1
|
Zhang L, Gu H, Li X, Wang Y, Yao S, Chen X, Zheng L, Yang X, Du Q, An J, Wen G, Zhu J, Jin H, Tuo B. Pathophysiological role of ion channels and transporters in hepatocellular carcinoma. Cancer Gene Ther 2024; 31:1611-1618. [PMID: 39048663 PMCID: PMC11567900 DOI: 10.1038/s41417-024-00782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 07/27/2024]
Abstract
The incidence of hepatocellular carcinoma (HCC) has continued to increase annually worldwide, and HCC has become a common cause of cancer-related death. Despite great progress in understanding the molecular mechanisms underlying HCC development, the treatment of HCC remains a considerable challenge. Thus, the survival and prognosis of HCC patients remain extremely poor. In recent years, the role of ion channels in the pathogenesis of diseases has become a hot topic. In normal liver tissue, ion channels and transporters maintain water and electrolyte balance and acid‒base homeostasis. However, dysfunction of these ion channels and transporters can lead to the development and progression of HCC, and thus these ion channels and transporters are expected to become new therapeutic targets. In this review, ion channels and transporters associated with HCC are reviewed, and potential targets for new and effective therapies are proposed.
Collapse
Affiliation(s)
- Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| | - Hong Gu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xin Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yongfeng Wang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Shun Yao
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xingyue Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Liming Zheng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xingyue Yang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qian Du
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiaxing An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Guorong Wen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Hai Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou, China.
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
2
|
Olver DJ, Azam I, Benson JD. HepG2 cells undergo regulatory volume decrease by mechanically induced efflux of water and solutes. Biomech Model Mechanobiol 2024; 23:1781-1799. [PMID: 39012455 DOI: 10.1007/s10237-024-01868-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/12/2024] [Indexed: 07/17/2024]
Abstract
This study challenges the conventional belief that animal cell membranes lack a significant hydrostatic gradient, particularly under anisotonic conditions, as demonstrated in the human hepatoma cell line HepG2. The Boyle van't Hoff (BvH) relation describes volumetric equilibration to anisotonic conditions for many cells. However, the BvH relation is simple and does not include many cellular components such as the cytoskeleton and actin cortex, mechanosensitive channels, and ion pumps. Here we present alternative models that account for mechanical resistance to volumetric expansion, solute leakage, and active ion pumping. We found the BvH relation works well to describe hypertonic volume equilibration but not hypotonic volume equilibration. After anisotonic exposure and return isotonic conditions cell volumes were smaller than their initial isotonic volume, indicating solutes had leaked out of the cell during swelling. Finally, we observed HepG2 cells undergo regulatory volume decrease at both 20 °C and 4 °C, indicating regulatory volume decrease to be a relatively passive phenomenon and not driven by ion pumps. We determined the turgor-leak model, which accounts for mechanical resistance and solute leakage, best fits the observations found in the suite of experiments performed, while other models were rejected.
Collapse
Affiliation(s)
- Dominic J Olver
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK, S7N 5E2, Canada
| | - Iqra Azam
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK, S7N 5E2, Canada
| | - James D Benson
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK, S7N 5E2, Canada.
| |
Collapse
|
3
|
Chandran C, Santra M, Rubin E, Geary ML, Yam GHF. Regenerative Therapy for Corneal Scarring Disorders. Biomedicines 2024; 12:649. [PMID: 38540264 PMCID: PMC10967722 DOI: 10.3390/biomedicines12030649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 05/09/2024] Open
Abstract
The cornea is a transparent and vitally multifaceted component of the eye, playing a pivotal role in vision and ocular health. It has primary refractive and protective functions. Typical corneal dysfunctions include opacities and deformities that result from injuries, infections, or other medical conditions. These can significantly impair vision. The conventional challenges in managing corneal ailments include the limited regenerative capacity (except corneal epithelium), immune response after donor tissue transplantation, a risk of long-term graft rejection, and the global shortage of transplantable donor materials. This review delves into the intricate composition of the cornea, the landscape of corneal regeneration, and the multifaceted repercussions of scar-related pathologies. It will elucidate the etiology and types of dysfunctions, assess current treatments and their limitations, and explore the potential of regenerative therapy that has emerged in both in vivo and clinical trials. This review will shed light on existing gaps in corneal disorder management and discuss the feasibility and challenges of advancing regenerative therapies for corneal stromal scarring.
Collapse
Affiliation(s)
- Christine Chandran
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Mithun Santra
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Elizabeth Rubin
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Moira L. Geary
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
| | - Gary Hin-Fai Yam
- Corneal Regeneration Laboratory, Department of Ophthalmology, Mercy Vision Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; (C.C.); (M.S.); (E.R.); (M.L.G.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
4
|
Chen X, Zhang L, He L, Zheng L, Tuo B. Potassium channels as novel molecular targets in hepatocellular carcinoma (Review). Oncol Rep 2023; 50:185. [PMID: 37654193 PMCID: PMC10485806 DOI: 10.3892/or.2023.8622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/31/2023] [Indexed: 09/02/2023] Open
Abstract
Hepatocellular carcinoma (HCC) poses a serious health burden worldwide. It is often not diagnosed until the patient is at an advanced stage of the disease, when treatment options are limited and the prognosis is poor. Therefore, novel treatment strategies are urgently required. Potassium (K+) channels have an important role in HCC, including regulating the proliferation, migration, invasion and drug resistance of HCC cells. The aim of the present review was therefore to survey the relevant publications that have investigated K+ channels not only as markers for the early diagnosis of HCC, but also as potential therapeutic targets for the treatment of HCC. Several of these channels have been indicated to be the sites of action for natural products previously known to inhibit HCC; however, more systematic studies are required to determine which K+ channels may be utilized for the clinical treatment of HCC, particularly in the advanced stages of the disease and in cases where patients are resistant to the existing drugs.
Collapse
Affiliation(s)
- Xingyue Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ling He
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Liming Zheng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
5
|
Wubulikasimu A, He Z, Long Y, Yuan F, Hou W, Liao Q, Chen H, Rong M. Molecular mechanism of HNTX-I activates the intermediate-conductance Ca 2+-activated K + (IK) channels. Int J Biol Macromol 2023:125197. [PMID: 37285887 DOI: 10.1016/j.ijbiomac.2023.125197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
The IK channel, a potassium ion channel regulated by calcium ions and voltages in a bidirectional manner, has been implicated in a range of diseases. However, there are currently few compounds available that can target the IK channel with high potency and specificity. Hainantoxin-I (HNTX-I) is the first peptide activator of IK channel discovered so far, but its activity is not ideal, and the underlying mechanism interaction between HNTX-I toxin and IK channel remains unclear. Thus, our study aimed to enhance the potency of IK channel activating peptides derived from HNTX-I and elucidate the molecular mechanism underlying the interaction between HNTX-I and the IK channel. By employing virtual alanine scanning mutagenesis, we generated 11 HNTX-I mutants using site-directed mutagenesis to pinpoint specific residues crucial for the HNTX-I and IK channel interaction. Subsequently, we identified key residues on the IK channel that are involved in the interaction with HNTX-I. Additionally, molecular docking was employed to guide the molecular engineering process and clarify the binding interface between HNTX-I and the IK channel. Our results demonstrate that HNTX-I primarily acts on the IK channel via the N-terminal amino acid, and its interaction with the IK channel is mediated by electrostatic and hydrophobic interactions, specifically the amino acid residues at positions 1, 3, 5, and 7 on HNTX-I. This study provides valuable insights into the peptide toxins that may serve as potential templates for the development of activators with enhanced potency and selectivity for the IK channel.
Collapse
Affiliation(s)
- Atikan Wubulikasimu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Ziyan He
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Yanping Long
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Fuchu Yuan
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Wenqian Hou
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Qiong Liao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China
| | - Haiyan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China.
| | - Mingqiang Rong
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, People's Republic of China.
| |
Collapse
|
6
|
Xia C, Liu C, Ren S, Cai Y, Zhang Q, Xia C. Potassium channels, tumorigenesis and targeted drugs. Biomed Pharmacother 2023; 162:114673. [PMID: 37031494 DOI: 10.1016/j.biopha.2023.114673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
Potassium channels play an important role in human physiological function. Recently, various molecular mechanisms have implicated abnormal functioning of potassium channels in the proliferation, migration, invasion, apoptosis, and cancer stem cell phenotype formation. Potassium channels also mediate the association of tumor cells with the tumor microenvironment. Meanwhile, potassium channels are important targets for cancer chemotherapy. A variety of drugs exert anti-cancer effects by modulating potassium channels in tumor cells. Therefore, there is a need to understand how potassium channels participate in tumor development and progression, which could reveal new, novel targets for cancer diagnosis and treatment. This review summarizes the roles of voltage-gated potassium channels, calcium-activated potassium channels, inwardly rectifying potassium channels, and two-pore domain potassium channels in tumorigenesis and the underlying mechanism of potassium channel-targeted drugs. Therefore, the study lays the foundation for rational and effective drug design and individualized clinical therapeutics.
