251
|
Tanwar J, Arora S, Motiani RK. Orai3: Oncochannel with therapeutic potential. Cell Calcium 2020; 90:102247. [PMID: 32659517 DOI: 10.1016/j.ceca.2020.102247] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 01/10/2023]
Abstract
Ion channels in particular Calcium (Ca2+) channels play a critical role in physiology by regulating plethora of cellular processes ranging from cell proliferation, differentiation, transcriptional regulation and programmed cell death. One such physiologically important and highly Ca2+ selective channel family is Orai channels consisting of three homologs Orai1, Orai2 and Orai3. Orai channels are responsible for Ca2+ influx across the plasma membrane in response to decrease in Endoplasmic Reticulum (ER) Ca2+ stores. STIM1/STIM2 proteins sense the reduction in ER Ca2+ levels and activate Orai channels for restoring ER Ca2+ as well as for driving cellular functions. This signaling cascade is known as Store Operated Ca2+ Entry (SOCE). Although Orai1 is the ubiquitous SOCE channel protein, Orai2 and Orai3 mediate SOCE in certain specific tissues. Further, mammalian specific homolog Orai3 forms heteromultimeric channel with Orai1 for constituting Arachidonic acid regulated Ca2+ (ARC) channels or arachidonic acid metabolite Leukotriene C4 (LTC4) regulated Ca2+ (LRC) channels. Literature suggests that Orai3 regulates Breast, Prostate, Lung and Gastrointestinal cancers by either forming Store Operated Ca2+ (SOC) or ARC/LRC channels in the cancerous cells but not in healthy tissue. In this review, we would discuss the role of Orai3 in these cancers and would highlight the potential of therapeutic targeting of Orai3 for better management and treatment of cancer. Finally, we will deliberate on key outstanding questions in the field that demand critical attention and further studies.
Collapse
Affiliation(s)
- Jyoti Tanwar
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India; CSIR-Institute of Genomics and Integrative Biology (IGIB), New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Samriddhi Arora
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India
| | - Rajender K Motiani
- Laboratory of Calciomics and Systemic Pathophysiology, Regional Centre for Biotechnology (RCB), Faridabad, Delhi-NCR, India.
| |
Collapse
|
252
|
Zhai X, Sterea AM, El Hiani Y. Lessons from the Endoplasmic Reticulum Ca 2+ Transporters-A Cancer Connection. Cells 2020; 9:E1536. [PMID: 32599788 PMCID: PMC7349521 DOI: 10.3390/cells9061536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023] Open
Abstract
Ca2+ is an integral mediator of intracellular signaling, impacting almost every aspect of cellular life. The Ca2+-conducting transporters located on the endoplasmic reticulum (ER) membrane shoulder the responsibility of constructing the global Ca2+ signaling landscape. These transporters gate the ER Ca2+ release and uptake, sculpt signaling duration and intensity, and compose the Ca2+ signaling rhythm to accommodate a plethora of biological activities. In this review, we explore the mechanisms of activation and functional regulation of ER Ca2+ transporters in the establishment of Ca2+ homeostasis. We also contextualize the aberrant alterations of these transporters in carcinogenesis, presenting Ca2+-based therapeutic interventions as a means to tackle malignancies.
Collapse
Affiliation(s)
- Xingjian Zhai
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | | | - Yassine El Hiani
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
253
|
Khateb A, Ronai ZA. Unfolded Protein Response in Leukemia: From Basic Understanding to Therapeutic Opportunities. Trends Cancer 2020; 6:960-973. [PMID: 32540455 DOI: 10.1016/j.trecan.2020.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/03/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022]
Abstract
Understanding genetic and epigenetic changes that underlie abnormal proliferation of hematopoietic stem and progenitor cells is critical for development of new approaches to monitor and treat leukemia. The unfolded protein response (UPR) is a conserved adaptive signaling pathway that governs protein folding, secretion, and energy production and serves to maintain protein homeostasis in various cellular compartments. Deregulated UPR signaling, which often occurs in hematopoietic stem cells and leukemia, defines the degree of cellular toxicity and perturbs protein homeostasis, and at the same time, offers a novel therapeutic target. Here, we review current knowledge related to altered UPR signaling in leukemia and highlight possible strategies for exploiting the UPR as treatment for this disease.
Collapse
Affiliation(s)
- Ali Khateb
- Tumor Initiation and Maintenance Program, National Cancer Institute (NCI) Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Ze'ev A Ronai
- Tumor Initiation and Maintenance Program, National Cancer Institute (NCI) Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
254
|
Shen B, Wang S, Bharathi G, Li Y, Lin F, Hu R, Liu L, Qu J. Rapid and Targeted Photoactivation of Ca 2+ Channels Mediated by Squaraine To Regulate Intracellular and Intercellular Signaling Processes. Anal Chem 2020; 92:8497-8505. [PMID: 32438796 DOI: 10.1021/acs.analchem.0c01243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As an important cellular signal transduction messenger, Ca2+ has the capability to regulate cell function and control many biochemical processes, including metabolism, gene expression, and cell survival and death. Here, we introduce an accessible method for the photoactivation of Ca2+ channels mediated by squaraine (SQ) to rapidly induce cellular Ca2+ release and activate signal transduction. With a short preparation time, the maximum Ca2+ concentration increase could reach approximately 450% in 30 s, resulting from marked Ca2+ release channel opening in the endoplasmic reticulum (ER). This release was enhanced by another target location of SQ, that is, the outer mitochondrial-associated membrane where Ca2+ channels accumulate, and by the consequent large amounts of reactive oxygen species resulting from the respiratory chain activity stimulated by Ca2+ load. We used this method to investigate cellular signal transduction in different cancer cells and revealed rapid intracellular Ca2+ flow, unidirectional intercellular signaling processes, and neuronal signaling activity, which demonstrated the potential and convenience of the method for routine Ca2+ research.
