1
|
De Fazio E, Pittarello M, Gans A, Ghosh B, Slika H, Alimonti P, Tyler B. Intrinsic and Microenvironmental Drivers of Glioblastoma Invasion. Int J Mol Sci 2024; 25:2563. [PMID: 38473812 PMCID: PMC10932253 DOI: 10.3390/ijms25052563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Gliomas are diffusely infiltrating brain tumors whose prognosis is strongly influenced by their extent of invasion into the surrounding brain tissue. While lower-grade gliomas present more circumscribed borders, high-grade gliomas are aggressive tumors with widespread brain infiltration and dissemination. Glioblastoma (GBM) is known for its high invasiveness and association with poor prognosis. Its low survival rate is due to the certainty of its recurrence, caused by microscopic brain infiltration which makes surgical eradication unattainable. New insights into GBM biology at the single-cell level have enabled the identification of mechanisms exploited by glioma cells for brain invasion. In this review, we explore the current understanding of several molecular pathways and mechanisms used by tumor cells to invade normal brain tissue. We address the intrinsic biological drivers of tumor cell invasion, by tackling how tumor cells interact with each other and with the tumor microenvironment (TME). We focus on the recently discovered neuronal niche in the TME, including local as well as distant neurons, contributing to glioma growth and invasion. We then address the mechanisms of invasion promoted by astrocytes and immune cells. Finally, we review the current literature on the therapeutic targeting of the molecular mechanisms of invasion.
Collapse
Affiliation(s)
- Emerson De Fazio
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy; (E.D.F.); (P.A.)
| | - Matilde Pittarello
- Department of Medicine, Humanitas University School of Medicine, 20089 Rozzano, Italy;
| | - Alessandro Gans
- Department of Neurology, University of Milan, 20122 Milan, Italy;
| | - Bikona Ghosh
- School of Medicine and Surgery, Dhaka Medical College, Dhaka 1000, Bangladesh;
| | - Hasan Slika
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Paolo Alimonti
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy; (E.D.F.); (P.A.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| |
Collapse
|
2
|
Ion Channels in Gliomas-From Molecular Basis to Treatment. Int J Mol Sci 2023; 24:ijms24032530. [PMID: 36768856 PMCID: PMC9916861 DOI: 10.3390/ijms24032530] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
Ion channels provide the basis for the nervous system's intrinsic electrical activity. Neuronal excitability is a characteristic property of neurons and is critical for all functions of the nervous system. Glia cells fulfill essential supportive roles, but unlike neurons, they also retain the ability to divide. This can lead to uncontrolled growth and the formation of gliomas. Ion channels are involved in the unique biology of gliomas pertaining to peritumoral pathology and seizures, diffuse invasion, and treatment resistance. The emerging picture shows ion channels in the brain at the crossroads of neurophysiology and fundamental pathophysiological processes of specific cancer behaviors as reflected by uncontrolled proliferation, infiltration, resistance to apoptosis, metabolism, and angiogenesis. Ion channels are highly druggable, making them an enticing therapeutic target. Targeting ion channels in difficult-to-treat brain tumors such as gliomas requires an understanding of their extremely heterogenous tumor microenvironment and highly diverse molecular profiles, both representing major causes of recurrence and treatment resistance. In this review, we survey the current knowledge on ion channels with oncogenic behavior within the heterogeneous group of gliomas, review ion channel gene expression as genomic biomarkers for glioma prognosis and provide an update on therapeutic perspectives for repurposed and novel ion channel inhibitors and electrotherapy.
Collapse
|
3
|
Stark RJ, Nguyen HN, Bacon MK, Rohrbough JC, Choi H, Lamb FS. Chloride Channel-3 (ClC-3) Modifies the Trafficking of Leucine-Rich Repeat-Containing 8A (LRRC8A) Anion Channels. J Membr Biol 2022; 256:125-135. [PMID: 36322172 PMCID: PMC10085862 DOI: 10.1007/s00232-022-00271-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/16/2022] [Indexed: 11/07/2022]
Abstract
Chloride channel-3 (ClC-3) Cl-/H+ antiporters and leucine-rich repeat-containing 8 (LRRC8) family anion channels have both been associated with volume-regulated anion currents (VRACs). VRACs are often altered in ClC-3 null cells but are absent in LRRC8A null cells. To explore the relationship between ClC-3, LRRC8A, and VRAC we localized tagged proteins in human epithelial kidney (HEK293) cells using multimodal microscopy. Expression of ClC-3-GFP induced large multivesicular bodies (MVBs) with ClC-3 in the delimiting membrane. LRRC8A-RFP localized to the plasma membrane and to small cytoplasmic vesicles. Co-expression demonstrated co-localization in small, highly mobile cytoplasmic vesicles that associated with the early endosomal marker Rab5A. However, most of the small LRRC8A-positive vesicles were constrained within large MVBs with abundant ClC-3 in the delimiting membrane. Dominant negative (S34A) Rab5A prevented ClC-3 overexpression from creating enlarged MVBs, while constitutively active (Q79L) Rab5A enhanced this phenotype. Thus, ClC-3 and LRRC8A are endocytosed together but independently sorted in Rab5A MVBs. Subsequently, LRRC8A-labeled vesicles were sorted to MVBs labeled by Rab27A and B exosomal compartment markers, but not to Rab11 recycling endosomes. VRAC currents were significantly larger in ClC-3 null HEK293 cells. This work demonstrates dependence of LRRC8A trafficking on ClC-3 which may explain the association between ClC-3 and VRACs.
Collapse
Affiliation(s)
- Ryan J Stark
- Department of Pediatrics, Vanderbilt University Medical Center, 2215 Garland Avenue, Light Hall-1055D, Nashville, TN, 37232-3122v, USA
| | - Hong N Nguyen
- Department of Pediatrics, Vanderbilt University Medical Center, 2215 Garland Avenue, Light Hall-1055D, Nashville, TN, 37232-3122v, USA
| | - Matthew K Bacon
- Department of Pediatrics, University of Kentucky, Lexington, KY, 40536, USA
| | - Jeffrey C Rohrbough
- Department of Pediatrics, Vanderbilt University Medical Center, 2215 Garland Avenue, Light Hall-1055D, Nashville, TN, 37232-3122v, USA
| | - Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, 2215 Garland Avenue, Light Hall-1055D, Nashville, TN, 37232-3122v, USA
| | - Fred S Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, 2215 Garland Avenue, Light Hall-1055D, Nashville, TN, 37232-3122v, USA.
| |
Collapse
|
4
|
Liu J, Tao M, Zhao W, Song Q, Yang X, Li M, Zhang Y, Xiu D, Zhang Z. Calcium Channel α2δ1 is Essential for Pancreatic Tumor-Initiating Cells through Sequential Phosphorylation of PKM2. Cell Mol Gastroenterol Hepatol 2022; 15:373-392. [PMID: 36244646 PMCID: PMC9791133 DOI: 10.1016/j.jcmgh.2022.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND & AIMS Tumor-initiating cells (TICs) drive pancreatic cancer tumorigenesis, therapeutic resistance, and metastasis. However, TICs are highly plastic and heterogenous, which impede the robust identification and targeted therapy of such a population. The aim of this study is to identify the surface marker and therapeutic target for pancreatic TICs. METHODS We isolated voltage-gated calcium channel α2δ1 subunit (isoform 5)-positive subpopulation from pancreatic cancer cell lines and freshly resected primary tissues by fluorescence-activated cell sorting and evaluated their TIC properties by spheroid formation and tumorigenic assays. Coimmunoprecipitation was used to identify the direct substrate of CaMKⅡδ. RESULTS We demonstrate that the voltage-gated calcium channel α2δ1 subunit (isoform 5) marks a subpopulation of pancreatic TICs with the highest TIC frequency among the known pancreatic TIC markers tested. Furthermore, α2δ1 is functionally sufficient and indispensable to promote TIC properties by mediating Ca2+ influx, which activates CaMKⅡδ to directly phosphorylate PKM2 at T454 that results in subsequent phosphorylation at Y105 to translocate into nucleus, enhancing the stem-like properties. Interestingly, blocking α2δ1 with its specific antibody has remarkably therapeutic effects on pancreatic cancer xenografts by reducing TICs. CONCLUSIONS α2δ1 promotes pancreatic TIC properties through sequential phosphorylation of PKM2 mediated by CaMKⅡδ, and targeting α2δ1 provides a therapeutic strategy against TICs for pancreatic cancer.
Collapse
Affiliation(s)
- Jingtao Liu
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, P.R. China; Department of Pharmacology, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| | - Ming Tao
- Department of General Surgery, Peking University Third Hospital, Beijing, P.R. China
| | - Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| | - Qingru Song
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| | - Xiaodan Yang
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| | - Meng Li
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| | - Yanhua Zhang
- Department of Pharmacology, Peking University Cancer Hospital and Institute, Beijing, P.R. China.
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, P.R. China.
| | - Zhiqian Zhang
- Key Laboratory of Carcinogenesis and Translational Research, (Ministry of Education/Beijing), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing, P.R. China.
| |
Collapse
|
5
|
Ogata G, Partida GJ, Fasoli A, Ishida AT. Calcium/calmodulin-dependent protein kinase II associates with the K + channel isoform Kv4.3 in adult rat optic nerve. Front Neuroanat 2022; 16:958986. [PMID: 36172564 PMCID: PMC9512010 DOI: 10.3389/fnana.2022.958986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Spikes are said to exhibit "memory" in that they can be altered by spikes that precede them. In retinal ganglion cell axons, for example, rapid spiking can slow the propagation of subsequent spikes. This increases inter-spike interval and, thus, low-pass filters instantaneous spike frequency. Similarly, a K+ ion channel blocker (4-aminopyridine, 4AP) increases the time-to-peak of compound action potentials recorded from optic nerve, and we recently found that reducing autophosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII) does too. These results would be expected if CaMKII modulates spike propagation by regulating 4AP-sensitive K+ channels. As steps toward identifying a possible substrate, we test whether (i) 4AP alters optic nerve spike shape in ways consistent with reducing K+ current, (ii) 4AP alters spike propagation consistent with effects of reducing CaMKII activation, (iii) antibodies directed against 4AP-sensitive and CaMKII-regulated K+ channels bind to optic nerve axons, and (iv) optic nerve CaMKII co-immunoprecipitates with 4AP-sensitive K+ channels. We find that, in adult rat optic nerve, (i) 4AP selectively slows spike repolarization, (ii) 4AP slows spike propagation, (iii) immunogen-blockable staining is achieved with anti-Kv4.3 antibodies but not with antibodies directed against Kv1.4 or Kv4.2, and (iv) CaMKII associates with Kv4.3. Kv4.3 may thus be a substrate that underlies activity-dependent spike regulation in adult visual system pathways.
Collapse
Affiliation(s)
- Genki Ogata
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA, United States
| | - Gloria J. Partida
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA, United States
| | - Anna Fasoli
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA, United States
| | - Andrew T. Ishida
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA, United States
- Department of Ophthalmology and Vision Science, University of California, Sacramento, Sacramento, CA, United States
| |
Collapse
|
6
|
Sinha M, Zabini D, Guntur D, Nagaraj C, Enyedi P, Olschewski H, Kuebler WM, Olschewski A. Chloride channels in the lung: Challenges and perspectives for viral infections, pulmonary arterial hypertension, and cystic fibrosis. Pharmacol Ther 2022; 237:108249. [PMID: 35878810 DOI: 10.1016/j.pharmthera.2022.108249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
Fine control over chloride homeostasis in the lung is required to maintain membrane excitability, transepithelial transport as well as intra- and extracellular ion and water homeostasis. Over the last decades, a growing number of chloride channels and transporters have been identified in the cells of the pulmonary vasculature and the respiratory tract. The importance of these proteins is underpinned by the fact that impairment of their physiological function is associated with functional dysregulation, structural remodeling, or hereditary diseases of the lung. This paper reviews the field of chloride channels and transporters in the lung and discusses chloride channels in disease processes such as viral infections including SARS-CoV- 2, pulmonary arterial hypertension, cystic fibrosis and asthma. Although chloride channels have become a hot research topic in recent years, remarkably few of them have been targeted by pharmacological agents. As such, we complement the putative pathophysiological role of chloride channels here with a summary of their therapeutic potential.
Collapse
Affiliation(s)
- Madhushri Sinha
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Diana Zabini
- Department of Physiology, Neue Stiftingtalstrasse 6/V, 8010 Graz, Austria.
| | - Divya Guntur
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria.
| | - Chandran Nagaraj
- Ludwig Boltzmann Institute for Lung Vascular Research, Neue Stiftingtalstraße 6, 8010 Graz, Austria.
| | - Peter Enyedi
- Department of Physiology, Semmelweis University, Tűzoltó utca 37-47, 1094 Budapest, Hungary.
| | - Horst Olschewski
- Department of Internal Medicine, Division of Pulmonology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria.