Collapse
Affiliation(s)
- Cong Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China
| | - Can Liu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Shuangyi Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China
| | - Yantao Cai
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China
| | - Qianshi Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116023, China.
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, Guangdong Province 528099, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
7
|
Zhang Y, Ma XZ, Zhao XY, Li JJ, Ma S, Pang ZD, Xu J, Du XJ, Deng XL, Wang JH. AGEs-RAGE-KCa3.1 pathway mediates palmitic acid-induced migration of PBMCs from patients with type 2 diabetes. Heliyon 2023; 9:e14823. [PMID: 37025887 PMCID: PMC10070889 DOI: 10.1016/j.heliyon.2023.e14823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by chronic low-grade systemic inflammation. Tissue infiltration by monocyte migration contributes to the pathogenesis of vascular complications in T2DM. We studied the role of intermediate-conductance Ca2+-activated K+ (KCa3.1) channels in the palmitic acid (PA)-induced migration of peripheral blood mononuclear cells (PBMCs) from T2DM patients and the influence of advanced glycation endproducts (AGEs). A total of 49 T2DM patients and 33 healthy subjects was recruited into this study. Using flow cytometry and Western blotting analysis as well as cell migration assay, we found that there was a significant decrease in frequency of T lymphocytes and monocytes in CD45+ leukocyte population. PA at 100 μM stimulated migration of PBMCs from T2DM individuals, which was inhibited by the specific KCa3.1 channel blocker TRAM-34 (1 μM). The PBMC migration was positively correlated with glycosylated hemoglobin A1 chain (HbA1c) level of T2DM patients, an indicator of AGEs, and PBMCs with higher level of HbA1c showed upregulated expression of toll-like receptor (TLR) 2/4 and KCa3.1 channels. In THP-1 cells, AGEs at 200 μg/ml increased protein expression of TLR 2/4 and KCa3.1 channels, and were synergistically involved in PA-induced migration through receptors of AGEs (RAGE)-mediated KCa3.1 upregulation. In conclusion, in PBMCs of T2DM patients, AGEs promotes PA-induced migration via upregulation of TLR2/4 and KCa3.1 channels.
Collapse
|
8
|
Cortés Franco KD, Brakmann IC, Feoktistova M, Panayotova-Dimitrova D, Gründer S, Tian Y. Aggressive migration in acidic pH of a glioblastoma cancer stem cell line in vitro is independent of ASIC and K Ca3.1 ion channels, but involves phosphoinositide 3-kinase. Pflugers Arch 2023; 475:405-416. [PMID: 36522586 PMCID: PMC9908655 DOI: 10.1007/s00424-022-02781-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
The microenvironment of proliferative and aggressive tumours, such as the brain tumour glioblastoma multiforme (GBM), is often acidic, hypoxic, and nutrient deficient. Acid-sensing ion channels (ASICs) are proton-sensitive Na+ channels that have been proposed to play a role in pH sensing and in modulation of cancer cell migration. We previously reported that primary glioblastoma stem cells (GSCs), which grow as multicellular tumour spheroids, express functional ASIC1a and ASIC3, whereas ASIC2a is downregulated in GSCs. Using a 2.5D migration assay, here we report that acidic pH dramatically increased migration of GSCs of the pro-neural subtype. Pharmacological blockade as well as CRISPR-Cas9-mediated gene knock-out of ASIC1a or stable overexpression of ASIC2a, however, revealed that neither ASIC1a nor ASIC3, nor downregulation of ASIC2a, mediated the aggressive migration at acidic pH. Therefore, we tested the role of two other proteins previously implicated in cancer cell migration: the Ca2+-activated K+ channel KCa3.1 (KCNN4) and phosphoinositide 3-kinase (PI3K). While pharmacological blockade of KCa3.1 did also not affect migration, blockade of PI3K decreased migration at acidic pH to control levels. In summary, our study reveals a strongly enhanced migration of GSCs at acidic pH in vitro and identifies PI3K as an important mediator of this effect.
Collapse
Affiliation(s)
| | - Ilka C Brakmann
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, D-52074, Aachen, Germany
| | - Maria Feoktistova
- Department of Dermatology, RWTH Aachen University, Pauwelsstraße 30, D-52074, Aachen, Germany
| | | | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, D-52074, Aachen, Germany.
| | - Yuemin Tian
- Institute of Physiology, RWTH Aachen University, Pauwelsstraße 30, D-52074, Aachen, Germany
| |
Collapse
|
9
|
Bulk E, Todesca LM, Bachmann M, Szabo I, Rieke M, Schwab A. Functional expression of mitochondrial K Ca3.1 channels in non-small cell lung cancer cells. Pflugers Arch 2022; 474:1147-1157. [PMID: 36152073 PMCID: PMC9560933 DOI: 10.1007/s00424-022-02748-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/21/2022] [Accepted: 09/05/2022] [Indexed: 12/01/2022]
Abstract
Lung cancer is one of the leading causes of cancer-related deaths worldwide. The Ca2+-activated K+ channel KCa3.1 contributes to the progression of non-small cell lung cancer (NSCLC). Recently, KCa3.1 channels were found in the inner membrane of mitochondria in different cancer cells. Mitochondria are the main sources for the generation of reactive oxygen species (ROS) that affect the progression of cancer cells. Here, we combined Western blotting, immunofluorescence, and fluorescent live-cell imaging to investigate the expression and function of KCa3.1 channels in the mitochondria of NSCLC cells. Western blotting revealed KCa3.1 expression in mitochondrial lysates from different NSCLC cells. Using immunofluorescence, we demonstrate a co-localization of KCa3.1 channels with mitochondria of NSCLC cells. Measurements of the mitochondrial membrane potential with TMRM reveal a hyperpolarization following the inhibition of KCa3.1 channels with the cell-permeable blocker senicapoc. This is not the case when cells are treated with the cell-impermeable peptidic toxin maurotoxin. The hyperpolarization of the mitochondrial membrane potential is accompanied by an increased generation of ROS in NSCLC cells. Collectively, our results provide firm evidence for the functional expression of KCa3.1 channels in the inner membrane of mitochondria of NSCLC cells.
Collapse
Affiliation(s)
- Etmar Bulk
- Institute of Physiology II, University of Münster, 48149, Münster, Germany.
| | | | | | - Ildiko Szabo
- Department of Biology, University of Padova, Padua, Italy
| | - Marius Rieke
- Institute of Physiology II, University of Münster, 48149, Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University of Münster, 48149, Münster, Germany
| |
Collapse
|
10
|
Zúñiga L, Cayo A, González W, Vilos C, Zúñiga R. Potassium Channels as a Target for Cancer Therapy: Current Perspectives. Onco Targets Ther 2022; 15:783-797. [PMID: 35899081 PMCID: PMC9309325 DOI: 10.2147/ott.s326614] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/08/2022] [Indexed: 12/18/2022] Open
Abstract
Potassium (K+) channels are highly regulated membrane proteins that control the potassium ion flux and respond to different cellular stimuli. These ion channels are grouped into three major families, Kv (voltage-gated K+ channel), Kir (inwardly rectifying K+ channel) and K2P (two-pore K+ channels), according to the structure, to mediate the K+ currents. In cancer, alterations in K+ channel function can promote the acquisition of the so-called hallmarks of cancer – cell proliferation, resistance to apoptosis, metabolic changes, angiogenesis, and migratory capabilities – emerging as targets for the development of new therapeutic drugs. In this review, we focus our attention on the different K+ channels associated with the most relevant and prevalent cancer types. We summarize our knowledge about the potassium channels structure and function, their cancer dysregulated expression and discuss the K+ channels modulator and the strategies for designing new drugs.