Collapse
Affiliation(s)
- Binglin Shen
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Ganapathi Bharathi
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Yanping Li
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Fangrui Lin
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Rui Hu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, No. 3688, Nanhai Avenue, Shenzhen 518060, China
| |
Collapse
|
255
|
Afzal M, Kren BT, Naveed AK, Trembley JH, Ahmed K. Protein kinase CK2 impact on intracellular calcium homeostasis in prostate cancer. Mol Cell Biochem 2020; 470:131-143. [PMID: 32436081 DOI: 10.1007/s11010-020-03752-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/08/2020] [Indexed: 01/06/2023]
Abstract
Protein kinase CK2 plays multiple roles in cell function in normal and disease states. CK2 is elevated in numerous types of cancer cells, and CK2 suppression of apoptosis represents a key link to the cancer cell phenotype. CK2 regulation of cell survival and death involves diverse processes, and our previous work suggested that mitochondrial machinery is a key locus of this function. One of the earliest responses of prostate cells to inhibition of CK2 is a change in mitochondrial membrane potential, possibly associated with Ca2+ signaling. Thus, in the present work, we investigated early impact of CK2 on intracellular Ca2+ dynamics. Three prostate cancer (PCa) cell lines, PC3-LN4, C4-2B, and 22Rv1, were studied. PCa cells were treated with the CK2 small molecule inhibitors 4,5,6,7-tetrabrombenzotriazole and CX-4945 followed by analysis of Ca2+ levels in various cellular compartments over time. The results showed dose-dependent loss in cytosolic Ca2+ levels starting within 2 min and reaching maximal loss within 5-10 min. There was a concomitant increase in Ca2+ in the endoplasmic reticulum (ER) and mitochondrial compartments. The results suggest that inhibition of CK2 activity results in a rapid movement of Ca2+ out of the cytosol and into the ER and mitochondria, which may be among the earliest contributory factors for induction of apoptosis in cells subjected to inhibition of CK2. In cells with death-inducing levels of CK2 inhibition, total cellular Ca2+ levels dropped at 2 h post-treatment. These novel observations represent a potential mechanism underlying regulation of cell survival and death by CK2 activity.
Collapse
Affiliation(s)
- Muhammad Afzal
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Biochemistry, Riphah International University, Islamabad, Pakistan
| | - Betsy T Kren
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - A Khaliq Naveed
- Department of Biochemistry, Riphah International University, Islamabad, Pakistan
| | - Janeen H Trembley
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Khalil Ahmed
- Research Service, Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Urology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
256
|
[Pt(O,O'-acac)(γ-acac)(DMS)]: Alternative Strategies to Overcome Cisplatin-Induced Side Effects and Resistance in T98G Glioma Cells. Cell Mol Neurobiol 2020; 41:563-587. [PMID: 32430779 DOI: 10.1007/s10571-020-00873-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (CDDP) is one of the most effective chemotherapeutic agents, used for the treatment of diverse tumors, including neuroblastoma and glioblastoma. CDDP induces cell death through different apoptotic pathways. Despite its clinical benefits, CDDP causes several side effects and drug resistance.[Pt(O,O'-acac)(γ-acac)(DMS)], namely PtAcacDMS, a new platinum(II) complex containing two acetylacetonate (acac) and a dimethylsulphide (DMS) in the coordination sphere of metal, has been recently synthesized and showed 100 times higher cytotoxicity than CDDP. Additionally, PtAcacDMS was associated to a decreased neurotoxicity in developing rat central nervous system, also displaying great antitumor and antiangiogenic activity both in vivo and in vitro. Thus, based on the knowledge that several chemotherapeutics induce cancer cell death through an aberrant increase in [Ca2+]i, in the present in vitro study we compared CDDP and PtAcacDMS effects on apoptosis and intracellular Ca2+ dynamics in human glioblastoma T98G cells, applying a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, electron microscopy, Western blotting, qRT-PCR, and epifluorescent Ca2+ imaging. The results confirmed that (i) platinum compounds may induce cell death through an aberrant increase in [Ca2+]i and (ii) PtAcacDMS exerted stronger cytotoxic effect than CDDP, associated to a larger increase in resting [Ca2+]i. These findings corroborate the use of PtAcacDMS as a promising approach to improve Pt-based chemotherapy against gliomas, either by inducing a chemosensitization or reducing chemoresistance in cell lineages resilient to CDDP treatment.