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Andrea Olschewski
- Experimental Anaesthesiology, Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Auenbruggerplatz 5, 8036 Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Neue Stiftingtalstraße 6, 8010 Graz, Austria.
| |
Collapse
|
7
|
Brignone MS, Lanciotti A, Michelucci A, Mallozzi C, Camerini S, Catacuzzeno L, Sforna L, Caramia M, D’Adamo MC, Ceccarini M, Molinari P, Macioce P, Macchia G, Petrucci TC, Pessia M, Visentin S, Ambrosini E. The CaMKII/MLC1 Axis Confers Ca2+-Dependence to Volume-Regulated Anion Channels (VRAC) in Astrocytes. Cells 2022; 11:cells11172656. [PMID: 36078064 PMCID: PMC9454758 DOI: 10.3390/cells11172656] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Astrocytes, the main glial cells of the central nervous system, play a key role in brain volume control due to their intimate contacts with cerebral blood vessels and the expression of a distinctive equipment of proteins involved in solute/water transport. Among these is MLC1, a protein highly expressed in perivascular astrocytes and whose mutations cause megalencephalic leukoencephalopathy with subcortical cysts (MLC), an incurable leukodystrophy characterized by macrocephaly, chronic brain edema, cysts, myelin vacuolation, and astrocyte swelling. Although, in astrocytes, MLC1 mutations are known to affect the swelling-activated chloride currents (ICl,swell) mediated by the volume-regulated anion channel (VRAC), and the regulatory volume decrease, MLC1′s proper function is still unknown. By combining molecular, biochemical, proteomic, electrophysiological, and imaging techniques, we here show that MLC1 is a Ca2+/Calmodulin-dependent protein kinase II (CaMKII) target protein, whose phosphorylation, occurring in response to intracellular Ca2+ release, potentiates VRAC-mediated ICl,swell. Overall, these findings reveal that MLC1 is a Ca2+-regulated protein, linking volume regulation to Ca2+ signaling in astrocytes. This knowledge provides new insight into the MLC1 protein function and into the mechanisms controlling ion/water exchanges in the brain, which may help identify possible molecular targets for the treatment of MLC and other pathological conditions caused by astrocyte swelling and brain edema.
Collapse
Affiliation(s)
| | - Angela Lanciotti
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Antonio Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Cinzia Mallozzi
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Serena Camerini
- Core Facilities (FAST), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Martino Caramia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Maria Cristina D’Adamo
- Department of Medicine and Surgery, LUM Giuseppe Degennaro University, 70010 Bari, Italy
| | - Marina Ceccarini
- National Centre for Rare Diseases, Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Paola Molinari
- National Centre for Drug Research and Evaluation (FARVA), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Pompeo Macioce
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
| | | | | | - Mauro Pessia
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates
| | - Sergio Visentin
- National Centre for Drug Research and Evaluation (FARVA), Istituto Superiore di Sanità, 00169 Rome, Italy
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy
- Correspondence: ; Tel.: +39-06-4990-2037
| |
Collapse
|
8
|
Jeong JY, Park H, Yoo H, Kim EJ, Jeon B, Lee JD, Kang D, Lee CJ, Paek SH, Roh EJ, Yi GS, Kang SS. Trifluoperazine and Its Analog Suppressed the Tumorigenicity of Non-Small Cell Lung Cancer Cell; Applicability of Antipsychotic Drugs to Lung Cancer Treatment. Biomedicines 2022; 10:biomedicines10051046. [PMID: 35625784 PMCID: PMC9138877 DOI: 10.3390/biomedicines10051046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Despite significant advances in diagnostic and therapeutic technologies, lung cancer remains the leading cause of cancer-related mortality worldwide. Non-small cell lung cancer (NSCLC) accounts for approximately 85% of lung cancer cases. Recently, some antipsychotics have been shown to possess anticancer activity. However, the effects of antipsychotics on NSCLC need to be further explored. We examined the effects of trifluoperazine (TFP), a commonly used antipsychotic drug, and its synthetic analogs on A549 human lung cancer cells. In addition, cell proliferation analysis, colony formation assay, flow cytometry, western blot analysis, and in vivo xenograft experiments were performed. Key genes and mechanisms possibly affected by TFP are significantly related to better survival outcomes in lung cancer patients. Treatment with TFP and a selected TFP analog 3dc significantly inhibited the proliferation, anchorage-dependent/independent colony formation, and migration of A549 cells. Treatment with 3dc affected the expression of genes related to the apoptosis and survival of A549 cells. Treatment with 3dc promoted apoptosis and DNA fragmentation. In all experiments, including in vivo studies of metastatic lung cancer development, 3dc had more substantial anticancer effects than TFP. According to our analysis of publicly available clinical data and in vitro and in vivo experiments, we suggest that some kinds of antipsychotics prevent the progression of NSCLC. Furthermore, this study indicates a synthetic TFP analog that could be a potential therapeutic for lung cancer.
Collapse
Affiliation(s)
- Joo Yeon Jeong
- Department of Anatomy & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
| | - Haangik Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea;
| | - Hong Yoo
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gyeongsang National University Hospital, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (H.Y.); (J.D.L.)
| | - Eun-Jin Kim
- Department of Physiology & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (E.-J.K.); (D.K.)
| | - Borami Jeon
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea; (B.J.); (E.J.R.)
| | - Jong Deog Lee
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gyeongsang National University Hospital, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (H.Y.); (J.D.L.)
| | - Dawon Kang
- Department of Physiology & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea; (E.-J.K.); (D.K.)
| | - Changjoon Justin Lee
- Center for Glia-Neuron Interaction and Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea;
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34141, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, College of Medicine, Seoul National University, Seoul 03080, Korea;
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul 02792, Korea; (B.J.); (E.J.R.)
| | - Gwan-Su Yi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea;
- Correspondence: (G.-S.Y.); (S.S.K.); Tel.: +82-42-350-4318 (G.-S.Y.); +82-55-772-8033 (S.S.K.)
| | - Sang Soo Kang
- Department of Anatomy & Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea;
- Correspondence: (G.-S.Y.); (S.S.K.); Tel.: +82-42-350-4318 (G.-S.Y.); +82-55-772-8033 (S.S.K.)
| |
Collapse
|
9
|
Sheth M, Esfandiari L. Bioelectric Dysregulation in Cancer Initiation, Promotion, and Progression. Front Oncol 2022; 12:846917. [PMID: 35359398 PMCID: PMC8964134 DOI: 10.3389/fonc.2022.846917] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is primarily a disease of dysregulation – both at the genetic level and at the tissue organization level. One way that tissue organization is dysregulated is by changes in the bioelectric regulation of cell signaling pathways. At the basis of bioelectricity lies the cellular membrane potential or Vmem, an intrinsic property associated with any cell. The bioelectric state of cancer cells is different from that of healthy cells, causing a disruption in the cellular signaling pathways. This disruption or dysregulation affects all three processes of carcinogenesis – initiation, promotion, and progression. Another mechanism that facilitates the homeostasis of cell signaling pathways is the production of extracellular vesicles (EVs) by cells. EVs also play a role in carcinogenesis by mediating cellular communication within the tumor microenvironment (TME). Furthermore, the production and release of EVs is altered in cancer. To this end, the change in cell electrical state and in EV production are responsible for the bioelectric dysregulation which occurs during cancer. This paper reviews the bioelectric dysregulation associated with carcinogenesis, including the TME and metastasis. We also look at the major ion channels associated with cancer and current technologies and tools used to detect and manipulate bioelectric properties of cells.
Collapse
Affiliation(s)
- Maulee Sheth
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
| | - Leyla Esfandiari
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, United States
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH, United States
- *Correspondence: Leyla Esfandiari,
| |
Collapse
|
10
|
Huang J, Sheng X, Zhuo Z, Xiao D, Wu K, Wan G, Chen H. ClC-c regulates the proliferation of intestinal stem cells via the EGFR signalling pathway in Drosophila. Cell Prolif 2021; 55:e13173. [PMID: 34952996 PMCID: PMC8780901 DOI: 10.1111/cpr.13173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/04/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023] Open
Abstract
Objectives Adult stem cells uphold a delicate balance between quiescent and active states, which is crucial for tissue homeostasis. Whereas many signalling pathways that regulate epithelial stem cells have been reported, many regulators remain unidentified. Materials and Methods Flies were used to generate tissue‐specific gene knockdown and gene knockout. qRT‐PCR was used to assess the relative mRNA levels. Immunofluorescence was used to determine protein localization and expression patterns. Clonal analyses were used to observe the phenotype. RNA‐seq was used to screen downstream mechanisms. Results Here, we report a member of the chloride channel family, ClC‐c, which is specifically expressed in Drosophila intestinal stem/progenitor cells and regulates intestinal stem cell (ISC) proliferation under physiological conditions and upon tissue damage. Mechanistically, we found that the ISC loss induced by the depletion of ClC‐c in intestinal stem/progenitor cells is due to inhibition of the EGFR signalling pathway. Conclusion Our findings reveal an ISC‐specific function of ClC‐c in regulating stem cell maintenance and proliferation, thereby providing new insights into the functional links among the chloride channel family, ISC proliferation and tissue homeostasis.
Collapse
Affiliation(s)
- Jinping Huang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao Sheng
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhangpeng Zhuo
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Danqing Xiao
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kun Wu
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Gang Wan
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haiyang Chen
- Laboratory of Metabolism and Aging Research, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Jenkins EPW, Finch A, Gerigk M, Triantis IF, Watts C, Malliaras GG. Electrotherapies for Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100978. [PMID: 34292672 PMCID: PMC8456216 DOI: 10.1002/advs.202100978] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/20/2021] [Indexed: 05/08/2023]
Abstract
Non-thermal, intermediate frequency (100-500 kHz) electrotherapies present a unique therapeutic strategy to treat malignant neoplasms. Here, pulsed electric fields (PEFs) which induce reversible or irreversible electroporation (IRE) and tumour-treating fields (TTFs) are reviewed highlighting the foundations, advances, and considerations of each method when applied to glioblastoma (GBM). Several biological aspects of GBM that contribute to treatment complexity (heterogeneity, recurrence, resistance, and blood-brain barrier(BBB)) and electrophysiological traits which are suggested to promote glioma progression are described. Particularly, the biological responses at the cellular and molecular level to specific parameters of the electrical stimuli are discussed offering ways to compare these parameters despite the lack of a universally adopted physical description. Reviewing the literature, a disconnect is found between electrotherapy techniques and how they target the biological complexities of GBM that make treatment difficult in the first place. An attempt is made to bridge the interdisciplinary gap by mapping biological characteristics to different methods of electrotherapy, suggesting important future research topics and directions in both understanding and treating GBM. To the authors' knowledge, this is the first paper that attempts an in-tandem assessment of the biological effects of different aspects of intermediate frequency electrotherapy methods, thus offering possible strategies toward GBM treatment.
Collapse
Affiliation(s)
- Elise P. W. Jenkins
- Division of Electrical EngineeringDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Alina Finch
- Institute of Cancer and Genomic ScienceUniversity of BirminghamBirminghamB15 2TTUK
| | - Magda Gerigk
- Division of Electrical EngineeringDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| | - Iasonas F. Triantis
- Department of Electrical and Electronic EngineeringCity, University of LondonLondonEC1V 0HBUK
| | - Colin Watts
- Institute of Cancer and Genomic ScienceUniversity of BirminghamBirminghamB15 2TTUK
| | - George G. Malliaras
- Division of Electrical EngineeringDepartment of EngineeringUniversity of CambridgeCambridgeCB3 0FAUK
| |
Collapse
|
12
|
Abstract
Chloride channel 3 (ClC-3), a Cl-/H+ antiporter, has been well established as a member of volume-regulated chloride channels (VRCCs). ClC-3 may be a crucial mediator for activating inflammation-associated signaling pathways by regulating protein phosphorylation. A growing number of studies have indicated that ClC-3 overexpression plays a crucial role in mediating increased plasma low-density lipoprotein levels, vascular endothelium dysfunction, pro-inflammatory activation of macrophages, hyper-proliferation and hyper-migration of vascular smooth muscle cells (VSMCs), as well as oxidative stress and foam cell formation, which are the main factors responsible for atherosclerotic plaque formation in the arterial wall. In the present review, we summarize the molecular structures and classical functions of ClC-3. We further discuss its emerging role in the atherosclerotic process. In conclusion, we explore the potential role of ClC-3 as a therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Dun Niu
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, 34706University of South China, Hengyang, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, 34706University of South China, Hengyang, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, 34706University of South China, Hengyang, China
| |
Collapse
|
13
|
Lee D, Hong JH. Ca 2+ Signaling as the Untact Mode during Signaling in Metastatic Breast Cancer. Cancers (Basel) 2021; 13:1473. [PMID: 33806911 PMCID: PMC8004807 DOI: 10.3390/cancers13061473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 01/06/2023] Open
Abstract
Metastatic features of breast cancer in the brain are considered a common pathology in female patients with late-stage breast cancer. Ca2+ signaling and the overexpression pattern of Ca2+ channels have been regarded as oncogenic markers of breast cancer. In other words, breast tumor development can be mediated by inhibiting Ca2+ channels. Although the therapeutic potential of inhibiting Ca2+ channels against breast cancer has been demonstrated, the relationship between breast cancer metastasis and Ca2+ channels is not yet understood. Thus, we focused on the metastatic features of breast cancer and summarized the basic mechanisms of Ca2+-related proteins and channels during the stages of metastatic breast cancer by evaluating Ca2+ signaling. In particular, we highlighted the metastasis of breast tumors to the brain. Thus, modulating Ca2+ channels with Ca2+ channel inhibitors and combined applications will advance treatment strategies for breast cancer metastasis to the brain.