Collapse
Affiliation(s)
- Leandro Zúñiga
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Angel Cayo
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Wendy González
- Centro de Bioinformática y Simulación Molecular, Universidad de Talca, Talca, Chile.,Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Talca, Talca, Chile
| | - Cristian Vilos
- Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile.,Laboratory of Nanomedicine and Targeted Delivery, School of Medicine, Universidad de Talca, Talca, 3460000, Chile.,Center for The Development of Nanoscience & Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, 8350709, Chile
| | - Rafael Zúñiga
- Laboratorio de Fisiología Molecular, Escuela de Medicina, Universidad de Talca, Talca, Chile.,Centro de Nanomedicina, Diagnóstico y Desarrollo de Fármacos (ND3), Escuela de Medicina, Universidad de Talca, Talca, Chile
| |
Collapse
|
11
|
Liu Z, Fu J, Yuan H, Ma B, Cao Z, Chen Y, Xing C, Niu X, Li N, Wang H, An H. Polyisocyanide hydrogels with tunable nonlinear elasticity mediate liver carcinoma cell functional response. Acta Biomater 2022; 148:152-162. [PMID: 35718101 DOI: 10.1016/j.actbio.2022.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 11/27/2022]
Abstract
Hepatocellular carcinoma development is closely related to the changes in tissue mechanics induced by excess collagen deposition and crosslinking, which leads to liver fibrosis and malignant progression. The role of matrix stiffness has been widely assessed using various linearly elastic materials. However, the liver, like many soft tissues, also exhibits nonlinear elasticity by strain-stiffening, allowing cells to mechanically interact with their micromilieus which has attracted much attention in cellular processes recently. Here, we use a biomimetic hydrogel grafting of GRGDS peptide with tunable nonlinear mechanical properties, polyisocyanides (PIC), to investigate the influence of strain-stiffening on HepG2 liver cancer cell behavior by tuning PIC polymer length. Compared to short PIC polymer with lower critical stress, PIC hydrogels composed of long polymer with higher critical stress promote the motility and invasiveness of HepG2 cells, and induce more actin stress fibers and higher expression level of mechanotransducer YAP and its nuclear translocation. Strikingly, the expression of calcium-activated potassium channel KCa3.1, an important biomarker in hepatocellular carcinoma, is also affected by the mechanical property of PIC hydrogels. It was also shown that downregulating the KCa3.1 channel can be achieved by inhibiting the formation of actin fibers. Our findings imply that the strain-stiffening property of PIC hydrogels affects the expression of KCa3.1 potassium channel via mediating cytoskeletal stress fiber formation, and ultimately influences the liver carcinoma cell functional response. STATEMENT OF SIGNIFICANCE: The effect of nonlinear elasticity by strain-stiffening, is assessed in HepG2 liver cancer cell behavior by using a biomimetic hydrogel with tunable mechanical properties, polyisocyanides (PIC). PIC gels with higher critical stress promote the motility and invasiveness of HepG2 cells and induce upregulated expression levels of KCa3.1 potassium channel and YAP, but which can be suppressed by inhibiting the formation of actin fibers. Our findings imply that the strain-stiffening property of PIC gels influences the expression of KCa3.1 potassium channel via mediating cytoskeletal stress fiber formation and, ultimately affects the liver carcinoma cell functional response.
Collapse
Affiliation(s)
- Zixin Liu
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Jingxuan Fu
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China; College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China; School of Chemical Engineering and Technology, Hebei University of Technology, GuangRong Dao 8, Hongqiao District, Tianjin 300130, PR China
| | - Hongbo Yuan
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China; Molecular Imaging and Photonics, Chemistry Department, KU Leuven, Celestijnenlaan 200F, 3001 Heverlee, Belgium.
| | - Biao Ma
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Zhanshuo Cao
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Yafei Chen
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Chengfen Xing
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Xuezhi Niu
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Ning Li
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China
| | - Hui Wang
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China; College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin 300071, PR China.
| | - Hailong An
- Key Laboratory of Hebei Province for Molecular Biophysics, Institute of Biophysics, School of Science, Hebei University of Technology, Tianjin 300401, PR China.
| |
Collapse
|
12
|
Khreesha L, Qaswal AB, Al Omari B, Albliwi MA, Ababneh O, Albanna A, Abunab'ah A, Iswaid M, Alarood S, Guzu H, Alshawabkeh G, Zayed FM, Abuhilaleh MA, Al-Jbour MN, Obeidat S, Suleiman A. Quantum Tunneling-Induced Membrane Depolarization Can Explain the Cellular Effects Mediated by Lithium: Mathematical Modeling and Hypothesis. MEMBRANES 2021; 11:851. [PMID: 34832080 PMCID: PMC8625630 DOI: 10.3390/membranes11110851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022]
Abstract
Lithium imposes several cellular effects allegedly through multiple physiological mechanisms. Membrane depolarization is a potential unifying concept of these mechanisms. Multiple inherent imperfections of classical electrophysiology limit its ability to fully explain the depolarizing effect of lithium ions; these include incapacity to explain the high resting permeability of lithium ions, the degree of depolarization with extracellular lithium concentration, depolarization at low therapeutic concentration, or the differences between the two lithium isotopes Li-6 and Li-7 in terms of depolarization. In this study, we implemented a mathematical model that explains the quantum tunneling of lithium ions through the closed gates of voltage-gated sodium channels as a conclusive approach that decodes the depolarizing action of lithium. Additionally, we compared our model to the classical model available and reported the differences. Our results showed that lithium can achieve high quantum membrane conductance at the resting state, which leads to significant depolarization. The quantum model infers that quantum membrane conductance of lithium ions emerges from quantum tunneling of lithium through the closed gates of sodium channels. It also differentiates between the two lithium isotopes (Li-6 and Li-7) in terms of depolarization compared with the previous classical model. Moreover, our study listed many examples of the cellular effects of lithium and membrane depolarization to show similarity and consistency with model predictions. In conclusion, the study suggests that lithium mediates its multiple cellular effects through membrane depolarization, and this can be comprehensively explained by the quantum tunneling model of lithium ions.
Collapse
Affiliation(s)
- Lubna Khreesha
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | | | - Baheth Al Omari
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | | | - Omar Ababneh
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Ahmad Albanna
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | | | - Mohammad Iswaid
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Salameh Alarood
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Hasan Guzu
- Anesthesia Department, Farah Medical Campus, 18 Mai Zeyadeh Street, Amman 11942, Jordan
| | - Ghadeer Alshawabkeh
- Anesthesia and Pain Management Department, King Hussein Cancer Center, Amman 11942, Jordan
| | | | | | | | - Salameh Obeidat
- Department of Anesthesia, Intensive Care and Pain Management, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Aiman Suleiman
- Department of Anesthesia, Intensive Care and Pain Management, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| |
Collapse
|
13
|
Qian Y, Li Y, Li R, Yang T, Jia R, Ge YZ. circ-ZNF609: A potent circRNA in human cancers. J Cell Mol Med 2021; 25:10349-10361. [PMID: 34697887 PMCID: PMC8581316 DOI: 10.1111/jcmm.16996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/22/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022] Open
Abstract
Circular RNAs (circRNAs) are a novel group of endogenous RNAs with a circular structure. Growing evidence indicates that circRNAs are involved in a variety of human diseases including malignancies. CircRNA ZNF609 (circ‐ZNF609), derived from the ZNF609 gene sequence, has been demonstrated to be involved in the development and progression of many diseases. circ‐ZNF609 is thought to be a viable diagnostic and prognostic biomarker for several diseases and might be a new therapeutic target, but further research is needed to accelerate clinical application. Here, we review the biogenesis and function of circRNAs and the functional roles and molecular mechanism related to circ‐ZNF609 in neoplasms and other diseases.
Collapse
Affiliation(s)
- Yiguan Qian
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Rongfei Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tianli Yang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yu-Zheng Ge
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Abstract
Neoplastic transformation is reportedly associated with alterations of the potassium transport across plasma and intracellular membranes. These alterations have been identified as crucial elements of the tumourigenic reprogramming of cells. Potassium channels may contribute to cancer initiation, malignant progression and therapy resistance of tumour cells. The book chapter focusses on (oncogenic) potassium channels frequently upregulated in different tumour entities, upstream and downstream signalling of these channels, their contribution to the maintenance of cancer stemness and the formation of an immunosuppressive tumour microenvironment. In addition, their role in adaptation to tumour hypoxia, metabolic reprogramming, as well as tumour spreading and metastasis is discussed. Finally, we discuss how (oncogenic) potassium channels may confer treatment resistance of tumours against radiation and chemotherapy and thus might be harnessed for new therapy strategies, for instance, by repurposing approved drugs known to target potassium channels.