Collapse
|
257
|
MCU-induced mitochondrial calcium uptake promotes mitochondrial biogenesis and colorectal cancer growth. Signal Transduct Target Ther 2020; 5:59. [PMID: 32371956 PMCID: PMC7200750 DOI: 10.1038/s41392-020-0155-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/30/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial calcium uniporter (MCU) has an important role in regulating mitochondrial calcium (Ca2+) homeostasis. Dysregulation of mitochondrial Ca2+ homeostasis has been implicated in various cancers. However, it remains unclear whether MCU regulates mitochondrial Ca2+ uptake to promote cell growth in colorectal cancer (CRC). Therefore, in the present study the expression of MCU in CRC tissues and its clinical significance were examined. Following which, the biological function of MCU-mediated mitochondrial Ca2+ uptake in CRC cell growth and the underlying mechanisms were systematically evaluated using in in vitro and in vivo assays, which included western blotting, cell viability and apoptosis assays, as well as xenograft nude mice models. Our results demonstrated that MCU was markedly upregulated in CRC tissues at both the mRNA and protein levels. Upregulated MCU was associated with poor prognosis in patients with CRC. Our data reported that upregulation of MCU enhanced the mitochondrial Ca2+ uptake to promote mitochondrial biogenesis, which in turn facilitated CRC cell growth in vitro and in vivo. In terms of the underlying mechanism, it was identified that MCU-mediated mitochondrial Ca2+ uptake inhibited the phosphorylation of transcription factor A, mitochondrial (TFAM), and thus enhanced its stability to promote mitochondrial biogenesis. Furthermore, our data indicated that increased mitochondrial Ca2+ uptake led to increased mitochondrial production of ROS via the upregulation of mitochondrial biogenesis, which subsequently activated NF-κB signaling to accelerate CRC growth. In conclusion, the results indicated that MCU-induced mitochondrial Ca2+ uptake promotes mitochondrial biogenesis by suppressing phosphorylation of TFAM, thus contributing to CRC cell growth. Our findings reveal a novel mechanism underlying mitochondrial Ca2+-mediated CRC cell growth and may provide a potential pharmacological target for CRC treatment.
Collapse
|
258
|
Grosshans HK, Fischer TT, Steinle JA, Brill AL, Ehrlich BE. Neuronal Calcium Sensor 1 is up-regulated in response to stress to promote cell survival and motility in cancer cells. Mol Oncol 2020; 14:1134-1151. [PMID: 32239615 PMCID: PMC7266285 DOI: 10.1002/1878-0261.12678] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 02/08/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Changes in intracellular calcium (Ca2+) signaling can modulate cellular machinery required for cancer progression. Neuronal calcium sensor 1 (NCS1) is a ubiquitously expressed Ca2+‐binding protein that promotes tumor aggressiveness by enhancing cell survival and metastasis. However, the underlying mechanism by which NCS1 contributes to increased tumor aggressiveness has yet to be identified. In this study, we aimed to determine (a) whether NCS1 expression changes in response to external stimuli, (b) the importance of NCS1 for cell survival and migration, and (c) the cellular mechanism(s) through which NSC1 modulates these outcomes. We found that NCS1 abundance increases under conditions of stress, most prominently after stimulation with the pro‐inflammatory cytokine tumor necrosis factor α, in a manner dependent on nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NFκB). We found that NFκB signaling is activated in human breast cancer tissue, which was accompanied by an increase in NCS1 mRNA expression. Further exploration into the relevance of NCS1 in breast cancer progression showed that knockout of NCS1 (NCS1 KO) caused decreased cell survival and motility, increased baseline intracellular Ca2+ levels, and decreased inositol 1,4,5‐trisphosphate‐mediated Ca2+ responses. Protein kinase B (Akt) activity was decreased in NCS1 KO cells, which could be rescued by buffering intracellular Ca2+. Conversely, Akt activity was increased in cells overexpressing NCS1 (NCS1 OE). We therefore conclude that NCS1 acts as cellular stress response protein up‐regulated by stress‐induced NFκB signaling and that NCS1 influences cell survival and motility through effects on Ca2+ signaling and Akt pathway activation.