Collapse
Affiliation(s)
| | - Jeong Hee Hong
- Department of Health Sciences and Technology, Lee Gil Ya Cancer and Diabetes Institute, GAIHST, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Korea;
| |
Collapse
|
14
|
Petralia F, Tignor N, Reva B, Koptyra M, Chowdhury S, Rykunov D, Krek A, Ma W, Zhu Y, Ji J, Calinawan A, Whiteaker JR, Colaprico A, Stathias V, Omelchenko T, Song X, Raman P, Guo Y, Brown MA, Ivey RG, Szpyt J, Guha Thakurta S, Gritsenko MA, Weitz KK, Lopez G, Kalayci S, Gümüş ZH, Yoo S, da Veiga Leprevost F, Chang HY, Krug K, Katsnelson L, Wang Y, Kennedy JJ, Voytovich UJ, Zhao L, Gaonkar KS, Ennis BM, Zhang B, Baubet V, Tauhid L, Lilly JV, Mason JL, Farrow B, Young N, Leary S, Moon J, Petyuk VA, Nazarian J, Adappa ND, Palmer JN, Lober RM, Rivero-Hinojosa S, Wang LB, Wang JM, Broberg M, Chu RK, Moore RJ, Monroe ME, Zhao R, Smith RD, Zhu J, Robles AI, Mesri M, Boja E, Hiltke T, Rodriguez H, Zhang B, Schadt EE, Mani DR, Ding L, Iavarone A, Wiznerowicz M, Schürer S, Chen XS, Heath AP, Rokita JL, Nesvizhskii AI, Fenyö D, Rodland KD, Liu T, Gygi SP, Paulovich AG, Resnick AC, Storm PB, Rood BR, Wang P. Integrated Proteogenomic Characterization across Major Histological Types of Pediatric Brain Cancer. Cell 2020; 183:1962-1985.e31. [PMID: 33242424 PMCID: PMC8143193 DOI: 10.1016/j.cell.2020.10.044] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/19/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
We report a comprehensive proteogenomics analysis, including whole-genome sequencing, RNA sequencing, and proteomics and phosphoproteomics profiling, of 218 tumors across 7 histological types of childhood brain cancer: low-grade glioma (n = 93), ependymoma (32), high-grade glioma (25), medulloblastoma (22), ganglioglioma (18), craniopharyngioma (16), and atypical teratoid rhabdoid tumor (12). Proteomics data identify common biological themes that span histological boundaries, suggesting that treatments used for one histological type may be applied effectively to other tumors sharing similar proteomics features. Immune landscape characterization reveals diverse tumor microenvironments across and within diagnoses. Proteomics data further reveal functional effects of somatic mutations and copy number variations (CNVs) not evident in transcriptomics data. Kinase-substrate association and co-expression network analysis identify important biological mechanisms of tumorigenesis. This is the first large-scale proteogenomics analysis across traditional histological boundaries to uncover foundational pediatric brain tumor biology and inform rational treatment selection.
Collapse
Affiliation(s)
- Francesca Petralia
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicole Tignor
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Boris Reva
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mateusz Koptyra
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shrabanti Chowdhury
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dmitry Rykunov
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jiayi Ji
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anna Calinawan
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Antonio Colaprico
- Department of Public Health Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vasileios Stathias
- Department of Pharmacology, Institute for Data Science and Computing, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146, USA
| | - Tatiana Omelchenko
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xiaoyu Song
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pichai Raman
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yiran Guo
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Miguel A Brown
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Richard G Ivey
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - John Szpyt
- Thermo Fisher Scientific Center for Multiplexed Proteomics, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sanjukta Guha Thakurta
- Thermo Fisher Scientific Center for Multiplexed Proteomics, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Marina A Gritsenko
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Gonzalo Lopez
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Selim Kalayci
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Seungyeul Yoo
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Hui-Yin Chang
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Karsten Krug
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02412, USA
| | - Lizabeth Katsnelson
- Institute for Systems Genetics; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ying Wang
- Institute for Systems Genetics; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Jacob J Kennedy
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Lei Zhao
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Krutika S Gaonkar
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian M Ennis
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bo Zhang
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Valerie Baubet
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lamiya Tauhid
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jena V Lilly
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jennifer L Mason
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Bailey Farrow
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nathan Young
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sarah Leary
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Cancer and Blood Disorders Center, Seattle Children's Hospital, Seattle, WA 98105, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Jamie Moon
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Javad Nazarian
- Children's National Research Institute, George Washington University School of Medicine, Washington, DC 20010, USA; Department of Oncology, Children's Research Center, University Children's Hospital Zürich, Zürich 8032, Switzerland
| | - Nithin D Adappa
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James N Palmer
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert M Lober
- Department of Neurosurgery, Dayton Children's Hospital, Dayton, OH 45404, USA
| | - Samuel Rivero-Hinojosa
- Children's National Research Institute, George Washington University School of Medicine, Washington, DC 20010, USA
| | - Liang-Bo Wang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 631110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Joshua M Wang
- Institute for Systems Genetics; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Matilda Broberg
- Institute for Systems Genetics; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Rosalie K Chu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Rui Zhao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily Boja
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tara Hiltke
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - D R Mani
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02412, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 631110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Neurology, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Maciej Wiznerowicz
- Poznan University of Medical Sciences, 61-701 Poznań, Poland; International Institute for Molecular Oncology, 61-203 Poznań, Poland
| | - Stephan Schürer
- Department of Pharmacology, Institute for Data Science and Computing, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146, USA
| | - Xi S Chen
- Department of Public Health Science, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Allison P Heath
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - David Fenyö
- Institute for Systems Genetics; Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Karin D Rodland
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR 97221, USA
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Steven P Gygi
- Thermo Fisher Scientific Center for Multiplexed Proteomics, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine, Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Brian R Rood
- Children's National Research Institute, George Washington University School of Medicine, Washington, DC 20010, USA.
| | - Pei Wang
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
15
|
Azab MA, Alomari A, Azzam AY. Featuring how calcium channels and calmodulin affect glioblastoma behavior. A review article. Cancer Treat Res Commun 2020; 25:100255. [PMID: 33341039 DOI: 10.1016/j.ctarc.2020.100255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/17/2020] [Accepted: 11/30/2020] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is considered to be the most aggressive primary brain tumor with an extremely bad prognosis. Recurrence after treatment is a major problem with a survival rate for one year ranging about 39.7%. Ideal outcomes are still difficult to be achieved despite the recent treatment combinations. The ultimate capacity to regrow after resection is considered to be related to the availability of self-regenerating populations of stem cells. We made a literature review interpreting how calcium channels and calcium-regulated proteins mechanistically elaborate glioblastoma virulence in different ways. Calcium channels, and calcium-regulated proteins have shown diverse interconnected roles in shaping different aspects of GBM biology as indicated in some experimental studies. The beneficial prospective of those roles granting GBM different aggressive potentials pose variable applications in targeted therapy whether it is experimental or clinical trials.
Collapse
Affiliation(s)
| | | | - Ahmed Y Azzam
- October 6 University Faculty of Medicine, Giza, Egypt.
| |
Collapse
|
16
|
Communication of Glioma cells with neuronal plasticity: What is the underlying mechanism? Neurochem Int 2020; 141:104879. [PMID: 33068685 DOI: 10.1016/j.neuint.2020.104879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/26/2020] [Accepted: 10/09/2020] [Indexed: 12/21/2022]
Abstract
There has been a significantly rising discussion on how neuronal plasticity communicates with the glioma growth and invasion. This literature review aims to determine which neurotransmitters, ion channels and signaling pathways are involved in this context, how information is transferred from synaptic sites to the glioma cells and how glioma cells apply established mechanics of synaptic plasticity for their own increment. This work is a compilation of some outstanding findings related to the influence of the glutamate, calcium, potassium, chloride and sodium channels and other important brain plasticity molecules over the glioma progression. These topics also include the relevant molecular signaling data which could prove to be helpful for an effective clinical management of brain tumors in the future.
Collapse
|
17
|
Takayasu T, Kurisu K, Esquenazi Y, Ballester LY. Ion Channels and Their Role in the Pathophysiology of Gliomas. Mol Cancer Ther 2020; 19:1959-1969. [PMID: 33008831 PMCID: PMC7577395 DOI: 10.1158/1535-7163.mct-19-0929] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/24/2020] [Accepted: 08/06/2020] [Indexed: 01/10/2023]
Abstract
Malignant gliomas are the most common primary central nervous system tumors and their prognosis is very poor. In recent years, ion channels have been demonstrated to play important roles in tumor pathophysiology such as regulation of gene expression, cell migration, and cell proliferation. In this review, we summarize the current knowledge on the role of ion channels on the development and progression of gliomas. Cell volume changes through the regulation of ion flux, accompanied by water flux, are essential for migration and invasion. Signaling pathways affected by ion channel activity play roles in cell survival and cell proliferation. Moreover, ion channels are involved in glioma-related seizures, sensitivity to chemotherapy, and tumor metabolism. Ion channels are potential targets for the treatment of these lethal tumors. Despite our increased understanding of the contributions of ion channels to glioma biology, this field remains poorly studied. This review summarizes the current literature on this important topic.
Collapse
Affiliation(s)
- Takeshi Takayasu
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
- Department of Neurosurgery, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Kaoru Kurisu
- Department of Neurosurgery, Institute of Biomedical and Health Sciences, Hiroshima University, Higashihiroshima, Hiroshima, Japan
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas.
- Memorial Hermann Hospital-TMC, Houston, Texas
| | - Leomar Y Ballester
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston, Texas.
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Medical School, Houston, Texas
- Memorial Hermann Hospital-TMC, Houston, Texas
| |
Collapse
|
18
|
Catacuzzeno L, Sforna L, Esposito V, Limatola C, Franciolini F. Ion Channels in Glioma Malignancy. Rev Physiol Biochem Pharmacol 2020; 181:223-267. [DOI: 10.1007/112_2020_44] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Jiang X, Liu Y, Wang G, Yao Y, Mei C, Wu X, Ma W, Yuan Y. Up-regulation of CLIC1 activates MYC signaling and forms a positive feedback regulatory loop with MYC in Hepatocellular carcinoma. Am J Cancer Res 2020; 10:2355-2370. [PMID: 32905514 PMCID: PMC7471371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/05/2020] [Indexed: 06/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is leading cause of tumor-related deaths worldwide. The intracellular chloride channel protein (CLIC1) plays a role in the occurrence and progression of HCC, although the underlying mechanisms are still unclear. We evaluated the CLIC1 mRNA and protein levels in both patient tissues and HCC cell lines, and analyzed the correlation between CLIC1 expression and clinical features. The biological function of CLIC1 in HCC was examined in vivo and in vitro. The upstream regulatory factors were identified by bioinformatics programs, and downstream mechanisms affecting HCC behavior have also been explored and validated. CLIC1 was up-regulated in HCC tissues and cell lines, and promoted the proliferation, invasion and migration of HCC cells in vivo and in vitro. TP53 was identified as the upstream transcription factor of CLIC1. MiR-122-5p also regulated CLIC1 levels by degrading the transcripts. More importantly, we found that the increased CLIC1 was significantly related to the activation of MYC signaling. By binding with MYC, CLIC1 enhanced the transcription activity of MYC to downstream genes, rather than by altering its expression. Finally, a positive feedback regulatory loop between CLIC1 and MYC was established. CLIC1 is closely related to the occurrence, progression and prognosis of HCC, and a promising novel therapeutic target.