Collapse
|
15
|
Bukhari M, Deng H, Sipes D, Ruane-Foster M, Purdy K, Woodworth CD, Sur S, Samways DSK. K Ca3.1-dependent uptake of the cytotoxic DNA-binding dye Hoechst 33258 into cancerous but not healthy cervical cells. J Biol Chem 2021; 296:100084. [PMID: 33199365 PMCID: PMC7948979 DOI: 10.1074/jbc.ra120.013997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/03/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
The poor and nonselective penetration of current chemotherapeutics across the plasma membranes of cancer cells, which is necessary for the targeted disruption of the intracellular machinery, remains a major pharmaceutical challenge. In several cell types, including mast cells and macrophages, exposure to extracellular ATP is known to stimulate passive entry of large and otherwise membrane impermeable cationic dyes, which is usually attributed to conduction through ionotropic P2X receptors. Here, we report that elevations in cytosolic Ca2+ stimulate the rapid uptake and nuclear accumulation of a DNA-binding fluorescent cation, Hoechst 33258 (H33258), in cervical cancer cells. The H33258 uptake was dependent on activation of intermediate conductance Ca2+-activated K+ channels (KCa3.1), and direct stimulation of the channel with the activators SKA 31 and DCEBIO was sufficient to induce cellular uptake of H33258 directly. In contrast to the results from cancerous cervical cells, KCa3.1-dependent H33258 uptake was rarely observed in epithelial cells derived from the ectocervix and transformation zone of healthy cervical tissue. Furthermore, whole-cell patch clamp experiments and assessment of membrane potential using the slow voltage-sensitive dye bis-(1,3-diethylthiobarbituric acid)trimethine oxonol revealed a significant difference in functional KCa3.1 activity between cancerous and healthy cervical epithelial cells, which correlated strongly with the incidence of KCa3.1-dependent H33258 uptake. Finally, we show that activation of KCa3.1 channels caused a modest but significant sensitization of cancer cells to the growth suppressant effects of H33258, lending plausibility to the idea of using KCa3.1 channel activators to enhance cell penetration of small cationic toxins into cancer cells expressing these channels.
Collapse
Affiliation(s)
- Maurish Bukhari
- Department of Biology, Clarkson University, Potsdam, New York, USA
| | - Han Deng
- Department of Biology, Clarkson University, Potsdam, New York, USA
| | - Darren Sipes
- Department of Biology, Clarkson University, Potsdam, New York, USA
| | | | - Kayla Purdy
- Department of Biology, Clarkson University, Potsdam, New York, USA
| | | | - Shantanu Sur
- Department of Biology, Clarkson University, Potsdam, New York, USA
| | | |
Collapse
|
16
|
Lin P, Li J, Ye F, Fu W, Hu X, Shao Z, Song C. KCNN4 induces multiple chemoresistance in breast cancer by regulating BCL2A1. Am J Cancer Res 2020; 10:3302-3315. [PMID: 33163271 PMCID: PMC7642670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/08/2020] [Indexed: 06/11/2023] Open
Abstract
Multidrug chemoresistance is a major clinical obstacle in breast cancer treatment. We aimed to elucidate the sensitivity to therapeutics in gemcitabine-resistant breast cancer models. Pooled library screening combined with RNA-seq was conducted to explore the potential targets involved in gemcitabine resistance in breast cancer cells. Cytotoxicity and tumor xenograft assays were used to evaluate the effect of calcium-activated channel subfamily N member 4 (KCNN4) inhibitors on the cellular sensitivity of breast cancer cells to chemotherapeutic drugs both in vitro and in vivo. We found that KCNN4 is an important determinant for the cytotoxicity of gemcitabine. Elevated KCNN4 expression enhanced resistance to chemotherapeutic antimetabolites and promoted cell proliferation. Conversely, silencing KCNN4 or chemical inhibition of KCNN4 by the specific inhibitor TRAM-34 inhibited the chemoresistance and cell proliferation. Mechanistically, KCNN4 upregulated BCL2-related protein A1 (BCL2A1) to suppress apoptosis by activating RAS-MAPK and PI3K-AKT signaling. Moreover, high expression levels of KCNN4 and BCL2A1 were associated with shortened disease-free survival in the cohort studies. Collectively, our findings showed that KCNN4 is a key modulator of progression and drug resistance in breast cancer, indicating that targeting KCNN4 may serve as a promising therapeutic strategy to overcome multidrug chemoresistance in this disease.
Collapse
Affiliation(s)
- Peiyang Lin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer CenterShanghai, China
- Department of Breast Surgery, Fujian Medical University Union HospitalFuzhou, China
| | - Junjing Li
- Department of Breast Surgery, Fujian Medical University Union HospitalFuzhou, China
| | - Fugui Ye
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer CenterShanghai, China
| | - Wenfen Fu
- Department of Breast Surgery, Fujian Medical University Union HospitalFuzhou, China
| | - Xin Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer CenterShanghai, China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer CenterShanghai, China
| | - Zhiming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer CenterShanghai, China
- Institutes of Biomedical Science, Fudan UniversityShanghai, China
- Precision Cancer Medicine Center, Fudan University Shanghai Cancer CenterShanghai, China
| | - Chuangui Song
- Department of Breast Surgery, Fujian Medical University Union HospitalFuzhou, China
| |
Collapse
|
17
|
How Dysregulated Ion Channels and Transporters Take a Hand in Esophageal, Liver, and Colorectal Cancer. Rev Physiol Biochem Pharmacol 2020; 181:129-222. [PMID: 32875386 DOI: 10.1007/112_2020_41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the last two decades, the understanding of how dysregulated ion channels and transporters are involved in carcinogenesis and tumor growth and progression, including invasiveness and metastasis, has been increasing exponentially. The present review specifies virtually all ion channels and transporters whose faulty expression or regulation contributes to esophageal, hepatocellular, and colorectal cancer. The variety reaches from Ca2+, K+, Na+, and Cl- channels over divalent metal transporters, Na+ or Cl- coupled Ca2+, HCO3- and H+ exchangers to monocarboxylate carriers and organic anion and cation transporters. In several cases, the underlying mechanisms by which these ion channels/transporters are interwoven with malignancies have been fully or at least partially unveiled. Ca2+, Akt/NF-κB, and Ca2+- or pH-dependent Wnt/β-catenin signaling emerge as cross points through which ion channels/transporters interfere with gene expression, modulate cell proliferation, trigger epithelial-to-mesenchymal transition, and promote cell motility and metastasis. Also miRs, lncRNAs, and DNA methylation represent potential links between the misexpression of genes encoding for ion channels/transporters, their malfunctioning, and cancer. The knowledge of all these molecular interactions has provided the basis for therapeutic strategies and approaches, some of which will be broached in this review.
Collapse
|
18
|
Shi C, Yang Q, Pan S, Lin X, Xu G, Luo Y, Zheng B, Xie X, Yu M. LncRNA OIP5-AS1 promotes cell proliferation and migration and induces angiogenesis via regulating miR-3163/VEGFA in hepatocellular carcinoma. Cancer Biol Ther 2020; 21:604-614. [PMID: 32329664 DOI: 10.1080/15384047.2020.1738908] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been reported to play a significant role in the occurrence and progression of tumors. In different tumors, they can either act as an oncogene or tumor suppressor via modulating various target mRNAs. OIP5-AS1 belongs to lncRNA family. It has been reported to be involved in the tumorigenesis of some cancers, such as bladder cancer, gastric cancer, and multiple myeloma. However, the role it plays in hepatocellular carcinoma (HCC) remains unclear. This study aims to explore the inherent mechanism of lncRNA OIP5-AS1 in HCC. In the first place, qRT-PCR found that OIP5-AS1 and VEGFA expressions were significantly increased while miR-3163 was obviously reduced in HCC cells and tissues. Next, a series of functional experiments found that knockdown of OIP5-AS1 suppressed HCC cell proliferation, migration and angiogenesis abilities while promoting cell apoptosis simultaneously. Last but not least, miR-3163 inhibition or VEGFA overexpression can reverse the anti-tumor effect of OIP5-AS1. In summary, OIP5-AS1 affects HCC proliferation, metastasis, and angiogenesis in HCC by regulating VEGFA expression through sponging miR-3163.