Collapse
Affiliation(s)
- Henrike K Grosshans
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Tom T Fischer
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.,Institute of Pharmacology, Heidelberg University, Germany
| | - Julia A Steinle
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Allison L Brill
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
259
|
Zhang J, Chen M, Zhao Y, Xiong H, Sneh T, Fan Y, Wang J, Zhou X, Gong C. Complement and coagulation cascades pathway correlates with chemosensitivity and overall survival in patients with soft tissue sarcoma. Eur J Pharmacol 2020; 879:173121. [PMID: 32339514 DOI: 10.1016/j.ejphar.2020.173121] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Chemotherapy is an indispensable method in treatment of Soft tissue sarcomas (STS), but variability in sensitivity as a result of tumor heterogeneity is a key factor in determining patient outcome. Several studies have investigated the phenomenon of chemotherapy resistance in STS, while its precise complex mechanism is still unknown. This study aims to identify potential biomarkers for predicting the STS chemosensitivity, with the goal of both aiding patient treatment determination in the clinic and providing insight into key parts of the underlying mechanism. Gene expression profiles of 265 patients were obtained from The Cancer Genome Atlas dataset and differentially expressed genes (DEGs) associated with chemosensitivity were identified in groups of varying chemosensitivity, including 177 upregulated and 21 downregulated genes (P < 0.05). Then, DEGs were found to be enriched in complement and coagulation cascades and the osteoclast differentiation pathway. Protein-protein interaction analysis showed 15 genes (52 edges) enriched in the complement and coagulation cascades while 11 genes (28 edges) enriched in the osteoclast differentiation pathway. Notably, all the genes that significantly correlated to disease-free survival (DFS) and overall survival (OS), such as C1QC, C3AR1, C7, CFI and SERPINE1, are enriched in complement and coagulation cascades pathway. The differential expression of these genes was further verified by using the GSE database. Our findings support that C1QC, C3AR1, C7, CFI and SERPINE1 in the complement and coagulation cascade pathway are potential biomarkers for chemotherapy resistance and survival of STS patients.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Maoshan Chen
- Myeloma Research Group, Australian Centre for Blood Diseases (ACBD), Clinical Central School, Monash University, Melbourne, 3004, Australia
| | - Yuanyuan Zhao
- Department of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Tal Sneh
- Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - Yang Fan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Jianhua Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiao Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
260
|
Nam Y, Ryu KD, Jang C, Moon YH, Kim M, Ko D, Chung KS, Gandini MA, Lee KT, Zamponi GW, Lee JY. Synthesis and cytotoxic effects of 2-thio-3,4-dihydroquinazoline derivatives as novel T-type calcium channel blockers. Bioorg Med Chem 2020; 28:115491. [PMID: 32327350 DOI: 10.1016/j.bmc.2020.115491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 10/24/2022]
Abstract
In our previous work, a series of 2-amino-3,4-dihydroquinazoline derivativesusing an electron acceptor group was reported to be potent T-type calcium channel blockers and exhibit strong cytotoxic effects against various cancerous cell lines. To investigate the role of the guanidine moiety in the 2-amino-3,4-dihydroquinazoline scaffold as a pharmacophore for dual biological activity, a new series of 2-thio-3,4-dihydroquniazoline derivatives using an electron donor group at the C2-position was synthesized and evaluated for T-type calcium channel blocking activity and cytotoxic effects against two human cancerous cell lines (lung cancer A549 and colon cancer HCT-116). Among them, compound 6g showed potent inhibition of Cav3.2 currents (83% inhibition) at 10 µM concentrations. The compound also exhibited IC50 values of 5.0 and 6.4 µM against A549 and HCT-116 cell lines, respectively, which are comparable to the parental lead compound KYS05090. These results indicate that the isothiourea moiety similar to the guanidine moiety of 2-amino-3,4-dihydroquinazoline derivatives may be an essential pharmacophore for the desired biological activities. Therefore, our preliminary work can provide the opportunity to expand a chemical repertoire to improve affinity and selectivity for T-type calcium channels.
Collapse
Affiliation(s)
- Yunchan Nam
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ki Deok Ryu
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Changyoung Jang
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoon Hyoung Moon
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Misong Kim
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dohyeong Ko
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sook Chung
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Maria A Gandini
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada.
| | - Jae Yeol Lee
- Research Institute for Basic Sciences and Department of Chemistry, College of Sciences, Kyung Hee University, Seoul 02447, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
261
|
Owusu Obeng E, Rusciano I, Marvi MV, Fazio A, Ratti S, Follo MY, Xian J, Manzoli L, Billi AM, Mongiorgi S, Ramazzotti G, Cocco L. Phosphoinositide-Dependent Signaling in Cancer: A Focus on Phospholipase C Isozymes. Int J Mol Sci 2020; 21:ijms21072581. [PMID: 32276377 PMCID: PMC7177890 DOI: 10.3390/ijms21072581] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
Phosphoinositides (PI) form just a minor portion of the total phospholipid content in cells but are significantly involved in cancer development and progression. In several cancer types, phosphatidylinositol 3,4,5-trisphosphate [PtdIns(3,4,5)P3] and phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] play significant roles in regulating survival, proliferation, invasion, and growth of cancer cells. Phosphoinositide-specific phospholipase C (PLC) catalyze the generation of the essential second messengers diacylglycerol (DAG) and inositol 1,4,5 trisphosphate (InsP3) by hydrolyzing PtdIns(4,5)P2. DAG and InsP3 regulate Protein Kinase C (PKC) activation and the release of calcium ions (Ca2+) into the cytosol, respectively. This event leads to the control of several important biological processes implicated in cancer. PLCs have been extensively studied in cancer but their regulatory roles in the oncogenic process are not fully understood. This review aims to provide up-to-date knowledge on the involvement of PLCs in cancer. We focus specifically on PLCβ, PLCγ, PLCδ, and PLCε isoforms due to the numerous evidence of their involvement in various cancer types.