Collapse
Affiliation(s)
- Xiang Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
- Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan UniversityWuhan 430071, PR China
| | - Yingyi Liu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
- Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan UniversityWuhan 430071, PR China
| | - Ganggang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
- Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan UniversityWuhan 430071, PR China
| | - Ye Yao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
- Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan UniversityWuhan 430071, PR China
| | - Chengjie Mei
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
- Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan UniversityWuhan 430071, PR China
| | - Xiaoling Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
- Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan UniversityWuhan 430071, PR China
| | - Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430071, Hubei, PR China
| |
Collapse
|
20
|
Archanjo AB, Assis ALEMD, Oliveira MMD, Mendes SO, Borçoi AR, Maia LDL, Souza RPD, Cicco RD, Saito KC, Kimura ET, Carvalho MBD, Nunes FD, Tajara EH, Santos MD, Nogueira BV, Trivilin LO, Pinheiro CJG, Álvares-da-Silva AM. Elemental characterization of oral cavity squamous cell carcinoma and its relationship with smoking, prognosis and survival. Sci Rep 2020; 10:10382. [PMID: 32587307 PMCID: PMC7316707 DOI: 10.1038/s41598-020-67270-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 06/03/2020] [Indexed: 11/29/2022] Open
Abstract
Oral cancer squamous cell carcinoma (OCSCC) mainly affects individuals aged between 50 and 70 years who consume tobacco and alcohol. Tobacco smoke contains hundreds of known toxic and carcinogenic molecules, and a few studies have sought to verify the relationship of such trace elements as risk or prognostic factors for head and neck cancer. We obtained 78 samples of tumor tissues from patients with OCSCC, and performed a qualitative elemental characterization using the micro X-Ray Fluorescence technique based on synchrotron radiation. We found the presence of magnesium, phosphorus, sulfur, chlorine, potassium, calcium, chromium, manganese, iron, zinc, cobalt, nickel, copper, arsenic and bromine in OCSCC samples. Magnesium, chlorine, chromium, manganese, nickel, arsenic and bromine are associated with smoking. We observed a significant association between relapse and chlorine and chromium. The presence of chlorine in the samples was an independent protective factor against relapse (OR = 0.105, CI = 0.01-0.63) and for best disease-free survival (HR = 0.194, CI = 0.04-0.87). Reporting for the first time in oral cancer, these results suggest a key relationship between smoking and the presence of certain elements. In addition, chlorine proved to be important in the context of patient prognosis and survival.
Collapse
Affiliation(s)
- Anderson Barros Archanjo
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil.
| | | | - Mayara Mota de Oliveira
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil
| | - Suzanny Oliveira Mendes
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil
| | - Aline Ribeiro Borçoi
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil
| | - Lucas de Lima Maia
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil
| | | | - Rafael de Cicco
- Cancer Institute Arnaldo Vieira de Carvalho, São Paulo, Brazil
| | | | - Edna Teruko Kimura
- Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | | | | | - Eloiza H Tajara
- Medical School of São José do Rio Preto, São José do Rio Preto, Brazil
| | - Marcelo Dos Santos
- Multicampi School of Medical Sciences of Rio Grande do Norte, Federal University of Rio Grande do Norte, Caicó, Brazil
| | - Breno Valentim Nogueira
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil
| | | | | | - Adriana Madeira Álvares-da-Silva
- Postgraduate Program in Biotechnology/RENORBIO, Federal University of Espirito Santo, Av. Marechal Campos, 1468, Vitoria, 29.040-090, ES, Brazil
| |
Collapse
|
21
|
Suppression of CaMKIIβ Inhibits ANO1-Mediated Glioblastoma Progression. Cells 2020; 9:cells9051079. [PMID: 32357567 PMCID: PMC7290681 DOI: 10.3390/cells9051079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
ANO1, a Ca2+-activated chloride channel, is highly expressed in glioblastoma cells and its surface expression is involved in their migration and invasion. However, the regulation of ANO1 surface expression in glioblastoma cells is largely unknown. In this study, we found that Ca2+/Calmodulin-dependent protein kinase II (CaMKII) β specifically enhances the surface expression and channel activity of ANO1 in U251 glioblastoma cells. When KN-93, a CaMKII inhibitor, was used to treat U251 cells, the surface expression and channel activity of ANO1 were significantly reduced. Only CaMKIIβ, among the four CaMKII isoforms, increased the surface expression and channel activity of ANO1 in a heterologous expression system. Additionally, gene silencing of CaMKIIβ suppressed the surface expression and channel activity of ANO1 in U251 cells. Moreover, gene silencing of CaMKIIβ or ANO1 prominently reduced the migration and invasion of U251 and U87 MG glioblastoma cells. We thus conclude that CaMKIIβ plays a specific role in the surface expression of ANO1 and in the ANO1-mediated tumorigenic properties of glioblastoma cells, such as migration and invasion.
Collapse
|
22
|
Yool AJ, Ramesh S. Molecular Targets for Combined Therapeutic Strategies to Limit Glioblastoma Cell Migration and Invasion. Front Pharmacol 2020; 11:358. [PMID: 32292341 PMCID: PMC7118801 DOI: 10.3389/fphar.2020.00358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
The highly invasive nature of glioblastoma imposes poor prospects for patient survival. Molecular evidence indicates glioblastoma cells undergo an intriguing expansion of phenotypic properties to include neuron-like signaling using excitable membrane ion channels and synaptic proteins, augmenting survival and motility. Neurotransmitter receptors, membrane signaling, excitatory receptors, and Ca2+ responses are important candidates for the design of customized treatments for cancers within the heterogeneous central nervous system. Relatively few published studies of glioblastoma multiforme (GBM) have evaluated pharmacological agents targeted to signaling pathways in limiting cancer cell motility. Transcriptomic analyses here identified classes of ion channels, ionotropic receptors, and synaptic proteins that are enriched in human glioblastoma biopsy samples. The pattern of GBM-enriched gene expression points to a major role for glutamate signaling. However, the predominant role of AMPA receptors in fast excitatory signaling throughout the central nervous system raises a challenge on how to target inhibitors selectively to cancer cells while maintaining tolerability. This review critically evaluates a panel of ligand- and voltage-gated ion channels and synaptic proteins upregulated in GBM, and the evidence for their potential roles in the pathological disease progress. Evidence suggests combinations of therapies could be more effective than single agents alone. Natural plant products used in traditional medicines for the treatment of glioblastoma contain flavonoids, terpenoids, polyphenols, epigallocatechin gallate, quinones, and saponins, which might serendipitously include agents that modulate some classes of signaling compounds highlighted in this review. New therapeutic strategies are likely to exploit evidence-based combinations of selected agents, each at a low dose, to create new cancer cell-specific therapeutics.
Collapse
Affiliation(s)
- Andrea J. Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sunita Ramesh
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
23
|
Villalobo A, Berchtold MW. The Role of Calmodulin in Tumor Cell Migration, Invasiveness, and Metastasis. Int J Mol Sci 2020; 21:ijms21030765. [PMID: 31991573 PMCID: PMC7037201 DOI: 10.3390/ijms21030765] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/18/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Calmodulin (CaM) is the principal Ca2+ sensor protein in all eukaryotic cells, that upon binding to target proteins transduces signals encoded by global or subcellular-specific changes of Ca2+ concentration within the cell. The Ca2+/CaM complex as well as Ca2+-free CaM modulate the activity of a vast number of enzymes, channels, signaling, adaptor and structural proteins, and hence the functionality of implicated signaling pathways, which control multiple cellular functions. A basic and important cellular function controlled by CaM in various ways is cell motility. Here we discuss the role of CaM-dependent systems involved in cell migration, tumor cell invasiveness, and metastasis development. Emphasis is given to phosphorylation/dephosphorylation events catalyzed by myosin light-chain kinase, CaM-dependent kinase-II, as well as other CaM-dependent kinases, and the CaM-dependent phosphatase calcineurin. In addition, the role of the CaM-regulated small GTPases Rac1 and Cdc42 (cell division cycle protein 42) as well as CaM-binding adaptor/scaffold proteins such as Grb7 (growth factor receptor bound protein 7), IQGAP (IQ motif containing GTPase activating protein) and AKAP12 (A kinase anchoring protein 12) will be reviewed. CaM-regulated mechanisms in cancer cells responsible for their greater migratory capacity compared to non-malignant cells, invasion of adjacent normal tissues and their systemic dissemination will be discussed, including closely linked processes such as the epithelial–mesenchymal transition and the activation of metalloproteases. This review covers as well the role of CaM in establishing metastatic foci in distant organs. Finally, the use of CaM antagonists and other blocking techniques to downregulate CaM-dependent systems aimed at preventing cancer cell invasiveness and metastasis development will be outlined.
Collapse
Affiliation(s)
- Antonio Villalobo
- Cancer and Human Molecular Genetics Area—Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain
- Correspondence: (A.V.); (M.W.B.)
| | - Martin W. Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen, Denmark
- Correspondence: (A.V.); (M.W.B.)
| |
Collapse
|
24
|
Kang S, Kwon HN, Kang S, Park S. Interaction between IDH1 WT and calmodulin and its implications for glioblastoma cell growth and migration. Biochem Biophys Res Commun 2020; 524:224-230. [PMID: 31983428 DOI: 10.1016/j.bbrc.2020.01.073] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 01/02/2023]
Abstract
Isocitrate dehydrogenase (IDH) mutations are found in low-grade gliomas, and the product of the IDH mutant (MT), 2-hydroxyglutarate (2-HG), is the first known oncometabolite. However, the roles of the IDH wild type (WT) in high-grade glioblastoma, which rarely has the IDH mutation, are still unknown. To investigate possible pathways related to IDH WT in gliomas, we carried out bioinformatics analysis, and found that IDH1 has several putative calmodulin (CaM) binding sites. Pull-down and quantitative dissociation constant (Kd) measurements using recombinant proteins showed that IDH1 WT indeed binds to CaM with a higher affinity than IDH1 R132H MT. This biochemical interaction was demonstrated also in the cellular environment by immunoprecipitation with glioblastoma cell extracts. A synthetic peptide for the suggested binding region interfered with the interaction between CaM and IDH1, confirming the specificity of the binding. Direct binding between the synthetic peptide and CaM was observed in an NMR binding experiment, which additionally revealed that the peptide initially binds to the C-lobe of CaM. The physiological meaning of the CaM-IDH1 WT binding was shown with trifluoperazine (TFP), a CaM antagonist, which disrupted the binding and inhibited survival and migration of glioblastoma cells with IDH1 WT. As CaM signaling is activated in glioblastoma, our results suggest that IDH1 WT may be involved in the CaM-signaling pathway in the tumorigenesis of high-grade gliomas.
Collapse
Affiliation(s)
- Sunmi Kang
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| | - Hyuk Nam Kwon
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Stem Cells and Metabolism Research Program, Faculty of Medicine / Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Soeun Kang
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea; Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Sunghyouk Park
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
25
|
|
26
|
Abstract
As the leading cause of death in cancer, there is an urgent need to develop treatments to target the dissemination of primary tumor cells to secondary organs, known as metastasis. Bioelectric signaling has emerged in the last century as an important controller of cell growth, and with the development of current molecular tools we are now beginning to identify its role in driving cell migration and metastasis in a variety of cancer types. This review summarizes the currently available research for bioelectric signaling in solid tumor metastasis. We review the steps of metastasis and discuss how these can be controlled by bioelectric cues at the level of a cell, a population of cells, and the tissue. The role of ion channel, pump, and exchanger activity and ion flux is discussed, along with the importance of the membrane potential and the relationship between ion flux and membrane potential. We also provide an overview of the evidence for control of metastasis by external electric fields (EFs) and draw from examples in embryogenesis and regeneration to discuss the implications for endogenous EFs. By increasing our understanding of the dynamic properties of bioelectric signaling, we can develop new strategies that target metastasis to be translated into the clinic.