Collapse
Affiliation(s)
- Changsheng Shi
- Department of Interventional Therapy, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Qing Yang
- Department of Gastroenterology, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Songsong Pan
- Wenzhou Medical University Wenzhou , Wenzhou, Zhejiang, China
| | - Xingcheng Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Gending Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Ya Luo
- Department of Interventional Therapy, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Bingru Zheng
- Department of Interventional Therapy, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Xiangpang Xie
- Department of Interventional Therapy, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| | - Mingxu Yu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Wenzhou Medical University , Wenzhou, Zhejiang, China
| |
Collapse
|
19
|
Wang Y, Lei X, Gao C, Xue Y, Li X, Wang H, Feng Y. MiR-506-3p suppresses the proliferation of ovarian cancer cells by negatively regulating the expression of MTMR6. J Biosci 2019; 44:126. [PMID: 31894107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
MicroRNAs have been reported to play a crucial role in ovarian cancer (OC) as the most lethal malignancy of the women. Here, we found miR-506-3p was significantly down-regulated in OC tissues compared with corresponding adjacent nontumor tissues. Ectopic miR-506-3p expression inhibited OC cell growth and proliferation using MTT and colony formation assay. Additionally, flow cytometry analysis showed that the overexpression of miR-506-3p induced cell cycle G0/G1 phase arrest and cell apoptosis in OC cells. A luciferase reporter assay confirmed that the myotubularin-related protein 6 (MTMR6) was the target of miR-506-3p. The expression of MTMR6 was increased in OC tissues compared with adjacent tissues using immunohistochemistry. Elevated MTMR6 protein levels were confirmed in OC cells lines compared with immortalized fallopian tube epithelial cell line FTE187 using western blotting. In addition, knockdown of MTMR6 imitated the effects of miR-506-3p on cell proliferation, cell cycle progression and apoptosis in OC cells. Furthermore, rescue experiments using MTMR6 overexpression further verified that MTMR6 was a functional target of miR-506-3p. Our data indicate that miR-506-3p might serve as a tumor suppressor gene and propose a new regulatory mechanism of MTMR6 by miR-506-3p in OC.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Gynaecology, Yan'an University Affiliated Hospital, Yanan, Shaanxi Province, China
| | | | | | | | | | | | | |
Collapse
|
20
|
MiR-506-3p suppresses the proliferation of ovarian cancer cells by negatively regulating the expression of MTMR6. J Biosci 2019. [DOI: 10.1007/s12038-019-9952-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Li QT, Feng YM, Ke ZH, Qiu MJ, He XX, Wang MM, Li YN, Xu J, Shi LL, Xiong ZF. KCNN4 promotes invasion and metastasis through the MAPK/ERK pathway in hepatocellular carcinoma. J Investig Med 2019; 68:68-74. [PMID: 31431469 DOI: 10.1136/jim-2019-001073] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2019] [Indexed: 01/15/2023]
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common malignancies in the world, and is well-known for its bad prognosis. Potassium calcium-activated channel subfamily N member 4 (KCNN4) is a type of intermediate conductance calcium-activated potassium channel, and increasing evidence suggests that KCNN4 contributes to the regulation of invasion and metastasis in a number of cancers. However, its clinical significance and biological function remain unclear in the HCC disease process. In this study, the expression levels of KCNN4 in 86 HCC samples were compared with corresponding paracancerous tissues. sh-RNA was used to reduce the expression of KCNN4 in Hep3B HCC cells in vitro; this was confirmed by Real time-PCR and western blotting. Wound healing, transwell assays and high content analysis were performed to investigate the tumor-promoting characteristics of KCNN4 in Hep3B HCC cells. As results, KCNN4 expression was significantly associated with preoperative serum alpha-fetoprotein level (p=0.038) and TNM stage (p=0.039). Additionally, patients with high KCNN4 amplification in HCC tissue exhibited shorter disease-free survival, whereas there was no statistical significance between KCNN4 amplification and overall survival. Wound healing and transwell assays showed that knockdown of KCNN4 expression could reduce migration and invasion abilities of HCC cells. High content analysis result showed that down-regulated KCNN4 could inhibit the ability of HCC cell proliferation. The mitogen-activated protein kinase (MAPK) pathway is active in cell proliferation, differentiation, migration, senescence, and apoptosis. Matrix metallopeptidase 9 and extracellular signal regulated kinase 1/2 (ERK1/2) were important biomarkers of MAPK/ERK pathway, knockdown of KCNN4 reduced the expression of MMP9 and ERK1/2. These findings showed that KCNN4 promotes HCC invasion and metastasis through the MAPK/ERK pathway.
Collapse
Affiliation(s)
- Qiu-Ting Li
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Ming Feng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zun-Hui Ke
- Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Meng-Jun Qiu
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Xiao He
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng-Meng Wang
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Nan Li
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Xu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Liang-Liang Shi
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Fan Xiong
- Division of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Wang X, Liao X, Huang K, Zeng X, Liu Z, Zhou X, Yu T, Yang C, Yu L, Wang Q, Han C, Zhu G, Ye X, Peng T. Clustered microRNAs hsa-miR-221-3p/hsa-miR-222-3p and their targeted genes might be prognostic predictors for hepatocellular carcinoma. J Cancer 2019; 10:2520-2533. [PMID: 31258758 PMCID: PMC6584338 DOI: 10.7150/jca.29207] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 04/27/2019] [Indexed: 12/16/2022] Open
Abstract
Objective: MicroRNAs (miRNAs) have been explored in malignancies. We investigated the functions of clustered miRNAs hsa-miR-221/222-3p in hepatocellular carcinoma (HCC). Methods: Human miRNA tissue atlas website was determined expression levels in liver tissue. Four databases, TarBase, miRTarBase, miRecords and miRPathDB, were found experimentally validated target genes of clustered miRNAs. TargetScanHuman was predicted target genes. The STRING website was depicted protein-protein interaction (PPI) networks. The OncoLnc website analyzed prognostic values for hsa-miR-221/222-3p and their target genes. The MCODE plugin calculated modules of PPI networks. Receiver operating characteristic (ROC) curves were predicted 1, 3, and 5 years prognostic values. Results: Expression of clustered miRNAs was high in liver tissues. A total of 1577 target genes were identified. Enrichment analysis showed that target genes were enriched mainly in cancer, Wnt signaling and ErbB signaling pathways. Two modules were calculated using PPI networks. Has-miR-221-3p was not associated with prognosis (P = 0.401). Has-miR-222-3p and target genes ESR1, TMED7, CBFB, ETS2, UBE2J1 and UBE2N of the clustered miRNAs were associated with HCC survival (all P < 0.05). Has-miR-222-3p, CBFB, and UBE2N showed good performance of ROC in prognosis prediction at 1, 3, and 5 years (all area under curves > 0.600). Conclusion: Has-miR-222-3p and target genes, especially CBFB, UBE2N, may serve as prognostic predictors for HCC.
Collapse
Affiliation(s)
- Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Ketuan Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xianmin Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Zhengqian Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Tingdong Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Chengkun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Long Yu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan Province, China
| | - Qiaoqi Wang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530000, Guangxi Province, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Xinping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, China
| |
Collapse
|
23
|
Balbaa AO, El-Fattah AA, Awad NM, Abdellatif A. Effects of nanoscale electric fields on the histology of liver cell dysplasia. Nanomedicine (Lond) 2019; 14:515-528. [PMID: 30807249 DOI: 10.2217/nnm-2018-0260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cells electrical fields have a significant role in cell function. AIM The current study examined the effects of nanoscale electric fields generated by magneto-electric nanoparticles (MENs) on precancerous liver tissue. METHODS & RESULTS A total of 30 nm MENs synthesized by sol-gel method were tested in vitro on HepG2 cells and in vivo on liver cell dysplasia in mice, which were exposed to 50 Hz 2 mT for 2 weeks, +/- MENs. MENs with alternating field (AF) reversed liver cells dysplastic features. In vitro cytotoxicity assay showed high lethal dose (LD 50) of 1.4 mg/ml. We also report on the expression of alpha-fetoprotein and cytochrome C. CONCLUSION MEN-generated nanoscale electric fields have significant biological effects on precancerous liver cells.