Collapse
|
262
|
Peng YS, Tang CW, Peng YY, Chang H, Chen CL, Guo SL, Wu LC, Huang MC, Lee HC. Comparative functional genomic analysis of Alzheimer's affected and naturally aging brains. PeerJ 2020; 8:e8682. [PMID: 32219020 PMCID: PMC7087547 DOI: 10.7717/peerj.8682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/04/2020] [Indexed: 11/27/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a prevalent progressive neurodegenerative human disease whose cause remains unclear. Numerous initially highly hopeful anti-AD drugs based on the amyloid-β (Aβ) hypothesis of AD have failed recent late-phase tests. Natural aging (AG) is a high-risk factor for AD. Here, we aim to gain insights in AD that may lead to its novel therapeutic treatment through conducting meta-analyses of gene expression microarray data from AG and AD-affected brain. Methods Five sets of gene expression microarray data from different regions of AD (hereafter, ALZ when referring to data)-affected brain, and one set from AG, were analyzed by means of the application of the methods of differentially expressed genes and differentially co-expressed gene pairs for the identification of putatively disrupted biological pathways and associated abnormal molecular contents. Results Brain-region specificity among ALZ cases and AG-ALZ differences in gene expression and in KEGG pathway disruption were identified. Strong heterogeneity in AD signatures among the five brain regions was observed: HC/PC/SFG showed clear and pronounced AD signatures, MTG moderately so, and EC showed essentially none. There were stark differences between ALZ and AG. OXPHOS and Proteasome were the most disrupted pathways in HC/PC/SFG, while AG showed no OXPHOS disruption and relatively weak Proteasome disruption in AG. Metabolic related pathways including TCA cycle and Pyruvate metabolism were disrupted in ALZ but not in AG. Three pathogenic infection related pathways were disrupted in ALZ. Many cancer and signaling related pathways were shown to be disrupted AG but far less so in ALZ, and not at all in HC. We identified 54 “ALZ-only” differentially expressed genes, all down-regulated and which, when used to augment the gene list of the KEGG AD pathway, made it significantly more AD-specific.
Collapse
Affiliation(s)
- Yi-Shian Peng
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chia-Wei Tang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Yi-Yun Peng
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Hung Chang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chien-Lung Chen
- Department of Nephrology, Landseed Hospital, Taoyuan, Taiwan
| | - Shu-Lin Guo
- Department of Anesthesiology, Cathay General Hospital, Taipei, Taiwan.,Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Li-Ching Wu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Min-Chang Huang
- Department of Physics, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Hoong-Chien Lee
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan.,Department of Physics, Chung Yuan Christian University, Taoyuan, Taiwan
| |
Collapse
|
263
|
Nunn AVW, Guy GW, Botchway SW, Bell JD. From sunscreens to medicines: Can a dissipation hypothesis explain the beneficial aspects of many plant compounds? Phytother Res 2020; 34:1868-1888. [PMID: 32166791 PMCID: PMC7496984 DOI: 10.1002/ptr.6654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 01/16/2020] [Accepted: 02/16/2020] [Indexed: 12/17/2022]
Abstract
Medicine has utilised plant‐based treatments for millennia, but precisely how they work is unclear. One approach is to use a thermodynamic viewpoint that life arose by dissipating geothermal and/or solar potential. Hence, the ability to dissipate energy to maintain homeostasis is a fundamental principle in all life, which can be viewed as an accretion system where layers of complexity have built upon core abiotic molecules. Many of these compounds are chromophoric and are now involved in multiple pathways. Plants have further evolved a plethora of chromophoric compounds that can not only act as sunscreens and redox modifiers, but also have now become integrated into a generalised stress adaptive system. This could be an extension of the dissipative process. In animals, many of these compounds are hormetic, modulating mitochondria and calcium signalling. They can also display anti‐pathogen effects. They could therefore modulate bioenergetics across all life due to the conserved electron transport chain and proton gradient. In this review paper, we focus on well‐described medicinal compounds, such as salicylic acid and cannabidiol and suggest, at least in animals, their activity reflects their evolved function in plants in relation to stress adaptation, which itself evolved to maintain dissipative homeostasis.
Collapse
Affiliation(s)
- Alistair V W Nunn
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| | | | - Stanley W Botchway
- STFC, UKRI & Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, UK
| |
Collapse
|
264
|
Rosa N, Sneyers F, Parys JB, Bultynck G. Type 3 IP 3 receptors: The chameleon in cancer. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 351:101-148. [PMID: 32247578 DOI: 10.1016/bs.ircmb.2020.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), intracellular calcium (Ca2+) release channels, fulfill key functions in cell death and survival processes, whose dysregulation contributes to oncogenesis. This is essentially due to the presence of IP3Rs in microdomains of the endoplasmic reticulum (ER) in close proximity to the mitochondria. As such, IP3Rs enable efficient Ca2+ transfers from the ER to the mitochondria, thus regulating metabolism and cell fate. This review focuses on one of the three IP3R isoforms, the type 3 IP3R (IP3R3), which is linked to proapoptotic ER-mitochondrial Ca2+ transfers. Alterations in IP3R3 expression have been highlighted in numerous cancer types, leading to dysregulations of Ca2+ signaling and cellular functions. However, the outcome of IP3R3-mediated Ca2+ transfers for mitochondrial function is complex with opposing effects on oncogenesis. IP3R3 can either suppress cancer by promoting cell death and cellular senescence or support cancer by driving metabolism, anabolic processes, cell cycle progression, proliferation and invasion. The aim of this review is to provide an overview of IP3R3 dysregulations in cancer and describe how such dysregulations alter critical cellular processes such as proliferation or cell death and survival. Here, we pose that the IP3R3 isoform is not only linked to proapoptotic ER-mitochondrial Ca2+ transfers but might also be involved in prosurvival signaling.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Flore Sneyers
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut (LKI), Leuven, Belgium.