Collapse
Affiliation(s)
- Samantha L. Payne
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
27
|
Morishita K, Watanabe K, Ichijo H. Cell volume regulation in cancer cell migration driven by osmotic water flow. Cancer Sci 2019; 110:2337-2347. [PMID: 31120184 PMCID: PMC6676112 DOI: 10.1111/cas.14079] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer metastasis is the most frequent cause of death for patients with cancer. The main current treatment for cancer metastasis is chemotherapy targeting cancer cells’ ability to proliferate. However, some types of cancer cells show resistance to chemotherapy. Recently, cancer cell migration has become the subject of interest as a novel target of cancer therapy. Cell migration requires many factors, such as the cytoskeleton, cell‐matrix adhesion and cell volume regulation. Here, we focus on cell volume regulation and the role of ion/water transport systems in cell migration. Transport proteins, such as ion channels, ion carriers, and aquaporins, are indispensable for cell volume regulation under steady‐state conditions and during exposure to osmotic stress. Studies from the last ~25 years have revealed that cell volume regulation also plays an important role in the process of cell migration. Water flow in accordance with localized osmotic gradients generated by ion transport contributes to the driving force for cell migration. Moreover, it has been reported that metastatic cancer cells have higher expression of these transport proteins than nonmetastatic cancer cells. Thus, ion/water transport proteins involved in cell volume regulation and cell migration could be novel therapeutic targets for cancer metastasis. In this review, after presenting the importance of ion/water transport systems in cell volume regulation, we discuss the roles of transport proteins in a pathophysiological context, especially in the context of cancer cell migration.
Collapse
Affiliation(s)
- Kazuhiro Morishita
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kengo Watanabe
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Hvinden IC, Berg HE, Sachse D, Skaga E, Skottvoll FS, Lundanes E, Sandberg CJ, Vik-Mo EO, Rise F, Wilson SR. Nuclear Magnetic Resonance Spectroscopy to Identify Metabolite Biomarkers of Nonresponsiveness to Targeted Therapy in Glioblastoma Tumor Stem Cells. J Proteome Res 2019; 18:2012-2020. [PMID: 30964684 DOI: 10.1021/acs.jproteome.8b00801] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Glioblastoma is the most common and malignant brain tumor, and current therapies confer only modest survival benefits. A major obstacle is our ability to monitor treatment effect on tumors. Current imaging modalities are ambiguous, and repeated biopsies are not encouraged. To scout for markers of treatment response, we used NMR spectroscopy to study the effects of a survivin inhibitor on the metabolome of primary glioblastoma cancer stem cells. Applying high resolution NMR spectroscopy (1H resonance frequency: 800.03 MHz) to just 3 million cells per sample, we achieved sensitive and high resolving determinations of, e.g., amino acids, nucleosides, and constituents of the citric acid cycle. For control samples that were cultured, prepared, and measured at varying dates, peak area relative standard deviations were 15-20%. Analyses of unfractionated lysates were performed for straightforward compound identification with COLMAR and HMDB databases. Principal component analysis revealed that citrate levels were clearly upregulated in nonresponsive cells, while lactate levels substantially decreased following treatment for both responsive and nonresponsive cells. Hence, lactate and citrate may be potential markers of successful drug uptake and poor response to survivin inhibitors, respectively. Our metabolomics approach provided alternative biomarker candidates compared to spectrometry-based proteomics, underlining benefits of complementary methodologies. These initial findings make a foundation for exploring in vivo MR spectroscopy (MRS) of brain tumors, as citrate and lactate are MRS-visible. In sum, NMR metabolomics is a tool for addressing glioblastoma.
Collapse
Affiliation(s)
- Ingvild Comfort Hvinden
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway.,Department of Chemistry , Chemistry Research Laboratory, University of Oxford , 12 Mansfield Road , Oxford OX1 3TA , United Kingdom
| | - Henriette Engen Berg
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Daniel Sachse
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Erlend Skaga
- Vilhelm Magnus Laboratory of Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery , Oslo University Hospital , 4950 Nydalen NO-0424 , Oslo , Norway.,Institute of Clinical Medicine, Faculty of Medicine , University of Oslo , Post Box 1171, Blindern NO-0318 , Oslo , Norway
| | - Frøydis Sved Skottvoll
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway.,Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine , University of Oslo , PO Box 1112, Blindern NO-0317 , Oslo , Norway
| | - Elsa Lundanes
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Cecilie J Sandberg
- Vilhelm Magnus Laboratory of Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery , Oslo University Hospital , 4950 Nydalen NO-0424 , Oslo , Norway
| | - Einar O Vik-Mo
- Vilhelm Magnus Laboratory of Neurosurgical Research, Institute for Surgical Research and Department of Neurosurgery , Oslo University Hospital , 4950 Nydalen NO-0424 , Oslo , Norway
| | - Frode Rise
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway
| | - Steven Ray Wilson
- Department of Chemistry , University of Oslo , Post Box 1033, Blindern NO-0315 , Oslo , Norway.,Hybrid Technology Hub, Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine , University of Oslo , PO Box 1112, Blindern NO-0317 , Oslo , Norway
| |
Collapse
|
29
|
Cheng W, Zheng S, Li L, Zhou Q, Zhu H, Hu J, Luo H. Chloride channel 3 (CIC-3) predicts the tumor size in hepatocarcinoma. Acta Histochem 2019; 121:284-288. [PMID: 30678806 DOI: 10.1016/j.acthis.2019.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 02/02/2023]
Abstract
Chloride channel 3 (CIC-3) has been suggested to be implicated in the carcinogenesis though; it still remains ill understood in hepatocarcinoma, especially in terms of clinicopathological meaning of its expression. Given this, herein, to understand the clinicopathological significance of CIC-3 expression in hepatocarcinoma, Immunohistochemistry was performed to examine the level of CIC-3, followed by statistical analysis of the correlation between expression versus clinicopathological variables, including gender, age, TNM classifications, tumor size, lymph node metastasis and overall prognosis. It was shown that positive staining of CIC-3 can be present in both hepatocarcinoma and its paired normal controls; and that CIC-3 was significantly over-expressed in hepatcarcioma on the whole relative to paired normal controls. Moreover, up-regulation of CIC-3 markedly correlated with tumor size and overall prognosis, suggesting that CIC-3 expression could predict both tumor size and overall prognosis in hepatocarcinoma.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Hematologic and Oncology, The Center Hospital of Karamay City, Karamay, 834000, Xinjiang, China
| | - Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, 830011, China; Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, 830011, China
| | - Li Li
- Department of Hematologic and Oncology, The Center Hospital of Karamay City, Karamay, 834000, Xinjiang, China
| | - Qin Zhou
- Department of Hematologic and Oncology, The Center Hospital of Karamay City, Karamay, 834000, Xinjiang, China
| | - Haipeng Zhu
- Department of Hematologic and Oncology, The Center Hospital of Karamay City, Karamay, 834000, Xinjiang, China
| | - Jun Hu
- Department of Hematologic and Oncology, The Center Hospital of Karamay City, Karamay, 834000, Xinjiang, China
| | - Hongbin Luo
- Department of Traditional Chinese Medicine, The Center Hospital of Karamay City, Karamay, 834000, Xinjiang, China.
| |
Collapse
|
30
|
Cenciarini M, Valentino M, Belia S, Sforna L, Rosa P, Ronchetti S, D'Adamo MC, Pessia M. Dexamethasone in Glioblastoma Multiforme Therapy: Mechanisms and Controversies. Front Mol Neurosci 2019; 12:65. [PMID: 30983966 PMCID: PMC6449729 DOI: 10.3389/fnmol.2019.00065] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/26/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant of the glial tumors. The world-wide estimates of new cases and deaths annually are remarkable, making GBM a crucial public health issue. Despite the combination of radical surgery, radio and chemotherapy prognosis is extremely poor (median survival is approximately 1 year). Thus, current therapeutic interventions are highly unsatisfactory. For many years, GBM-induced brain oedema and inflammation have been widely treated with dexamethasone (DEX), a synthetic glucocorticoid (GC). A number of studies have reported that DEX also inhibits GBM cell proliferation and migration. Nevertheless, recent controversial results provided by different laboratories have challenged the widely accepted dogma concerning DEX therapy for GBM. Here, we have reviewed the main clinical features and genetic and epigenetic abnormalities underlying GBM. Finally, we analyzed current notions and concerns related to DEX effects on cerebral oedema, cancer cell proliferation and migration and clinical outcome.
Collapse
Affiliation(s)
- Marta Cenciarini
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Simona Ronchetti
- Section of Pharmacology, Department of Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Mauro Pessia
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy.,Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| |
Collapse
|
31
|
Guan YT, Xie Y, Zhou H, Shi HY, Zhu YY, Zhang XL, Luan Y, Shen XM, Chen YP, Xu LJ, Lin ZQ, Wang G. Overexpression of chloride channel-3 (ClC-3) is associated with human cervical carcinoma development and prognosis. Cancer Cell Int 2019; 19:8. [PMID: 30636929 PMCID: PMC6325671 DOI: 10.1186/s12935-018-0721-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/24/2018] [Indexed: 02/06/2023] Open
Abstract
Background Cervical carcinoma is a major gynecological cancer and causes cancer-related deaths in worldwide, the latent pathogenesis and progress of cervical cancer is still under research. ClC-3 may be an important promoter for aggressive metastasis of malignant tumors. In this research, we explore the ClC-3 expression in cervical carcinoma and its underlying clinical significance, trying to illuminate ClC-3 probable function in the neoplasm malignant behavior, development and prognosis. Methods Paraffin-embedded cervical (n = 168) and lymph node (n = 100) tissue specimens were analysed by immunohistochemistry. Fresh human cervical tissue specimens (n = 165) and four human cervical cell lines were tested for ClC-3 mRNA and protein expression levels by quantitative real-time PCR and western blotting. The relationship between the expression levels of ClC-3, the pathological characteristics of the carcinoma, and the clinical prognosis were statistically analysed. Results In normal and precancerous (LSIL, HSIL) cervical tissues as well as cervical carcinoma tissues, both ClC-3 mRNA and protein expression levels increased significantly (p < 0.05). The expression level of ClC-3 was closely-related to the histological differentiation (p = 0.029), tumour staging (p = 0.016), tumour size (p = 0.039), vascular invasion (p = 0.045), interstitial infiltration depth (p = 0.012), lymphatic metastasis (p = 0.036), and HPV infection (p = 0.022). In an in vitro experiment, ClC-3 mRNA and protein were found to be overexpressed both in the HeLa and SiHa cell lines, but low expression levels were detected in the C-33A and H8 cell lines (p < 0.05). Furthermore, the high expression levels of ClC-3 was significantly correlated to poor survival in cervical carcinoma patients (Log-rank test, p = 0.046). Conclusions These data suggest that overexpression of ClC-3 is closely associated with human cervical carcinoma progression and poor prognosis; this suggests that ClC-3 may function as a patent tumour biomarker and a latent therapeutic target for cervical carcinoma patients.
Collapse
Affiliation(s)
- Yu-Tao Guan
- 1Department of Obstetrics and Gynaecology, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China.,2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| | - Yong Xie
- 2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| | - Hui Zhou
- 1Department of Obstetrics and Gynaecology, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hai-Yan Shi
- 3Department of Pathology, The First People's Hospital of Foshan, Foshan, China
| | - Yu-Yuan Zhu
- 2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| | - Xiao-Lu Zhang
- 2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| | - Yi Luan
- 1Department of Obstetrics and Gynaecology, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xi-Ming Shen
- 4Department of Pathology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yang-Ping Chen
- 2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| | - Li-Jiang Xu
- 2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| | - Zhong-Qiu Lin
- 1Department of Obstetrics and Gynaecology, Sun Yat-sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Gang Wang
- 2Department of Obstetrics and Gynaecology, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
32
|
Brzozowski JS, Skelding KA. The Multi-Functional Calcium/Calmodulin Stimulated Protein Kinase (CaMK) Family: Emerging Targets for Anti-Cancer Therapeutic Intervention. Pharmaceuticals (Basel) 2019; 12:ph12010008. [PMID: 30621060 PMCID: PMC6469190 DOI: 10.3390/ph12010008] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 01/25/2023] Open
Abstract
The importance of Ca2+ signalling in key events of cancer cell function and tumour progression, such as proliferation, migration, invasion and survival, has recently begun to be appreciated. Many cellular Ca2+-stimulated signalling cascades utilise the intermediate, calmodulin (CaM). The Ca2+/CaM complex binds and activates a variety of enzymes, including members of the multifunctional Ca2+/calmodulin-stimulated protein kinase (CaMK) family. These enzymes control a broad range of cancer-related functions in a multitude of tumour types. Herein, we explore the cancer-related functions of these kinases and discuss their potential as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joshua S Brzozowski
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Kathryn A Skelding
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute (HMRI) and University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
33
|
Prevarskaya N, Skryma R, Shuba Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol Rev 2018; 98:559-621. [PMID: 29412049 DOI: 10.1152/physrev.00044.2016] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Genomic instability is a primary cause and fundamental feature of human cancer. However, all cancer cell genotypes generally translate into several common pathophysiological features, often referred to as cancer hallmarks. Although nowadays the catalog of cancer hallmarks is quite broad, the most common and obvious of them are 1) uncontrolled proliferation, 2) resistance to programmed cell death (apoptosis), 3) tissue invasion and metastasis, and 4) sustained angiogenesis. Among the genes affected by cancer, those encoding ion channels are present. Membrane proteins responsible for signaling within cell and among cells, for coupling of extracellular events with intracellular responses, and for maintaining intracellular ionic homeostasis ion channels contribute to various extents to pathophysiological features of each cancer hallmark. Moreover, tight association of these hallmarks with ion channel dysfunction gives a good reason to classify them as special type of channelopathies, namely oncochannelopathies. Although the relation of cancer hallmarks to ion channel dysfunction differs from classical definition of channelopathies, as disease states causally linked with inherited mutations of ion channel genes that alter channel's biophysical properties, in a broader context of the disease state, to which pathogenesis ion channels essentially contribute, such classification seems absolutely appropriate. In this review the authors provide arguments to substantiate such point of view.