Collapse
Affiliation(s)
- Aya O Balbaa
- Medical Research Institute, Faculty of Medicine, Ain Shams University, Cairo, Egypt.,Biology Department, School of Sciences & Engineering, American University in Cairo, New Cairo 11835, Egypt
| | - Ahmed Abd El-Fattah
- Department of Materials Science, Institute of Graduate Studies & Research, Alexandria University, Alexandria, Egypt.,Department of Chemistry, College of Science, University of Bahrain
| | - Nahla M Awad
- Early Cancer Detection Unit. Ain Shams University Hospitals, Cairo, Egypt
| | - Ahmed Abdellatif
- Biology Department, School of Sciences & Engineering, American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
24
|
Du Y, Song W, Chen J, Chen H, Xuan Z, Zhao L, Chen J, Jin C, Zhou M, Tuo B, Zhao Y, Zheng S, Song P. The potassium channel KCa3.1 promotes cell proliferation by activating SKP2 and metastasis through the EMT pathway in hepatocellular carcinoma. Int J Cancer 2019; 145:503-516. [PMID: 30628729 DOI: 10.1002/ijc.32121] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 11/17/2018] [Accepted: 12/19/2018] [Indexed: 12/19/2022]
Abstract
The intermediate conductance calcium-activated potassium channel (KCa3.1) plays an important role in maintaining intracellular calcium homeostasis and is involved in the tumorigenesis of many human cancers. However, it is unknown whether KCa3.1 plays a role in the genesis of hepatocellular carcinoma (HCC), one of the most common malignant tumors worldwide with a very poor prognosis. In our study, we found that the expression of KCa3.1 was significantly elevated in poorly differentiated HCC tissues compared to adjacent noncancerous tissues. In vitro and in vivo experiments showed that KCa3.1 could promote cell proliferation, migration, and invasion of HCC. Mechanistically, KCa3.1 promoted cell cycle progression and migration and invasion of HCC cells by activating S-phase protein kinase 2 (SKP2) to trigger the degradation of p21 and p27 and targeting Reelin (RELN) to induce epithelial-mesenchymal transition (EMT), respectively. Taken together, our results demonstrate that KCa3.1 plays an important role in the genesis and progression of HCC, implying that it might be a promising therapeutic target in HCC.
Collapse
Affiliation(s)
- Yehui Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Hao Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Zefeng Xuan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Long Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Jun Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Cheng Jin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Mengqiao Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Yongchao Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
25
|
Cancer-Associated Intermediate Conductance Ca 2+-Activated K⁺ Channel K Ca3.1. Cancers (Basel) 2019; 11:cancers11010109. [PMID: 30658505 PMCID: PMC6357066 DOI: 10.3390/cancers11010109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Several tumor entities have been reported to overexpress KCa3.1 potassium channels due to epigenetic, transcriptional, or post-translational modifications. By modulating membrane potential, cell volume, or Ca2+ signaling, KCa3.1 has been proposed to exert pivotal oncogenic functions in tumorigenesis, malignant progression, metastasis, and therapy resistance. Moreover, KCa3.1 is expressed by tumor-promoting stroma cells such as fibroblasts and the tumor vasculature suggesting a role of KCa3.1 in the adaptation of the tumor microenvironment. Combined, this features KCa3.1 as a candidate target for innovative anti-cancer therapy. However, immune cells also express KCa3.1 thereby contributing to T cell activation. Thus, any strategy targeting KCa3.1 in anti-cancer therapy may also modulate anti-tumor immune activity and/or immunosuppression. The present review article highlights the potential of KCa3.1 as an anti-tumor target providing an overview of the current knowledge on its function in tumor pathogenesis with emphasis on vasculo- and angiogenesis as well as anti-cancer immune responses.
Collapse
|
26
|
Marshall HT, Djamgoz MBA. Immuno-Oncology: Emerging Targets and Combination Therapies. Front Oncol 2018; 8:315. [PMID: 30191140 PMCID: PMC6115503 DOI: 10.3389/fonc.2018.00315] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022] Open
Abstract
Host immunity recognizes and eliminates most early tumor cells, yet immunological checkpoints, exemplified by CTLA-4, PD-1, and PD-L1, pose a significant obstacle to effective antitumor immune responses. T-lymphocyte co-inhibitory pathways influence intensity, inflammation and duration of antitumor immunity. However, tumors and their immunosuppressive microenvironments exploit them to evade immune destruction. Recent PD-1 checkpoint inhibitors yielded unprecedented efficacies and durable responses across advanced-stage melanoma, showcasing potential to replace conventional radiotherapy regimens. Neverthless, many clinical problems remain in terms of efficacy, patient-to-patient variability, and undesirable outcomes and side effects. In this review, we evaluate recent advances in the immuno-oncology field and discuss ways forward. First, we give an overview of current immunotherapy modalities, involving mainy single agents, including inhibitor monoclonal antibodies (mAbs) targeting T-cell checkpoints of PD-1 and CTLA-4. However, neoantigen recognition alone cannot eliminate tumors effectively in vivo given their inherent complex micro-environment, heterogeneous nature and stemness. Then, based mainly upon CTLA-4 and PD-1 checkpoint inhibitors as a "backbone," we cover a range of emerging ("second-generation") therapies incorporating other immunotherapies or non-immune based strategies in synergistic combination. These include targeted therapies such as tyrosine kinase inhibitors, co-stimulatory mAbs, bifunctional agents, epigenetic modulators (such as inhibitors of histone deacetylases or DNA methyltransferase), vaccines, adoptive-T-cell therapy, nanoparticles, oncolytic viruses, and even synthetic "gene circuits." A number of novel immunotherapy co-targets in pre-clinical development are also introduced. The latter include metabolic components, exosomes and ion channels. We discuss in some detail of the personalization of immunotherapy essential for ultimate maximization of clinical outcomes. Finally, we outline possible future technical and conceptual developments including realistic in vitro and in vivo models and inputs from physics, engineering, and artificial intelligence. We conclude that the breadth and quality of immunotherapeutic approaches and the types of cancers that can be treated will increase significantly in the foreseeable future.
Collapse
Affiliation(s)
- Henry T Marshall
- Neuroscience Solutions to Cancer Research Group, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mustafa B A Djamgoz
- Neuroscience Solutions to Cancer Research Group, Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
27
|
Thurber AE, Nelson M, Frost CL, Levin M, Brackenbury WJ, Kaplan DL. IK channel activation increases tumor growth and induces differential behavioral responses in two breast epithelial cell lines. Oncotarget 2017; 8:42382-42397. [PMID: 28415575 PMCID: PMC5522074 DOI: 10.18632/oncotarget.16389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 03/08/2017] [Indexed: 12/26/2022] Open
Abstract
Many potassium channel families are over-expressed in cancer, but their mechanistic role in disease progression is poorly understood. Potassium channels modulate membrane potential (Vmem) and thereby influence calcium ion dynamics and other voltage-sensitive signaling mechanisms, potentially acting as transcriptional regulators. This study investigated the differential response to over-expression and activation of a cancer-associated potassium channel, the intermediate conductance calcium-activated potassium channel (IK), on aggressive behaviors in mammary epithelial and breast cancer cell lines. IK was over-expressed in the highly metastatic breast cancer cell line MDA-MB-231 and the spontaneously immortalized breast epithelial cell line MCF-10A, and the effect on cancer-associated behaviors was assessed. IK over-expression increased primary tumor growth and metastasis of MDA-MB-231 in orthotopic xenografts, demonstrating for the first time in any cancer type that increased IK is sufficient to promote cancer aggression. The primary tumors had similar vascularization as determined by CD31 staining and similar histological characteristics. Interestingly, despite the increased in vivo growth and metastasis, neither IK over-expression nor activation with agonist had a significant effect on MDA-MB-231 proliferation, invasion, or migration in vitro. In contrast, IK decreased MCF-10A proliferation and invasion through Matrigel but had no effect on migration in a scratch-wound assay. We conclude that IK activity is sufficient to promote cell aggression in vivo. Our data provide novel evidence supporting IK and downstream signaling networks as potential targets for cancer therapies.