| |
Collapse
|
265
|
Taylor J, Azimi I, Monteith G, Bebawy M. Ca 2+ mediates extracellular vesicle biogenesis through alternate pathways in malignancy. J Extracell Vesicles 2020; 9:1734326. [PMID: 32194926 PMCID: PMC7067202 DOI: 10.1080/20013078.2020.1734326] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 01/30/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) are small membrane vesicles that serve as important intercellular signalling intermediaries in both malignant and non-malignant cells. For EVs formed by the plasma membrane, their biogenesis is characterized by an increase in intracellular calcium followed by successive membrane and cytoskeletal changes. EV-production is significantly higher in malignant cells relative to non-malignant cells and previous work suggests this is dependent on increased calcium mobilization and activity of calpain. However, calcium-signalling pathways involved in malignant and non-malignant EV biogenesis remain unexplored. Here we demonstrate; malignant cells have high basal production of plasma membrane EVs compared to non-malignant cells and this is driven by a calcium–calpain dependent pathway. Resting vesiculation in malignant cells occurs via mobilization of calcium from endoplasmic reticulum (ER) stores rather than from the activity of plasma membrane calcium channels. In the event of ER store depletion however, the store-operated calcium entry (SOCE) pathway is activated to restore ER calcium stores. Depleting both ER calcium stores and blocking SOCE, inhibits EV biogenesis. In contrast, calcium signalling pathways are not activated in resting non-malignant cells. Consequently, these cells are relatively low vesiculators in the resting state. Following cellular activation however, an increase in cytosolic calcium and activation of calpain increase in EV biogenesis. These findings contribute to furthering our understanding of extracellular vesicle biogenesis. As EVs are key mediators in the intercellular transfer of deleterious cancer traits such as cancer multidrug resistance (MDR), understanding the molecular mechanisms governing their biogenesis in cancer is the crucial first step in finding novel therapeutic targets that circumvent EV-mediated MDR.
Collapse
Affiliation(s)
- Jack Taylor
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Australia
| | - Iman Azimi
- Division of Pharmacy, College of Health and Medicine, University of Tasmania, Australia
| | - Gregory Monteith
- School of Pharmacy, The University of Queensland, Brisbane, Australia.,Mater Research, Translational Research Institute, the University of Queensland, Brisbane, Australia.,Translational Research Institute, The University of Queensland, Brisbane, Australia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, The University of Technology Sydney, Australia
| |
Collapse
|
266
|
Asghar MY, Törnquist K. Transient Receptor Potential Canonical (TRPC) Channels as Modulators of Migration and Invasion. Int J Mol Sci 2020; 21:E1739. [PMID: 32138386 PMCID: PMC7084769 DOI: 10.3390/ijms21051739] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Calcium (Ca2+) is perhaps the most versatile signaling molecule in cells. Ca2+ regulates a large number of key events in cells, ranging from gene transcription, motility, and contraction, to energy production and channel gating. To accomplish all these different functions, a multitude of channels, pumps, and transporters are necessary. A group of channels participating in these processes is the transient receptor potential (TRP) family of cation channels. These channels are divided into 29 subfamilies, and are differentially expressed in man, rodents, worms, and flies. One of these subfamilies is the transient receptor potential canonical (TRPC) family of channels. This ion channel family comprises of seven isoforms, labeled TRPC1-7. In man, six functional forms are expressed (TRPC1, TRPC3-7), whereas TRPC2 is a pseudogene; thus, not functionally expressed. In this review, we will describe the importance of the TRPC channels and their interacting molecular partners in the etiology of cancer, particularly in regard to regulating migration and invasion.
Collapse
Affiliation(s)
- Muhammad Yasir Asghar
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland;
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| | - Kid Törnquist
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2U, Tukholmankatu 8, 00290 Helsinki, Finland;
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6A, 20520 Turku, Finland
| |
Collapse
|
267
|
Peng R, Xu L, Wang H, Lyu Y, Wang D, Bi C, Cui C, Fan C, Liu Q, Zhang X, Tan W. DNA-based artificial molecular signaling system that mimics basic elements of reception and response. Nat Commun 2020; 11:978. [PMID: 32080196 PMCID: PMC7033183 DOI: 10.1038/s41467-020-14739-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
In order to maintain tissue homeostasis, cells communicate with the outside environment by receiving molecular signals, transmitting them, and responding accordingly with signaling pathways. Thus, one key challenge in engineering molecular signaling systems involves the design and construction of different modules into a rationally integrated system that mimics the cascade of molecular events. Herein, we rationally design a DNA-based artificial molecular signaling system that uses the confined microenvironment of a giant vesicle, derived from a living cell. This system consists of two main components. First, we build an adenosine triphosphate (ATP)-driven DNA nanogatekeeper. Second, we encapsulate a signaling network in the biomimetic vesicle, consisting of distinct modules, able to sequentially initiate a series of downstream reactions playing the roles of reception, transduction and response. Operationally, in the presence of ATP, nanogatekeeper switches from the closed to open state. The open state then triggers the sequential activation of confined downstream signaling modules.
Collapse
Affiliation(s)
- Ruizi Peng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Liujun Xu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China
| | - Huijing Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China
| | - Yifan Lyu
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Dan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China
| | - Cheng Bi
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China
| | - Cheng Cui
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Foundation for Applied Molecular Evolution, 13709 Progress Boulevard, Alachua, FL, 32615, USA
| | - Chunhai Fan
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiaoling Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China.
| | - Xiaobing Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China.