Collapse
Affiliation(s)
- Natalia Prevarskaya
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Roman Skryma
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| | - Yaroslav Shuba
- INSERM U-1003, Equipe Labellisée par la Ligue Nationale contre le Cancer et LABEX, Université Lille1 , Villeneuve d'Ascq , France ; Bogomoletz Institute of Physiology and International Center of Molecular Physiology, NASU, Kyiv-24, Ukraine
| |
Collapse
|
34
|
Jentsch TJ, Pusch M. CLC Chloride Channels and Transporters: Structure, Function, Physiology, and Disease. Physiol Rev 2018; 98:1493-1590. [DOI: 10.1152/physrev.00047.2017] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
CLC anion transporters are found in all phyla and form a gene family of eight members in mammals. Two CLC proteins, each of which completely contains an ion translocation parthway, assemble to homo- or heteromeric dimers that sometimes require accessory β-subunits for function. CLC proteins come in two flavors: anion channels and anion/proton exchangers. Structures of these two CLC protein classes are surprisingly similar. Extensive structure-function analysis identified residues involved in ion permeation, anion-proton coupling and gating and led to attractive biophysical models. In mammals, ClC-1, -2, -Ka/-Kb are plasma membrane Cl−channels, whereas ClC-3 through ClC-7 are 2Cl−/H+-exchangers in endolysosomal membranes. Biological roles of CLCs were mostly studied in mammals, but also in plants and model organisms like yeast and Caenorhabditis elegans. CLC Cl−channels have roles in the control of electrical excitability, extra- and intracellular ion homeostasis, and transepithelial transport, whereas anion/proton exchangers influence vesicular ion composition and impinge on endocytosis and lysosomal function. The surprisingly diverse roles of CLCs are highlighted by human and mouse disorders elicited by mutations in their genes. These pathologies include neurodegeneration, leukodystrophy, mental retardation, deafness, blindness, myotonia, hyperaldosteronism, renal salt loss, proteinuria, kidney stones, male infertility, and osteopetrosis. In this review, emphasis is laid on biophysical structure-function analysis and on the cell biological and organismal roles of mammalian CLCs and their role in disease.
Collapse
Affiliation(s)
- Thomas J. Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; and Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy
| | - Michael Pusch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP) and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany; and Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy
| |
Collapse
|
35
|
Zhou FM, Huang YY, Tian T, Li XY, Tang YB. Knockdown of Chloride Channel-3 Inhibits Breast Cancer Growth In Vitro and In Vivo. J Breast Cancer 2018; 21:103-111. [PMID: 29963105 PMCID: PMC6015970 DOI: 10.4048/jbc.2018.21.2.103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 05/04/2018] [Indexed: 12/11/2022] Open
Abstract
Purpose Chloride channel-3 (ClC-3) is a member of the chloride channel family and plays a critical role in a variety of cellular activities. The aim of the present study is to explore the molecular mechanisms underlying the antitumor effect of silencing ClC-3 in breast cancer. Methods Human breast cancer cell lines MDA-MB-231 and MCF-7 were used in the experiments. Messenger RNA and protein expression were examined by quantitative real-time polymerase chain reaction and western blot analysis. Cell proliferation was measured by the bromodeoxyuridine method, and the cell cycle was evaluated using fluorescence-activated cell sorting. Protein interaction in cells was analyzed by co-immunoprecipitation. Tumor tissues were stained with hematoxylin-eosin and tumor burden was measured using the Metamorph software. Results Breast cancer tissues collected from patients showed an increase in ClC-3 expression. Knockdown of ClC-3 inhibited the secretion of insulin-like growth factor (IGF)-1, cell proliferation, and G1/S transition in breast cancer cells. In the mouse xenograft model of human breast carcinoma, tumor growth was significantly slower in animals injected with ClC-3-deficient cells compared with the growth of normal human breast cancer cells. In addition, silencing of ClC-3 attenuated the expression of proliferating cell nuclear antigen, Ki-67, cyclin D1, and cyclin E, as well as the activation of extracellular signalregulated protein kinases (ERK) 1/2, both in vitro and in vivo. Conclusion Together, our data suggest that upregulation of ClC-3 by IGF-1 contributes to cell proliferation and tumor growth in breast cancer, and ClC-3 deficiency suppresses cell proliferation and tumor growth via the IGF/IGF receptor/ERK pathway.
Collapse
Affiliation(s)
- Fang-Min Zhou
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yun-Ying Huang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacy, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Tian
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Yan Li
- Department of Clinical Pharmacology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong-Bo Tang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
Qi Y, Mair N, Kummer KK, Leitner MG, Camprubí-Robles M, Langeslag M, Kress M. Identification of Chloride Channels CLCN3 and CLCN5 Mediating the Excitatory Cl - Currents Activated by Sphingosine-1-Phosphate in Sensory Neurons. Front Mol Neurosci 2018; 11:33. [PMID: 29479306 PMCID: PMC5811518 DOI: 10.3389/fnmol.2018.00033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/24/2018] [Indexed: 12/27/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid involved in numerous physiological and pathophysiological processes. We have previously reported a S1P-induced nocifensive response in mice by excitation of sensory neurons via activation of an excitatory chloride current. The underlying molecular mechanism for the S1P-induced chloride conductance remains elusive. In the present study, we identified two CLCN voltage-gated chloride channels, CLCN3 and CLCN5, which mediated a S1P-induced excitatory Cl- current in sensory neurons by combining RNA-seq, adenovirus-based gene silencing and whole-cell electrophysiological voltage-clamp recordings. Downregulation of CLCN3 and CLCN5 channels by adenovirus-mediated delivery of shRNA dramatically reduced S1P-induced Cl- current and membrane depolarization in sensory neurons. The mechanism of S1P-induced activation of the chloride current involved Rho GTPase but not Rho-associated protein kinase. Although S1P-induced potentiation of TRPV1-mediated ionic currents also involved Rho-dependent process, the lack of correlation of the S1P-activated Cl- current and the potentiation of TRPV1 by S1P suggests that CLCN3 and CLCN5 are necessary components for S1P-induced excitatory Cl- currents but not for the amplification of TRPV1-mediated currents in sensory neurons. This study provides a novel mechanistic insight into the importance of bioactive sphingolipids in nociception.
Collapse
Affiliation(s)
- Yanmei Qi
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Norbert Mair
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai K Kummer
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael G Leitner
- Department of Neurophysiology, Philipps University of Marburg, Marburg, Germany
| | - María Camprubí-Robles
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Michiel Langeslag
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Division of Physiology, Department of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
37
|
Umans RA, Sontheimer H. Combating malignant astrocytes: Strategies mitigating tumor invasion. Neurosci Res 2018; 126:22-30. [PMID: 29054465 PMCID: PMC6880651 DOI: 10.1016/j.neures.2017.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 08/30/2017] [Accepted: 08/30/2017] [Indexed: 01/08/2023]
Abstract
Malignant gliomas are glial-derived, primary brain tumors that carry poor prognosis. Existing therapeutics are largely ineffective and dramatically affect quality of life. The standard of care details a taxing combination of surgical resection, radiation of the resection cavity, and temozolomide (TMZ) chemotherapy, with treatment extending life by only an average of months (Maher et al., 2001; Stupp et al., 2005). Despite scientific and technological advancement, surgery remains the most important treatment modality. Therapeutic obstacles include xenobiotic protection conveyed by the blood-brain barrier (Zhang et al., 2015), invasiveness and therapeutic resistance of tumor cell populations (Bao et al., 2006), and distinctive attributes of secondary glioma occurrence (Ohgaki and Kleihues, 2013). While these brain malignancies can be classified by grade or grouped by molecular subclass, each tumor presents itself as its own complication. Based on all of these obstacles, new therapeutic approaches are urgently needed. These will likely emerge from numerous exciting studies of glioma biology that are ongoing and reviewed here. These show unexpected roles for ion channels, amino-acid transporters, and connexin gap junctions in supporting the invasive growth of gliomas. These studies have identified a number of proteins that may be targeted for therapy in the future.
Collapse
Affiliation(s)
- Robyn A Umans
- Center for Glial Biology in Health and Disease, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA
| | - Harald Sontheimer
- Center for Glial Biology in Health and Disease, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA.
| |
Collapse
|
38
|
Chen TJ, He HL, Shiue YL, Yang CC, Lin LC, Tian YF, Chen SH. High chloride channel accessory 1 expression predicts poor prognoses in patients with rectal cancer receiving chemoradiotherapy. Int J Med Sci 2018; 15:1171-1178. [PMID: 30123054 PMCID: PMC6097263 DOI: 10.7150/ijms.26685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 06/30/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Concurrent chemoradiotherapy (CCRT) has now become the standard of treatments for advanced rectal cancer before surgery. To search the biological molecules with prognostic and therapeutic potential of CCRT could be beneficial for these patients. Recently, aberrant expression of chloride channels has been linked to radio-resistance in glioblastoma; however, its clinical implication has not been well-studied in rectal cancers. Therefore, we examined the clinical significance of targetable drivers associated with chloride channel activity in patients with rectal cancer receiving CCRT. Methods: After datamining from a published transcriptome of rectal cancers, upregulation of CLCA1 gene was recognized to be significantly correlated with non-responders of CCRT. In validation cohort of rectal cancers, the expression levels of CLCA1 were accessed by using immunohistochemistry assays in 172 tumor specimens that were obtained before any treatment. Expression levels of CLCA1 were statistically analyzed with principal clinicopathological features and survival outcomes in this substantial cohort. Results: In validation cohort, high expression of CLCA1 was significantly associated with higher pre-treatment tumor nodal stages (P=0.032), vascular invasion (P=0.028), and inferior tumor regression grade (P=0.042). In survival evaluations, high expression of CLCA1 was significantly correlated with worse local recurrence-free survival (LRFS; P=0.0012), metastasis-free survival (MeFS; P =0.0114), and disease-specific survival (DSS; P=0.0041). Furthermore, high expression of CLCA1 remained an independent prognosticator of shorter LRFS (P=0.029, hazard ratio=2.555), MeFS (P=0.044, hazard ratio=2.125) and DSS (P=0.044, hazard ratio=2.172). Conclusions: High expression of CLCA1 is significantly associated with poor therapeutic response and survival outcomes in rectal cancer patients with CCRT treatment before surgery. With the development of specific inhibitors, our findings indicate not only prognostic but also therapeutic potential of CLCA1 in rectal cancers.
Collapse
Affiliation(s)
- Tzu-Ju Chen
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Optometry, Chung Hwa University of Medical Technology, Tainan, Taiwan.,Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hong-Lin He
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Tainan, Taiwan.,Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Yu-Feng Tian
- Division of General Surgery, Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan.,Department of Health & Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Shang-Hung Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
39
|
TRPM8 is required for survival and radioresistance of glioblastoma cells. Oncotarget 2017; 8:95896-95913. [PMID: 29221175 PMCID: PMC5707069 DOI: 10.18632/oncotarget.21436] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/25/2017] [Indexed: 01/29/2023] Open
Abstract
TRPM8 is a Ca2+-permeable nonselective cation channel belonging to the melastatin sub-group of the transient receptor potential (TRP) family. TRPM8 is aberrantly overexpressed in a variety of tumor entities including glioblastoma multiforme where it reportedly contributes to tumor invasion. The present study aimed to disclose further functions of TRPM8 in glioma biology in particular upon cell injury by ionizing radiation. To this end, TCGA data base was queried to expose the TRPM8 mRNA abundance in human glioblastoma specimens and immunoblotting was performed to analyze the TRPM8 protein abundance in primary cultures of human glioblastoma. Moreover, human glioblastoma cell lines were irradiated with 6 MV photons and TRPM8 channels were targeted pharmacologically or by RNA interference. TRPM8 abundance, Ca2+ signaling and resulting K+ channel activity, chemotaxis, cell migration, clonogenic survival, DNA repair, apoptotic cell death, and cell cycle control were determined by qRT-PCR, fura-2 Ca2+ imaging, patch-clamp recording, transfilter migration assay, wound healing assay, colony formation assay, immunohistology, flow cytometry, and immunoblotting. As a result, human glioblastoma upregulates TRPM8 channels to variable extent. TRPM8 inhibition or knockdown slowed down cell migration and chemotaxis, attenuated DNA repair and clonogenic survival, triggered apoptotic cell death, impaired cell cycle and radiosensitized glioblastoma cells. Mechanistically, ionizing radiation activated and upregulated TRPM8-mediated Ca2+ signaling that interfered with cell cycle control probably via CaMKII, cdc25C and cdc2. Combined, our data suggest that TRPM8 channels contribute to spreading, survival and radioresistance of human glioblastoma and, therefore, might represent a promising target in future anti-glioblastoma therapy.