Collapse
Affiliation(s)
- Amy E. Thurber
- Program in Cell, Molecular, and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Michaela Nelson
- Department of Biology, University of York, Heslington, York, UK
| | | | - Michael Levin
- Biology Department, and Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, Massachusetts, USA
| | | | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| |
Collapse
|
28
|
Song P, Du Y, Song W, Chen H, Xuan Z, Zhao L, Chen J, Chen J, Guo D, Jin C, Zhao Y, Tuo B, Zheng S. KCa3.1 as an Effective Target for Inhibition of Growth and Progression of Intrahepatic Cholangiocarcinoma. J Cancer 2017; 8:1568-1578. [PMID: 28775776 PMCID: PMC5535712 DOI: 10.7150/jca.18697] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/31/2017] [Indexed: 12/15/2022] Open
Abstract
Background: Intrahepatic cholangiocarcinoma (ICC) is a high malignant tumor arising from the bile ducts in the liver with a poor prognosis. As current molecular targeted therapies and systemic chemotherapies had limited success in ICC, novel therapeutic targets are needed. In this study, we attempted to investigate the expression and the role of the intermediate conductance calcium-activated potassium channel (KCa3.1) in ICC. Methods: The expression levels of KCa3.1 channel were measured in 81 resected ICC tumor specimens and the clinicopathological significance of these levels were determined. KCa3.1 channel inhibitor and siRNA were used to study the role of KCa3.1 in proliferation, migration, and invasion of ICC cell lines. The effect of KCa3.1 channel blockade on tumor growth in vivo was also studied using xenograft model in nude mice. Results: The protein expression of KCa3.1 channel was upregulated in ICC tissues and was correlated with age, lymph node metastasis and TNM stage. And high KCa3.1 expression indicated a worse prognosis in ICC patients. Blocking KCa3.1 channel with a specific inhibitor TRAM-34 reduced the proliferation and invasion of ICC cells. Knockdown of KCa3.1 could achieve the same effects through decreasing NF-κB activation. Further in vivo studies demonstrated that KCa3.1 channel blockade suppressed ICC tumor growth. Conclusions: Our observations suggested KCa3.1 might be a promising novel therapeutic target in intrahepatic cholangiocarcinoma.
Collapse
Affiliation(s)
- Penghong Song
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Yehui Du
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Wenfeng Song
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Hao Chen
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Zefeng Xuan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Long Zhao
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Jun Chen
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Jian Chen
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Danjing Guo
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Cheng Jin
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| | - Yongchao Zhao
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi 563003, China
| | - Shusen Zheng
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou 310003, China
| |
Collapse
|
29
|
Liu L, Zhan P, Nie D, Fan L, Lin H, Gao L, Mao X. Intermediate-Conductance-Ca2-Activated K Channel IKCa1 Is Upregulated and Promotes Cell Proliferation in Cervical Cancer. Med Sci Monit Basic Res 2017; 23:45-57. [PMID: 28280257 PMCID: PMC5358865 DOI: 10.12659/msmbr.901462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Accumulating data point to intermediate-conductance calcium-activated potassium channel (IKCa1) as a key player in controlling cell cycle progression and proliferation of human cancer cells. However, the role that IKCa1 plays in the growth of human cervical cancer cells is largely unexplored. MATERIAL AND METHODS In this study, Western blot analysis, immunohistochemical staining, and RT-PCR were first used for IKCa1protein and gene expression assays in cervical cancer tissues and HeLa cells. Then, IKCa1 channel blocker and siRNA were employed to inhibit the functionality of IKCa1 and downregulate gene expression in HeLa cells, respectively. After these treatments, we examined the level of cell proliferation by MTT method and measured IKCa1 currents by conventional whole-cell patch clamp technique. Cell apoptosis was assessed using the Annexin V-FITC/Propidium Iodide (PI) double-staining apoptosis detection kit. RESULTS We demonstrated that IKCa1 mRNA and protein are preferentially expressed in cervical cancer tissues and HeLa cells. We also showed that the IKCa1 channel blocker, clotrimazole, and IKCa1 channel siRNA can be used to suppress cervical cancer cell proliferation and decrease IKCa1 channel current. IKCa1 downregulation by specific siRNAs induced a significant increase in the proportion of apoptotic cells in HeLa cells. CONCLUSIONS IKCa1 is overexpressed in cervical cancer tissues, and IKCa1 upregulation in cervical cancer cell linea enhances cell proliferation, partly by reducing the proportion of apoptotic cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiguang Mao
- Corresponding Authors: Xiguang Mao, e-mail: ; Lanyang Gao, e-mail:
| |
Collapse
|
30
|
Bauer D, Werth F, Nguyen HA, Kiecker F, Eberle J. Critical role of reactive oxygen species (ROS) for synergistic enhancement of apoptosis by vemurafenib and the potassium channel inhibitor TRAM-34 in melanoma cells. Cell Death Dis 2017; 8:e2594. [PMID: 28151482 PMCID: PMC5386497 DOI: 10.1038/cddis.2017.6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/11/2016] [Accepted: 12/27/2016] [Indexed: 12/13/2022]
Abstract
Inhibition of MAP kinase pathways by selective BRAF inhibitors, such as vemurafenib and dabrafenib, have evolved as key therapies of BRAF-mutated melanoma. However, tumor relapse and therapy resistance have remained as major problems, which may be addressed by combination with other pathway inhibitors. Here we identified the potassium channel inhibitor TRAM-34 as highly effective in combination with vemurafenib. Thus apoptosis was significantly enhanced and cell viability was decreased. The combination vemurafenib/TRAM-34 was also effective in vemurafenib-resistant cells, suggesting that acquired resistance may be overcome. Vemurafenib decreased ERK phosphorylation, suppressed antiapoptotic Mcl-1 and enhanced proapoptotic Puma and Bim. The combination resulted in enhancement of proapoptotic pathways as caspase-3 and loss of mitochondrial membrane potential. Indicating a special mechanism of vemurafenib-induced apoptosis, we found strong enhancement of intracellular ROS levels already at 1 h of treatment. The critical role of ROS was demonstrated by the antioxidant vitamin E (α-tocopherol), which decreased intracellular ROS as well as apoptosis. Also caspase activation and loss of mitochondrial membrane potential were suppressed, proving ROS as an upstream effect. Thus ROS represents an initial and independent apoptosis pathway in melanoma cells that is of particular importance for vemurafenib and its combination with TRAM-34.
Collapse
Affiliation(s)
- Daniel Bauer
- Department of Dermatology, Venerology und Allergology, Skin Cancer Center Charité, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Molecular Medicine Master's Program, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Werth
- Department of Dermatology, Venerology und Allergology, Skin Cancer Center Charité, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute for Biochemistry and Biology, Faculty of Science, University of Potsdam, Potsdam, Germany
| | - Ha An Nguyen
- Department of Dermatology, Venerology und Allergology, Skin Cancer Center Charité, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Molecular Medicine Master's Program, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Kiecker
- Department of Dermatology, Venerology und Allergology, Skin Cancer Center Charité, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jürgen Eberle
- Department of Dermatology, Venerology und Allergology, Skin Cancer Center Charité, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
31
|
Muratori L, Petroni G, Antonuzzo L, Boni L, Iorio J, Lastraioli E, Bartoli G, Messerini L, Di Costanzo F, Arcangeli A. hERG1 positivity and Glut-1 negativity identifies high-risk TNM stage I and II colorectal cancer patients, regardless of adjuvant chemotherapy. Onco Targets Ther 2016; 9:6325-6332. [PMID: 27789963 PMCID: PMC5072508 DOI: 10.2147/ott.s114090] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The identification of early-stage colorectal cancer (CRC) with high risk of progression is one major clinical challenge, mainly due to lack of validated biomarkers. The aims of the present study were to analyze the prognostic impact of three molecular markers belonging to the ion channels and transporters family: the ether-à-go-go-related gene 1 (hERG1) and the calcium-activated KCa3.1 potassium channels, as well as the glucose transporter 1 (Glut-1); and to define the impact of adjuvant chemotherapy in conjunction with the abovementioned biomarkers, in a cohort of radically resected stage I-III CRC patients. PATIENTS AND METHODS The expressions of hERG1, KCa3.1, and Glut-1 were tested by immunohistochemistry on 162 surgical samples of nonmetastatic, stage I-III CRC patients. The median follow-up was 32 months. The association between biological markers, clinicopathological features, and survival outcomes was investigated by evaluating both disease-free survival and overall survival. RESULTS Although no prognostic valence emerged for KCa3.1, evidence of a negative impact of hERG1 expression on survival outcomes was provided. On the contrary, Glut-1 expression had a positive impact. According to the results of the multivariate analysis, patients were stratified in four risk groups, based on TNM stage and hERG1/Glut-1 expression. After adjusting for adjuvant therapy, stage I and II, Glut-1-negative, and hERG1-positive patients showed the worst survival experience. CONCLUSION This study strongly indicates that the combination of hERG1 positivity and Glut-1 negativity behaves as a prognostic biomarker in radically resected CRC patients. This combination identifies a group of stage I and II CRC patients with a bad prognosis, even worse than that of stage III patients, regardless of adjuvant therapy accomplishment.