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, and Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, People's Republic of China.
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, The Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
- Foundation for Applied Molecular Evolution, 13709 Progress Boulevard, Alachua, FL, 32615, USA.
| |
Collapse
|
268
|
So CL, Milevskiy MJG, Monteith GR. Transient receptor potential cation channel subfamily V and breast cancer. J Transl Med 2020; 100:199-206. [PMID: 31822791 DOI: 10.1038/s41374-019-0348-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential cation channel subfamily V (TRPV) channels play important roles in a variety of cellular processes. One example includes the sensory role of TRPV1 that is sensitive to elevated temperatures and acidic environments and is activated by the hot pepper component capsaicin. Another example is the importance of the highly Ca2+ selective channels TRPV5 and TRPV6 in Ca2+ absorption/reabsorption in the intestine and kidney. However, in some cases such as TRPV4 and TRPV6, breast cancer cells appear to overexpress TRPV channels. Moreover, TRPV mediated Ca2+ influx may contribute to enhanced breast cancer cell proliferation and other processes important in tumor progression such as angiogenesis. It appears that the overexpression of some TRPV channels in breast cancer and/or their involvement in breast cancer cell processes, processes important in the tumor microenvironment or pain may make some TRPV channels potential targets for breast cancer therapy. In this review, we provide an overview of TRPV expression in breast cancer subtypes, the roles of TRPV channels in various aspects of breast cancer progression and consider implications for future therapeutic approaches.
Collapse
Affiliation(s)
- Choon Leng So
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Michael J G Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia. .,Mater Research, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia. .,Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
269
|
Ding J, Jin Z, Yang X, Lou J, Shan W, Hu Y, Du Q, Liao Q, Xu J, Xie R. Plasma membrane Ca 2+-permeable channels and sodium/calcium exchangers in tumorigenesis and tumor development of the upper gastrointestinal tract. Cancer Lett 2020; 475:14-21. [PMID: 32004573 DOI: 10.1016/j.canlet.2020.01.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/30/2019] [Accepted: 01/23/2020] [Indexed: 12/17/2022]
Abstract
The upper gastrointestinal (GI) tumors are multifactorial diseases associated with a combination of oncogenes and environmental factors. Currently, surgery, chemotherapy, radiotherapy and immunotherapy are relatively effective treatment options for the patients with these tumors. However, the asymptomatic phenotype of these tumors during the early stages poses as a significant limiting factor to diagnosis and often renders treatments ineffective. Therefore, new early diagnosis and effective therapy for upper GI tumors are urgently needed. Ca2+ is a pivotal intracellular second messenger and plays a crucial role in living cells by regulating several processes from cell division to death. The aberrant Ca2+ homeostasis is related to many human pathological conditions and diseases, including cancer, and thus the changes in the expression and function of plasma membrane Ca2+ permeable channels and sodium/calcium exchangers are frequently described in tumorigenesis and tumor development of the upper GI tract, including voltage-gated Ca2+ channels (VGCC), transient receptor potential (TRP) channels, store-operated channels (SOC) and Na+/Ca2+ exchanger (NCX). This review will summarize the current knowledge about plasma membrane Ca2+ permeable channels and sodium/calcium exchangers in the upper GI tumors and provide a synopsis of recent advancements on the role and involvement of these channels in upper GI tumors as well as a discussion of the possible strategies to target these channels and exchangers for diagnosis and therapy of the upper GI tumors.
Collapse
Affiliation(s)
- JianHong Ding
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Zhe Jin
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Xiaoxu Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Jun Lou
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Weixi Shan
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Yanxia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Qiushi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China.
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, 563003, PR China.
| |
Collapse
|
270
|
Jurj A, Zanoaga O, Braicu C, Lazar V, Tomuleasa C, Irimie A, Berindan-Neagoe I. A Comprehensive Picture of Extracellular Vesicles and Their Contents. Molecular Transfer to Cancer Cells. Cancers (Basel) 2020; 12:cancers12020298. [PMID: 32012717 PMCID: PMC7072213 DOI: 10.3390/cancers12020298] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Critical processes such as growth, invasion, and metastasis of cancer cells are sustained via bidirectional cell-to-cell communication in tissue complex environments. Such communication involves the secretion of soluble factors by stromal cells and/or cancer cells within the tumor microenvironment (TME). Both stromal and cancer cells have been shown to export bilayer nanoparticles: encapsulated regulatory molecules that contribute to cell-to-cell communication. These nanoparticles are known as extracellular vesicles (EVs) being classified into exosomes, microvesicles, and apoptotic bodies. EVs carry a vast repertoire of molecules such as oncoproteins and oncopeptides, DNA fragments from parental to target cells, RNA species (mRNAs, microRNAs, and long non-coding RNA), and lipids, initiating phenotypic changes in TME. According to their specific cargo, EVs have crucial roles in several early and late processes associated with tumor development and metastasis. Emerging evidence suggests that EVs are being investigated for their implication in early cancer detection, monitoring cancer progression and chemotherapeutic response, and more relevant, the development of novel targeted therapeutics. In this study, we provide a comprehensive understanding of the biophysical properties and physiological functions of EVs, their implications in TME, and highlight the applicability of EVs for the development of cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Vladimir Lazar
- Worldwide Innovative Network for Personalized Cancer Therapy, 94800 Villejuif, France;
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
- Department of Hematology, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Alexandru Irimie
- 11th Department of Surgical Oncology and Gynaecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
- Department of Surgery, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
- Correspondence: (A.I.); (I.B.-N.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
- MEDFUTURE—Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
- Correspondence: (A.I.); (I.B.-N.)