Collapse
|
40
|
Wang B, Xie J, He HY, Huang EW, Cao QH, Luo L, Liao YS, Guo Y. Suppression of CLC-3 chloride channel reduces the aggressiveness of glioma through inhibiting nuclear factor-κB pathway. Oncotarget 2017; 8:63788-63798. [PMID: 28969029 PMCID: PMC5609961 DOI: 10.18632/oncotarget.19093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/06/2017] [Indexed: 11/25/2022] Open
Abstract
CLC-3 chloride channel plays important roles on cell volume regulation, proliferation and migration in normal and cancer cells. Recent growing evidence supports a critical role of CLC-3 in glioma metastasis, however, the mechanism underlying is unclear. This study finds that CLC-3 is upregulated in glioma tissues and positively correlated with WHO histological grade. Patients with high CLC-3 expression had an overall shorter survival time, whereas patients with low expression of CLC-3 had a better survival time. Silencing endogenous CLC-3 with ShCLC-3 adenovirus significantly decreases volume-regulated chloride currents, inhibits the nuclear translocation of p65 subunit of Nuclear Factor-κB (NF-κB), decreases transcriptional activity of NF-κB, reduces MMP-3 and MMP-9 expression and decreases glioma cell migration and invasion. Taken together, these results suggest CLC-3 promotes the aggressiveness of glioma at least in part through nuclear factor-κB pathway, and might be a novel prognostic biomarker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Bing Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Department of Neurosurgery, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Jing Xie
- Department of Integrative Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Hai-Yong He
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - En-Wen Huang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qing-Hua Cao
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lun Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yong-Shi Liao
- Department of Neurosurgery, The Second Affiliated Hospital, University of South China, Hengyang, China
| | - Ying Guo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
41
|
Nguyen CH, Huttary N, Atanasov AG, Chatuphonprasert W, Brenner S, Fristiohady A, Hong J, Stadler S, Holzner S, Milovanovic D, Dirsch VM, Kopp B, Saiko P, Krenn L, Jäger W, Krupitza G. Fenofibrate inhibits tumour intravasation by several independent mechanisms in a 3-dimensional co-culture model. Int J Oncol 2017; 50:1879-1888. [PMID: 28393180 DOI: 10.3892/ijo.2017.3956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 01/16/2017] [Indexed: 11/06/2022] Open
Abstract
Lymph node metastasis of breast cancer is a clinical marker of poor prognosis. Yet, there exist no therapies targeting mechanisms of intravasation into lymphatics. Herein we report on an effect of the antidyslipidemic drug fenofibrate with vasoprotective activity, which attenuates breast cancer intravasation in vitro, and describe the potential mechanisms. To measure intravasation in a 3-dimensional co-culture model MDA-MB231 and MCF-7 breast cancer spheroids were placed on immortalised lymphendothelial cell (LEC) monolayers. This provokes the formation of circular chemorepellent induced defects (CCIDs) in the LEC barrier resembling entry ports for the intravasating tumour. Furthermore, the expression of adhesion molecules ICAM-1, CD31 and FAK was investigated in LECs by western blotting as well as cell-cell adhesion and NF-κB activity by respective assays. In MDA-MB231 cells the activity of CYP1A1 was measured by EROD assay. Fenofibrate inhibited CCID formation in the MDA-MB231/LEC- and MCF-7/LEC models and the activity of NF-κB, which in turn downregulated ICAM-1 in LECs and the adhesion of cancer cells to LECs. Furthermore, CD31 and the activity of FAK were inhibited. In MDA-MB231 cells, fenofibrate attenuated CYP1A1 activity. Combinations with other FDA-approved drugs, which reportedly inhibit different ion channels, attenuated CCID formation additively or synergistically. In summary, fenofibrate inhibited NF-κB and ICAM-1, and inactivated FAK, thereby attenuating tumour intravasation in vitro. A combination with other FDA-approved drugs further improved this effect. Our new concept may lead to a novel therapy for cancer patients.
Collapse
Affiliation(s)
- Chi Huu Nguyen
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Nicole Huttary
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | | | | | - Stefan Brenner
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Adryan Fristiohady
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Junli Hong
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Serena Stadler
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Silvio Holzner
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Daniela Milovanovic
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Brigitte Kopp
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Philipp Saiko
- Department of Medical and Chemical Laboratory Diagnostics, Medical University of Vienna, Vienna, Austria
| | - Liselotte Krenn
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Georg Krupitza
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Tavalin SJ, Colbran RJ. CaMKII-mediated phosphorylation of GluN2B regulates recombinant NMDA receptor currents in a chloride-dependent manner. Mol Cell Neurosci 2016; 79:45-52. [PMID: 27998718 DOI: 10.1016/j.mcn.2016.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 11/29/2016] [Accepted: 12/15/2016] [Indexed: 01/09/2023] Open
Abstract
Some forms of long-term synaptic plasticity require docking of Ca2+/calmodulin-dependent protein kinase II α (CaMKIIα) to residues 1290-1309 within the intracellular C-terminal tail of the N-methyl-d-aspartate (NMDA) receptor GluN2B subunit. The phosphorylation of Ser1303 within this region destabilizes CaMKII binding. Interestingly, Ser1303 is a substrate for CaMKII itself, as well as PKC and DAPK1, but these kinases have been reported to have contradictory effects on the activity of GluN2B-containing NMDA receptors. Here, we re-assessed the effect of CaMKII on NMDA receptor desensitization in heterologous cells, as measured by the ratio of steady-state to peak currents induced during 3s agonist applications. CaMKIIα co-expression or infusion of constitutively active CaMKII limits the extent of desensitization and preserves current amplitude with repeated stimulation of recombinant GluN1A/GluN2B when examined using low intracellular chloride (Cl-) levels, characteristic of neurons beyond the first postnatal week. In contrast, CaMKIIα enhances the acute rate and extent of desensitization when intracellular Cl- concentrations are high. The apparent dependence of CaMKIIα effects on NMDA receptor desensitization on Cl- concentrations is consistent with the presence of a Ca2+-activated Cl- conductance endogenous to HEK 293 cells, which was confirmed by photolysis of caged-Ca2+. However, Ca2+-activated Cl- conductances are unaffected by CaMKIIα expression, indicating that CaMKII affects agonist-induced whole cell currents via modulation of the NMDA receptor. In support of this idea, CaMKIIα modulation of GluN2B-NMDA receptors is abrogated by the phospho-null mutation of Ser1303 in GluN2B to alanine and occluded by phospho-mimetic mutation of Ser1303 to aspartate regardless of intracellular Cl- concentration. Thus, CaMKII-mediated phosphorylation of GluN2B-containing NMDA receptors reduces desensitization at physiological (low) intracellular Cl-, perhaps serving as a feed-forward mechanism to sustain NMDA-mediated Ca2+ entry and continued CaMKII activation during learning and memory.
Collapse
Affiliation(s)
- Steven J Tavalin
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38103, United States.
| | - Roger J Colbran
- Department of Molecular Physiology & Biophysics, Brain Institute, and Kennedy Center for Research on Human Development, Vanderbilt University School of Medicine, Nashville, TN, 37232-0615, United States
| |
Collapse
|
43
|
Sforna L, Cenciarini M, Belia S, Michelucci A, Pessia M, Franciolini F, Catacuzzeno L. Hypoxia Modulates the Swelling-Activated Cl Current in Human Glioblastoma Cells: Role in Volume Regulation and Cell Survival. J Cell Physiol 2016; 232:91-100. [PMID: 27028592 DOI: 10.1002/jcp.25393] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/25/2016] [Indexed: 12/18/2022]
Abstract
The malignancy of glioblastoma multiforme (GBM), the most common human brain tumor, correlates with the presence of hypoxic areas, but the underlying mechanisms are unclear. GBM cells express abundant Cl channels whose activity supports cell volume and membrane potential changes, ultimately leading to cell proliferation, migration, and escaping death. In non-tumor tissues Cl channels are modulated by hypoxia, which prompted us to verify whether hypoxia would also modulate Cl channels in GBM cells. Our results show that in GBM cell lines, acute application of a hypoxic solution activates a Cl current displaying the biophysical and pharmacological features of the swelling-activated Cl current (ICl,swell ). We also found that acute hypoxia increased the cell volume by about 20%, and a 30% hypertonic solution partially inhibited the hypoxia-activated Cl current, suggesting that cell swelling and the activation of the Cl current are sequential events. Notably, the hypoxia-induced cell swelling was followed by a regulatory volume decrease (RVD) mediated mainly by ICl,swell . Since, a hypoxia-induced prolonged cell swelling is usually regarded as a death insult, we hypothesized that the hypoxia-activated Cl current could limit cell swelling and prevent necrotic death of GBM cells under hypoxic conditions. In accordance, we found that the ICl,swell inhibitor DCPIB hampered the RVD process, and more importantly it sensibly increased the hypoxia-induced necrotic death in these cells. Taken together, these results suggest that Cl channels are strongly involved in the survival of GBM cells in a hypoxic environment, and may thus represent a new therapeutic target for this malignant tumor. J. Cell. Physiol. 232: 91-100, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.,Department of Experimental Medicine, University of Perugia, Italy
| | - Marta Cenciarini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy
| | - Antonio Michelucci
- Department of Neuroscience, Imaging and Clinical Sciences, University of Chieti 'G. d'Annunzio', Italy
| | - Mauro Pessia
- Department of Experimental Medicine, University of Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Italy.
| |
Collapse
|
44
|
Thompson EG, Sontheimer H. A role for ion channels in perivascular glioma invasion. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:635-648. [PMID: 27424110 DOI: 10.1007/s00249-016-1154-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 06/21/2016] [Accepted: 07/01/2016] [Indexed: 11/28/2022]
Abstract
Malignant gliomas are devastating tumors, frequently killing those diagnosed in little over a year. The profuse infiltration of glioma cells into healthy tissue surrounding the main tumor mass is one of the major obstacles limiting the improvement of patient survival. Migration along the abluminal side of blood vessels is one of the salient features of glioma cell invasion. Invading glioma cells are attracted to the vascular network, in part by the neuropeptide bradykinin, where glioma cells actively modify the gliovascular interface and undergo volumetric alterations to navigate the confined space. Critical to these volume modifications is a proposed hydrodynamic model that involves the flux of ions in and out of the cell, followed by osmotically obligated water. Ion and water channels expressed by the glioma cell are essential in this model of invasion and make opportune therapeutic targets. Lastly, there is growing evidence that vascular-associated glioma cells are able to control the vascular tone, presumably to free up space for invasion and growth. The unique mechanisms that enable perivascular glioma invasion may offer critical targets for therapeutic intervention in this devastating disease. Indeed, a chloride channel-blocking peptide has already been successfully tested in human clinical trials.
