Poisson
- EA4245 Transplantation, Immunologie, Inflammation; Université de Tours, Tours, France.,Inserm UMR1069, Nutrition, Croissance et Cancer; Université de Tours, Tours, France
| | - Lobna Ouldamer
- Inserm UMR1069, Nutrition, Croissance et Cancer; Université de Tours, Tours, France.,CHRU de Tours, Tours, France
| | - Caroline Goupille
- Inserm UMR1069, Nutrition, Croissance et Cancer; Université de Tours, Tours, France.,CHRU de Tours, Tours, France
| | - Marie-Lise Jourdan
- Inserm UMR1069, Nutrition, Croissance et Cancer; Université de Tours, Tours, France.,CHRU de Tours, Tours, France
| | - Stéphan Chevalier
- Inserm UMR1069, Nutrition, Croissance et Cancer; Université de Tours, Tours, France
| | - Driffa Moussata
- EA4245 Transplantation, Immunologie, Inflammation; Université de Tours, Tours, France.,CHRU de Tours, Tours, France
| | - Pierre Besson
- Inserm UMR1069, Nutrition, Croissance et Cancer; Université de Tours, Tours, France
| | - Sébastien Roger
- EA4245 Transplantation, Immunologie, Inflammation; Université de Tours, Tours, France. .,Institut Universitaire de France, Paris, France.
| |
Collapse
|
45
|
Piggott BJ, Peters CJ, He Y, Huang X, Younger S, Jan LY, Jan YN. Paralytic, the Drosophila voltage-gated sodium channel, regulates proliferation of neural progenitors. Genes Dev 2019; 33:1739-1750. [PMID: 31753914 PMCID: PMC6942049 DOI: 10.1101/gad.330597.119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/10/2019] [Accepted: 10/28/2019] [Indexed: 12/23/2022]
Abstract
Proliferating cells, typically considered "nonexcitable," nevertheless, exhibit regulation by bioelectric signals. Notably, voltage-gated sodium channels (VGSC) that are crucial for neuronal excitability are also found in progenitors and up-regulated in cancer. Here, we identify a role for VGSC in proliferation of Drosophila neuroblast (NB) lineages within the central nervous system. Loss of paralytic (para), the sole gene that encodes Drosophila VGSC, reduces neuroblast progeny cell number. The type II neuroblast lineages, featuring a population of transit-amplifying intermediate neural progenitors (INP) similar to that found in the developing human cortex, are particularly sensitive to para manipulation. Following a series of asymmetric divisions, INPs normally exit the cell cycle through a final symmetric division. Our data suggests that loss of Para induces apoptosis in this population, whereas overexpression leads to an increase in INPs and overall neuroblast progeny cell numbers. These effects are cell autonomous and depend on Para channel activity. Reduction of Para expression not only affects normal NB development, but also strongly suppresses brain tumor mass, implicating a role for Para in cancer progression. To our knowledge, our studies are the first to identify a role for VGSC in neural progenitor proliferation. Elucidating the contribution of VGSC in proliferation will advance our understanding of bioelectric signaling within development and disease states.
Collapse
Affiliation(s)
- Beverly J Piggott
- Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
- Howard Hughes Medical Institute
| | - Christian J Peters
- Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
| | - Ye He
- Neuroscience Initiative, Advanced Science Research Center, the Graduate Center, City University of New York, New York 10031, New York
| | - Xi Huang
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5S 3E1, Canada
| | - Susan Younger
- Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
- Howard Hughes Medical Institute
| | - Lily Yeh Jan
- Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
- Howard Hughes Medical Institute
| | - Yuh Nung Jan
- Department of Physiology, Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, California 94158, USA
- Howard Hughes Medical Institute
| |
Collapse
|
46
|
Wang W, Xie G, Ren Z, Xie T, Li J. Gene Selection for the Discrimination of Colorectal Cancer. Curr Mol Med 2019; 20:415-428. [PMID: 31746296 DOI: 10.2174/1566524019666191119105209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/20/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer worldwide. Cancer discrimination is a typical application of gene expression analysis using a microarray technique. However, microarray data suffer from the curse of dimensionality and usual imbalanced class distribution between the majority (tumor samples) and minority (normal samples) classes. Feature gene selection is necessary and important for cancer discrimination. OBJECTIVES To select feature genes for the discrimination of CRC. METHODS We improve the feature selection algorithm based on differential evolution, DEFSw by using RUSBoost classifier and weight accuracy instead of the common classifier and evaluation measure for selecting feature genes from imbalance data. We firstly extract differently expressed genes (DEGs) from the CRC dataset of the TCGA and then select the feature genes from the DEGs using the improved DEFSw algorithm. Finally, we validate the selected feature gene sets using independent datasets and retrieve the cancer related information for these genes based on text mining through the Coremine Medical online database. RESULTS We select out 16 single-gene feature sets for colorectal cancer discrimination and 19 single-gene feature sets only for colon cancer discrimination. CONCLUSIONS In summary, we find a series of high potential candidate biomarkers or signatures, which can discriminate either or both of colon cancer and rectal cancer with high sensitivity and specificity.