Collapse
Affiliation(s)
- Leonardo Muratori
- Department of Experimental and Clinical Medicine, University of Florence
| | - Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence
| | - Lorenzo Antonuzzo
- Medical Oncology, Azienda Ospedaliero-Universitaria Careggi, Florence; Department of Medical Biotechnologies, University of Siena, Siena
| | - Luca Boni
- Clinical Trials Coordinating Center, Istituto Toscano Tumori, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Jessica Iorio
- Department of Experimental and Clinical Medicine, University of Florence; Department of Medical Biotechnologies, University of Siena, Siena
| | - Elena Lastraioli
- Department of Experimental and Clinical Medicine, University of Florence
| | - Gianluca Bartoli
- Department of Experimental and Clinical Medicine, University of Florence
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence
| | | | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence
| |
Collapse
|
32
|
Sevelsted Møller L, Fialla AD, Schierwagen R, Biagini M, Liedtke C, Laleman W, Klein S, Reul W, Koch Hansen L, Rabjerg M, Singh V, Surra J, Osada J, Reinehr R, de Muckadell OBS, Köhler R, Trebicka J. The calcium-activated potassium channel KCa3.1 is an important modulator of hepatic injury. Sci Rep 2016; 6:28770. [PMID: 27354175 PMCID: PMC4926059 DOI: 10.1038/srep28770] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/10/2016] [Indexed: 12/12/2022] Open
Abstract
The calcium-activated potassium channel KCa3.1 controls different cellular processes such as proliferation and volume homeostasis. We investigated the role of KCa3.1 in experimental and human liver fibrosis. KCa3.1 gene expression was investigated in healthy and injured human and rodent liver. Effect of genetic depletion and pharmacological inhibition of KCa3.1 was evaluated in mice during carbon tetrachloride induced hepatic fibrogenesis. Transcription, protein expression and localisation of KCa3.1 was analysed by reverse transcription polymerase chain reaction, Western blot and immunohistochemistry. Hemodynamic effects of KCa3.1 inhibition were investigated in bile duct-ligated and carbon tetrachloride intoxicated rats. In vitro experiments were performed in rat hepatic stellate cells and hepatocytes. KCa3.1 expression was increased in rodent and human liver fibrosis and was predominantly observed in the hepatocytes. Inhibition of KCa3.1 aggravated liver fibrosis during carbon tetrachloride challenge but did not change hemodynamic parameters in portal hypertensive rats. In vitro, KCa3.1 inhibition leads to increased hepatocyte apoptosis and DNA damage, whereas proliferation of hepatic stellate cells was stimulated by KCa3.1 inhibition. Our data identifies KCa3.1 channels as important modulators in hepatocellular homeostasis. In contrast to previous studies in vitro and other tissues this channel appears to be anti-fibrotic and protective during liver injury.
Collapse
Affiliation(s)
- Linda Sevelsted Møller
- Department of Medical Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark.,Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Annette Dam Fialla
- Department of Medical Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | | | - Matteo Biagini
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Wim Laleman
- Department of Liver and Biliopancreatic disorders, University of Leuven, Leuven, Belgium
| | - Sabine Klein
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Winfried Reul
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Lars Koch Hansen
- Department of Medical Gastroenterology and Hepatology, Vejle Hospital, Vejle, Denmark
| | - Maj Rabjerg
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Vikrant Singh
- Department of Pharmacology, University of California, Davis, California, USA
| | - Joaquin Surra
- Departament de Producción Animal, Escuela Politécnica Superior, Huesca, Spain
| | - Jesus Osada
- Departamento Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón (IIS), Universidad de Zaragoza-CIBEROBN, Zaragoza, Spain
| | - Roland Reinehr
- Elbe-Elster Klinikum, Krankenhaus Herzberg, Herzberg, Germany
| | | | - Ralf Köhler
- Aragon Institute of Health Science I CS, Zaragoza, Spain
| | - Jonel Trebicka
- Department of Medical Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark.,Department of Internal Medicine I, University of Bonn, Bonn, Germany
| |
Collapse
|
33
|
Martial S. Involvement of ion channels and transporters in carcinoma angiogenesis and metastasis. Am J Physiol Cell Physiol 2016; 310:C710-27. [PMID: 26791487 DOI: 10.1152/ajpcell.00218.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Angiogenesis is a finely tuned process, which is the result of the equilibrium between pro- and antiangiogenic factors. In solid tumor angiogenesis, the balance is highly in favor of the production of new, but poorly functional blood vessels, initially intended to provide growing tumors with nutrients and oxygen. Among the numerous proteins involved in tumor development, several types of ion channels are overexpressed in tumor cells, as well as in stromal and endothelial cells. Ion channels thus actively participate in the different hallmarks of cancer, especially in tumor angiogenesis and metastasis. Indeed, from their strategic localization in the plasma membrane, ion channels are key operators of cell signaling, as they sense and respond to environmental changes. This review aims to decipher how ion channels of different families are intricately involved in the fundamental angiogenesis and metastasis hallmarks, which lead from a nascent tumor to systemic dissemination. An overview of the possible use of ion channels as therapeutic targets will also be given, showing that ion channel inhibitors or specific antibodies may provide effective tools, in the near future, in the treatment of carcinomas.
Collapse
Affiliation(s)
- Sonia Martial
- Institut de Recherche sur le Cancer et le Vieillissement, CNRS UMR 7284, Inserm U1081, Université Nice-Sophia Antipolis, Nice, France
| |
Collapse
|
34
|
Xiong G, Wang Y, Ding Q, Yang L. Hsa-mir-1269 genetic variant contributes to hepatocellular carcinoma susceptibility through affecting SOX6. Am J Transl Res 2015; 7:2091-2098. [PMID: 26692953 PMCID: PMC4656786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 07/31/2015] [Indexed: 06/05/2023]
Abstract
MiR-1269 is an essential oncogene that plays crucial roles in regulating the development of hepatocellular carcinoma (HCC). In this study, we mainly focused on the polymorphisms (rs73239138) in miR-1069 to explore its potential role in regulation of target genes in liver cancer. We detected increased level of miR-1269 in 80 HCC patients. SOX6 was predicted as a potential target gene of miR-as. Notably, Pearson correlation analysis indicated that patients harbored with miR-1269 wild type (rs73239138, GG genotype), positively correlated with SOX6 expression. Over-expression of miR-1269 with GG genotype promoted cell proliferation comparing with AA genotype, which is acompanied by a decreased level of SOX6. Further dual luciferase reporter assay showed that miR-1269 with GG genotype have a stronger binding ability with SOX6. SNP rs73239138 in miR-1269 was very likey to be involved in the development of HCC by acting as a protective factor, as the carriers of GA and GG genotype resulted in a smaller tumor size. In conclusion, our results support that SNP rs73239138 in miR-1269 is a protective factor which prevents binding to 3'UTR of SOX6 and there by suppresses tumor growth in HCC.
Collapse
Affiliation(s)
- Guanying Xiong
- The Medical Center for Digestive Diseases of The Second Affiliated Hospital of Nanjing Medical University Nanjing 210011, Jiangsu Province, P.R. China
| | - Yun Wang
- The Medical Center for Digestive Diseases of The Second Affiliated Hospital of Nanjing Medical University Nanjing 210011, Jiangsu Province, P.R. China
| | - Quchen Ding
- The Medical Center for Digestive Diseases of The Second Affiliated Hospital of Nanjing Medical University Nanjing 210011, Jiangsu Province, P.R. China
| | - Lihua Yang
- The Medical Center for Digestive Diseases of The Second Affiliated Hospital of Nanjing Medical University Nanjing 210011, Jiangsu Province, P.R. China
| |
Collapse
|
35
|
Blockade of KCa3.1 potassium channels protects against cisplatin-induced acute kidney injury. Arch Toxicol 2015; 90:2249-2260. [DOI: 10.1007/s00204-015-1607-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/21/2015] [Indexed: 12/31/2022]
|