Collapse
Affiliation(s)
- Emily G Thompson
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, Roanoke, VA, USA
| | - Harald Sontheimer
- Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, Roanoke, VA, USA. .,Virginia Tech School of Neuroscience, Blacksburg, VA, USA.
| |
Collapse
|
45
|
Boursi B, Han HJ, Haynes K, Mamtani R, Yang YX. Ion channel blockers and glioblastoma risk and outcome: a nested case-control and retrospective cohort studies. Pharmacoepidemiol Drug Saf 2016; 25:1179-1185. [PMID: 27384764 DOI: 10.1002/pds.4054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/16/2016] [Accepted: 05/30/2016] [Indexed: 11/08/2022]
Abstract
PURPOSE Mutations in ion channels are common among patients with glioblastoma multiforme (GBM) and promote cell migration and invasion. We sought to evaluate the association between the use of specific ion channel blockers such as digoxin, amiodarone, diltiazem and verapamil and GBM risk and survival. METHODS We conducted a nested case-control study in a large primary care database from the UK. Cases were defined as all individuals with incident diagnosis of GBM during follow-up. For each case, up to four controls were selected using incidence density sampling. The primary exposure of interest was active treatment with each of the four ion channel blockers. We used conditional logistic regression to estimate odds ratios and 95% confidence interval (CI) for the association between ion channel blocker use and GBM risk. We then performed a Cox regression analysis among those diagnosed with GBM in order to evaluate the association between use of ion channel blockers and overall survival. Both analyses were adjusted to common confounders. RESULTS The study included 1076 cases and 4253 matched controls. There was no statistically significant difference between cases and controls in cardiac and metabolic risk factors. There was no change in GBM risk in active users of ion channel blockers compared with non-users. Among patients with GBM, active users of amiodarone had worse survival compared with never users with an HR of 4.41 (95%CI 1.95-9.96). There was no statistically significant change in survival among diltiazem, verapamil or digoxin users. CONCLUSION Treatment with specific ion channel blockers was not associated with the risk of GBM but was associated with worse survival in patients with GBM. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ben Boursi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.,Tel-Aviv University, Tel-Aviv, Israel
| | - Harry J Han
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin Haynes
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ronac Mamtani
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Yu-Xiao Yang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
46
|
Stegen B, Klumpp L, Misovic M, Edalat L, Eckert M, Klumpp D, Ruth P, Huber SM. K + channel signaling in irradiated tumor cells. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2016; 45:585-598. [PMID: 27165704 DOI: 10.1007/s00249-016-1136-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/24/2016] [Accepted: 04/20/2016] [Indexed: 12/17/2022]
Abstract
K+ channels crosstalk with biochemical signaling cascades and regulate virtually all cellular processes by adjusting the intracellular K+ concentration, generating the membrane potential, mediating cell volume changes, contributing to Ca2+ signaling, and directly interacting within molecular complexes with membrane receptors and downstream effectors. Tumor cells exhibit aberrant expression and activity patterns of K+ channels. The upregulation of highly "oncogenic" K+ channels such as the Ca2+-activated IK channel may drive the neoplastic transformation, malignant progression, metastasis, or therapy resistance of tumor cells. In particular, ionizing radiation in doses used for fractionated radiotherapy in the clinic has been shown to activate K+ channels. Radiogenic K+ channel activity, in turn, contributes to the DNA damage response and promotes survival of the irradiated tumor cells. Tumor-specific overexpression of certain K+ channel types together with the fact that pharmacological K+ channel modulators are already in clinical use or well tolerated in clinical trials suggests that K+ channel targeting alone or in combination with radiotherapy might become a promising new strategy of anti-cancer therapy. The present article aims to review our current knowledge on K+ channel signaling in irradiated tumor cells. Moreover, it provides new data on molecular mechanisms of radiogenic K+ channel activation and downstream signaling events.
Collapse
Affiliation(s)
- Benjamin Stegen
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Lukas Klumpp
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Milan Misovic
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Lena Edalat
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Marita Eckert
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Dominik Klumpp
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
47
|
Wang YY, Zhao R, Zhe H. The emerging role of CaMKII in cancer. Oncotarget 2016; 6:11725-34. [PMID: 25961153 PMCID: PMC4494900 DOI: 10.18632/oncotarget.3955] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 04/08/2015] [Indexed: 12/13/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a multifunctional serine/threonine kinases best known for its critical role in learning and memory. Recent studies suggested that high levels of CaMKII also expressed in variety of malignant diseases. In this review, we focus on the structure and biology properties of CaMKII, including the role of CaMKII in the regulation of cancer progression and therapy response. We also describe the role of CaMKII in the diagnosis of different kinds of cancer and recent progress in the development of CaMKII inhibitors. These data establishes CaMKII as a novel target whose modulation presents new opportunities for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yan-yang Wang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ren Zhao
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hong Zhe
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.,Cancer Institute, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
48
|
Park M, Song C, Yoon H, Choi KH. Double Blockade of Glioma Cell Proliferation and Migration by Temozolomide Conjugated with NPPB, a Chloride Channel Blocker. ACS Chem Neurosci 2016; 7:275-85. [PMID: 26711895 DOI: 10.1021/acschemneuro.5b00178] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma is the most common and aggressive primary malignant brain tumor. Temozolomide (TMZ), a chemotherapeutic agent combined with radiation therapy, is used as a standard treatment. The infiltrative nature of glioblastoma, however, interrupts effective treatment with TMZ and increases the tendency to relapse. Voltage-gated chloride channels have been identified as crucial regulators of glioma cell migration and invasion by mediating cell shape and volume change. Accordingly, chloride current inhibition by 5-nitro-2-(3-phenylpropylamino)-benzoate (NPPB), a chloride channel blocker, suppresses cell movement by diminishing the osmotic cell volume regulation. In this study, we developed a novel compound, TMZ conjugated with NPPB (TMZ-NPPB), as a potential anticancer drug. TMZ-NPPB blocked chloride currents in U373MG, a severely invasive human glioma cell line, and suppressed migration and invasion of U373MG cells. Moreover, TMZ-NPPB exhibited DNA modification activity similar to that of TMZ, and surprisingly showed remarkably enhanced cytotoxicity relative to TMZ by inducing apoptotic cell death via DNA damage. These findings indicate that TMZ-NPPB has a dual function in blocking both proliferation and migration of human glioma cells, thereby suggesting its potential to overcome challenges in current glioblastoma therapy.
Collapse
Affiliation(s)
- Miri Park
- Department of Biological Chemistry, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Chiman Song
- Materials
and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Hojong Yoon
- Materials
and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Kee-Hyun Choi
- Department of Biological Chemistry, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
- Materials
and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
49
|
Zhao X, Xu L, Zheng L, Yin L, Qi Y, Han X, Xu Y, Peng J. Potent effects of dioscin against gastric cancer in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:274-282. [PMID: 26969381 DOI: 10.1016/j.phymed.2016.01.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/24/2016] [Accepted: 01/26/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND We previously reported the effect of dioscin on human gastric carcinoma SGC-7901 cells, but its effects on other gastric cancers are still unknown. PURPOSE The present paper aimed to demonstrate the activity of dioscin against human gastric carcinoma MGC-803 and MKN-45. STUDY DESIGN In our study, MGC-803 and MKN-45 cells were used to examine the effects of dioscin on human gastric carcinoma in vitro. The effects of dioscin against human gastric carcinoma in vivo were accomplished by the xenografts of MGC-803 cells in BALB/c nude mice. METHODS AO/EB and DAPI staining, TEM, single cell gel electrophoresis and flow cytometry assays were used in cell experiments. Then, an iTRAQ-based proteomics approach, DNA and siRNA transfection experiments were carried out for mechanism investigation. RESULTS In MGC-803 cells, dioscin caused DNA damage and mitochondrial change, induced ROS generation, Ca(2+) release and cell apoptosis, and blocked cell cycle at S phase. In vivo results showed that dioscin significantly suppressed the tumor growth of MGC-803 cell xenografts in nude mice. In addition, dioscin markedly inhibited cell migration, caused Cytochrome c release and adjusted mitochondrial signal pathway. Then, an iTRAQ-based proteomics approach was carried out and 121 differentially expressed proteins were found, in which five biomarkers associated with cell cycle, apoptosis and migration were evaluated. Dioscin significantly up-regulated the levels of GALR-2 and RBM-3, and down-regulated CAP-1, Tribbles-2 and CliC-3. Furthermore, overexpressed DNA transfection of CAP-1 enhanced cell migration and invasion, which was decreased by dioscin. SiRNA to Tribbles-2 affected the protein levels of Bcl-2, Bax and MAPKs, suggesting that dioscin decreased Tribbles-2 level leading to cell apoptosis. CONCLUSION Our works confirmed the activity of dioscin against gastric cancer. In addition, this work also provided that dioscin is a new potent candidate for treating gastric cancer in the future.
Collapse
Affiliation(s)
- Xinwei Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lingli Zheng
- Department of Pharmaceuticals, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
50
|
Guan YT, Huang YQ, Wu JB, Deng ZQ, Wang Y, Lai ZY, Wang HB, Sun XX, Zhu YL, Du MM, Zhu LY, Chen LX, Wang LW. Overexpression of chloride channel-3 is associated with the increased migration and invasion ability of ectopic endometrial cells from patients with endometriosis. Hum Reprod 2016; 31:986-98. [PMID: 26965430 DOI: 10.1093/humrep/dew034] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/07/2016] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION Is chloride channel-3 (ClC-3) involved in regulating the biological behavior of endometrial stromal cells (ESCs)? SUMMARY ANSWER ClC-3 promotes endometriotic cell migration and invasion. WHAT IS KNOWN ALREADY ClC-3 plays a significant role in the migration and invasion of various kinds of cells. STUDY DESIGN, SIZE, DURATION An ITALIC! in vitro investigation of the effect of ClC-3 on the migration and invasion of ectopic ESCs from patients with endometriosis. PARTICIPANTS/MATERIALS, SETTING, METHODS The ectopic and eutopic endometrial samples from 43 female patients with endometriosis and the endometrial samples from 39 non-endometriotic female patients were collected. Primary cells from these samples were isolated and cultured. Real-time RT-PCR, immunohistochemistry and western blot were used to detect the expression of ClC-3 and matrix metalloproteinase 9 (MMP-9). Small interfering RNA (siRNA) technology was employed to knock down ClC-3 expression. The migration and invasion ability of ESCs was measured by the transwell assay with uncoated or Matrigel-coated membranes. MAIN RESULTS AND THE ROLE OF CHANCE The expression of ClC-3 mRNA and proteins was significantly up-regulated in the ectopic tissues from endometriotic patients, while that in the eutopic endometrial tissues of the same patients did not significantly differ from that in non-endometriotic patients. The migration and invasion ability and MMP-9 expression was increased in the ESCs from ectopic endometrial tissues. The knockdown of ClC-3 expression by ClC-3 siRNA inhibited ESC migration and invasion and attenuated the expression of MMP-9. ClC-3 expression level was well-correlated to the clinical characteristics and symptoms of endometriosis patients, including infertility, dysmenorrhea, chronic pelvic pain, dyspareunia and diameter of endometriosis lesion. LIMITATIONS, REASONS FOR CAUTION Further studies are needed to examine the regulatory mechanism of estrogen on ClC-3 expression of ESCs. WIDER IMPLICATIONS OF THE FINDINGS ClC-3 is involved in the migration and invasion processes of ESCs and can regulate MMP-9 expression. Up-regulation of ClC-3 expression may contribute to endometriosis development by regulating MMP-9 expression. STUDY FUNDING/COMPETING INTERESTS This work was supported by the National Natural Science Foundation of China (81173064, 81272223, 81273539), the Ministry of Education of China (20124401110009), the Natural Science Foundation of Guangdong Province (S2011010001589) and the Science and Technology Programs of Guangdong (2013B051000059), Guangzhou (2013J500015) and Dongguan (2011108102006). The authors have no conflict of interest.
Collapse
Affiliation(s)
- Yu-tao Guan
- Department of Pharmacology, Medical College, Jinan University, Guangzhou 510632, China Department of Pathophysiology, Medical College, Jinan University, Guangzhou, China
| | - Yan-qing Huang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Jia-bao Wu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou 510632, China
| | - Zhi-qin Deng
- Department of Pharmacology, Medical College, Jinan University, Guangzhou 510632, China Department of Pathophysiology, Medical College, Jinan University, Guangzhou, China
| | - Yuan Wang
- Department of Physiology, Medical College, Jinan University, Guangzhou 510632, China
| | - Zhou-yi Lai
- Department of Physiology, Medical College, Jinan University, Guangzhou 510632, China
| | - Hai-bo Wang
- Department of Pharmacology, Medical College, Jinan University, Guangzhou 510632, China
| | - Xiao-xue Sun
- Department of Physiology, Medical College, Jinan University, Guangzhou 510632, China
| | - Ya-li Zhu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Miao-miao Du
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Lin-yan Zhu
- Department of Pharmacology, Medical College, Jinan University, Guangzhou 510632, China
| | - Li-xin Chen
- Department of Pharmacology, Medical College, Jinan University, Guangzhou 510632, China Department of Pathophysiology, Medical College, Jinan University, Guangzhou, China
| | - Li-wei Wang
- Department of Pathophysiology, Medical College, Jinan University, Guangzhou, China Department of Physiology, Medical College, Jinan University, Guangzhou 510632, China
| |
Collapse
|