Collapse
Affiliation(s)
- Wenhui Wang
- Network Information Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,National Engineering Research Center of Digital Life, Sun Yat-sen University, Guangzhou, China.,Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guanglei Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhonglu Ren
- College of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tingyan Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
47
|
Lin S, Lv Y, Xu J, Mao X, Chen Z, Lu W. Over-expression of Nav1.6 channels is associated with lymph node metastases in colorectal cancer. World J Surg Oncol 2019; 17:175. [PMID: 31672162 PMCID: PMC6824047 DOI: 10.1186/s12957-019-1715-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/14/2019] [Accepted: 09/27/2019] [Indexed: 11/15/2022] Open
Abstract
Background and objectives Lymph node metastasis is a key factor in predicting and determining the prognosis of patients with colorectal cancer (CRC). Sodium channels are highly expressed in a variety of tumors and are closely related to tumor development, metastasis, and invasion. We investigated the relationship between the expressions of different subtypes of Nav channels and lymph node metastasis of CRC. Methods Real-time PCR (RT-qPCR) was carried out to measure the expressions of different sodium channel subtypes, chemokine receptors (CCR2, CCR4, CCR7), and lymphocyte infiltration-related biomarkers (CD3e, CD8a, IL-2RA) in CRC tissues from 97 patients. The expressions of Nav1.5 and Nav1.6 in surgically isolated lymph nodes were detected by immunohistochemistry. Correlation analysis between expressions of different genes and lymph node metastasis was performed by two-tailed t test. Results Nav1.1 and Nav1.6 were highly expressed in CRC tissues and positively correlated with CRC lymph node metastasis. Nav1.6 was also highly expressed in metastatic lymph nodes. Further analysis showed that the high expression of Nav1.6 was closely related to the one of CCR2\CCR4 in tumor lymph node metastasis. Conclusions These results suggested that Nav1.6 might be a novel marker for CRC lymph node metastasis.
Collapse
Affiliation(s)
- Shuiquan Lin
- Department of Anorectal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Yangbo Lv
- Department of Anorectal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Jianguang Xu
- Department of Digestive System, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Xinglong Mao
- Department of Gastrointestinal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China
| | - Zhenhong Chen
- Department of Anorectal Surgery, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, People's Republic of China.
| | - Wuguang Lu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, People's Republic of China.
| |
Collapse
|
48
|
Djamgoz MBA, Fraser SP, Brackenbury WJ. In Vivo Evidence for Voltage-Gated Sodium Channel Expression in Carcinomas and Potentiation of Metastasis. Cancers (Basel) 2019; 11:E1675. [PMID: 31661908 PMCID: PMC6895836 DOI: 10.3390/cancers11111675] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/16/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 12/16/2022] Open
Abstract
A wide body of evidence suggests that voltage-gated sodium channels (VGSCs) are expressed de novo in several human carcinomas where channel activity promotes a variety of cellular behaviours integral to the metastatic cascade. These include directional motility (including galvanotaxis), pH balance, extracellular proteolysis, and invasion. Contrary to the substantial in vitro data, however, evidence for VGSC involvement in the cancer process in vivo is limited. Here, we critically assess, for the first time, the available in vivo evidence, hierarchically from mRNA level to emerging clinical aspects, including protein-level studies, electrolyte content, animal tests, and clinical imaging. The evidence strongly suggests that different VGSC subtypes (mainly Nav1.5 and Nav1.7) are expressed de novo in human carcinoma tissues and generally parallel the situation in vitro. Consistent with this, tissue electrolyte (sodium) levels, quantified by clinical imaging, are significantly higher in cancer vs. matched non-cancer tissues. These are early events in the acquisition of metastatic potential by the cancer cells. Taken together, the multi-faceted evidence suggests that the VGSC expression has clinical (diagnostic and therapeutic) potential as a prognostic marker, as well as an anti-metastatic target. The distinct advantages offered by the VGSC include especially (1) its embryonic nature, demonstrated most clearly for the predominant neonatal Nav1.5 expression in breast and colon cancer, and (2) the specifically druggable persistent current that VGSCs develop under hypoxic conditions, as in growing tumours, which promotes invasiveness and metastasis.
Collapse
Affiliation(s)
- Mustafa B A Djamgoz
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Scott P Fraser
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - William J Brackenbury
- Department of Biology and York Biomedical Research Institute, University of York, Heslington, York, YO10 5DD, UK.
| |
Collapse
|
49
|
Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol 2019; 25:5732-5772. [PMID: 31636470 PMCID: PMC6801186 DOI: 10.3748/wjg.v25.i38.5732] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 05/05/2019] [Revised: 07/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023] Open
Abstract
In their seminal papers Hanahan and Weinberg described oncogenic processes a normal cell undergoes to be transformed into a cancer cell. The functions of ion channels in the gastrointestinal (GI) tract influence a variety of cellular processes, many of which overlap with these hallmarks of cancer. In this review we focus on the roles of the calcium (Ca2+), sodium (Na+), potassium (K+), chloride (Cl-) and zinc (Zn2+) transporters in GI cancer, with a special emphasis on the roles of the KCNQ1 K+ channel and CFTR Cl- channel in colorectal cancer (CRC). Ca2+ is a ubiquitous second messenger, serving as a signaling molecule for a variety of cellular processes such as control of the cell cycle, apoptosis, and migration. Various members of the TRP superfamily, including TRPM8, TRPM7, TRPM6 and TRPM2, have been implicated in GI cancers, especially through overexpression in pancreatic adenocarcinomas and down-regulation in colon cancer. Voltage-gated sodium channels (VGSCs) are classically associated with the initiation and conduction of action potentials in electrically excitable cells such as neurons and muscle cells. The VGSC NaV1.5 is abundantly expressed in human colorectal CRC cell lines as well as being highly expressed in primary CRC samples. Studies have demonstrated that conductance through NaV1.5 contributes significantly to CRC cell invasiveness and cancer progression. Zn2+ transporters of the ZIP/SLC39A and ZnT/SLC30A families are dysregulated in all major GI organ cancers, in particular, ZIP4 up-regulation in pancreatic cancer (PC). More than 70 K+ channel genes, clustered in four families, are found expressed in the GI tract, where they regulate a range of cellular processes, including gastrin secretion in the stomach and anion secretion and fluid balance in the intestinal tract. Several distinct types of K+ channels are found dysregulated in the GI tract. Notable are hERG1 upregulation in PC, gastric cancer (GC) and CRC, leading to enhanced cancer angiogenesis and invasion, and KCNQ1 down-regulation in CRC, where KCNQ1 expression is associated with enhanced disease-free survival in stage II, III, and IV disease. Cl- channels are critical for a range of cellular and tissue processes in the GI tract, especially fluid balance in the colon. Most notable is CFTR, whose deficiency leads to mucus blockage, microbial dysbiosis and inflammation in the intestinal tract. CFTR is a tumor suppressor in several GI cancers. Cystic fibrosis patients are at a significant risk for CRC and low levels of CFTR expression are associated with poor overall disease-free survival in sporadic CRC. Two other classes of chloride channels that are dysregulated in GI cancers are the chloride intracellular channels (CLIC1, 3 & 4) and the chloride channel accessory proteins (CLCA1,2,4). CLIC1 & 4 are upregulated in PC, GC, gallbladder cancer, and CRC, while the CLCA proteins have been reported to be down-regulated in CRC. In summary, it is clear, from the diverse influences of ion channels, that their aberrant expression and/or activity can contribute to malignant transformation and tumor progression. Further, because ion channels are often localized to the plasma membrane and subject to multiple layers of regulation, they represent promising clinical targets for therapeutic intervention including the repurposing of current drugs.
Collapse
Affiliation(s)
- Kyle J Anderson
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Robert T Cormier
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| | - Patricia M Scott
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, United States
| |
Collapse
|
50
|
Xie MX, Zhang XL, Xu J, Zeng WA, Li D, Xu T, Pang RP, Ma K, Liu XG. Nuclear Factor-kappaB Gates Na v1.7 Channels in DRG Neurons via Protein-Protein Interaction. iScience 2019; 19:623-633. [PMID: 31446225 PMCID: PMC6715905 DOI: 10.1016/j.isci.2019.08.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/12/2019] [Revised: 07/04/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
It is well known that nuclear factor-kappaB (NF-κB) regulates neuronal structures and functions by nuclear transcription. Here, we showed that phospho-p65 (p-p65), an active form of NF-κB subunit, reversibly interacted with Nav1.7 channels in the membrane of dorsal root ganglion (DRG) neurons of rats. The interaction increased Nav1.7 currents by slowing inactivation of Nav1.7 channels and facilitating their recovery from inactivation, which may increase the resting state of the channels ready for activation. In cultured DRG neurons TNF-α upregulated the membrane p-p65 and enhanced Nav1.7 currents within 5 min but did not affect nuclear NF-κB within 40 min. This non-transcriptional effect on Nav1.7 may underlie a rapid regulation of the sensibility of the somatosensory system. Both NF-κB and Nav1.7 channels are critically implicated in many physiological functions and diseases. Our finding may shed new light on the investigation into the underlying mechanisms. NF-κB p-p65 interacts with Nav1.7 in the membrane of DRG neurons The interaction is reversible, depending on the cytoplasmic p-p65 content Reducing cytoplasmic p-p65 rapidly attenuates the interaction and Nav1.7 currents The rapid effect on Nav1.7 channels is independent of p-p65 nuclear translocation
Collapse
Affiliation(s)
- Man-Xiu Xie
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Xiao-Long Zhang
- Medical Research Center of Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan Road 2, Guangzhou 510080, China
| | - Jing Xu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Wei-An Zeng
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Dai Li
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Ting Xu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Rui-Ping Pang
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China
| | - Ke Ma
- Department of Pain Management, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 91603, China.
| | - Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou 510080, China; Department of Pain Management, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 91603, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, 74 Zhongshan Road 2, Guangzhou 510080, China.
| |
Collapse
|