| |
Collapse
|
271
|
Plasma Membrane Ca 2+ ATPase Isoform 4 (PMCA4) Has an Important Role in Numerous Hallmarks of Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12010218. [PMID: 31963119 PMCID: PMC7016988 DOI: 10.3390/cancers12010218] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is largely resistant to standard treatments leading to poor patient survival. The expression of plasma membrane calcium ATPase-4 (PMCA4) is reported to modulate key cancer hallmarks including cell migration, growth, and apoptotic resistance. Data-mining revealed that PMCA4 was over-expressed in pancreatic ductal adenocarcinoma (PDAC) tumors which correlated with poor patient survival. Western blot and RT-qPCR revealed that MIA PaCa-2 cells almost exclusively express PMCA4 making these a suitable cellular model of PDAC with poor patient survival. Knockdown of PMCA4 in MIA PaCa-2 cells (using siRNA) reduced cytosolic Ca2+ ([Ca2+]i) clearance, cell migration, and sensitized cells to apoptosis, without affecting cell growth. Knocking down PMCA4 had minimal effects on numerous metabolic parameters (as assessed using the Seahorse XF analyzer). In summary, this study provides the first evidence that PMCA4 is over-expressed in PDAC and plays a role in cell migration and apoptotic resistance in MIA PaCa-2 cells. This suggests that PMCA4 may offer an attractive novel therapeutic target in PDAC.
Collapse
|
272
|
Fiorio Pla A, Gkika D. Ca2+ Channel Toolkit in Neuroendocrine Tumors. Neuroendocrinology 2020; 110:147-154. [PMID: 31177261 DOI: 10.1159/000501397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/06/2019] [Indexed: 11/19/2022]
Abstract
Neuroendocrine tumors (NET) constitute a heterogeneous group of malignancies with various clinical presentations and growth rates but a common origin in neuroendocrine cells located all over the body. NET are a relatively low-frequency disease mostly represented by gastroenteropancreatic (GEP) and bronchopulmonary tumors (pNET); on the other hand, an increasing frequency and prevalence have been associated with NET. Despite great efforts in recent years, the management of NET is still a critical unmet need due to the lack of knowledge of the biology of the disease, the lack of adequate biomarkers, late presentation, the relative insensitivity of imaging modalities, and a paucity of predictably effective treatment options. In this context Ca2+ signals, being pivotal molecular devices in sensing and integrating signals from the microenvironment, are emerging to be particularly relevant in cancer, where they mediate interactions between tumor cells and the tumor microenvironment to drive different aspects of neoplastic progression (e.g., cell proliferation and survival, cell invasiveness, and proangiogenetic programs). Indeed, ion channels represent good potential pharmacological targets due to their location on the plasma membrane, where they can be easily accessed by drugs. The present review aims to provide a critical and up-to-date overview of NET development integrating Ca2+ signal involvement. In this perspective, we first give an introduction to NET and Ca2+ channels and then describe the different families of Ca2+ channels implicated in NET, i.e., ionotropic receptors, voltage-dependent Ca2+ channels, and transient receptor potential channels, as well as intracellular Ca2+ channels and their signaling molecules.
Collapse
Affiliation(s)
- Alessandra Fiorio Pla
- Department of Life Science and Systems Biology, University of Torino, Turin, Italy,
- Inserm, U1003 - PHYCEL (Physiologie Cellulaire), Université de Lille, Lille, France,
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France,
| | - Dimitra Gkika
- Inserm, U1003 - PHYCEL (Physiologie Cellulaire), Université de Lille, Lille, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| |
Collapse
|
273
|
Elingaard-Larsen LO, Rolver MG, Sørensen EE, Pedersen SF. How Reciprocal Interactions Between the Tumor Microenvironment and Ion Transport Proteins Drive Cancer Progression. Rev Physiol Biochem Pharmacol 2020; 182:1-38. [PMID: 32737753 DOI: 10.1007/112_2020_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Solid tumors comprise two major components: the cancer cells and the tumor stroma. The stroma is a mixture of cellular and acellular components including fibroblasts, mesenchymal and cancer stem cells, endothelial cells, immune cells, extracellular matrix, and tumor interstitial fluid. The insufficient tumor perfusion and the highly proliferative state and dysregulated metabolism of the cancer cells collectively create a physicochemical microenvironment characterized by altered nutrient concentrations and varying degrees of hypoxia and acidosis. Furthermore, both cancer and stromal cells secrete numerous growth factors, cytokines, and extracellular matrix proteins which further shape the tumor microenvironment (TME), favoring cancer progression.Transport proteins expressed by cancer and stromal cells localize at the interface between the cells and the TME and are in a reciprocal relationship with it, as both sensors and modulators of TME properties. It has been amply demonstrated how acid-base and nutrient transporters of cancer cells enable their growth, presumably by contributing both to the extracellular acidosis and the exchange of metabolic substrates and waste products between cells and TME. However, the TME also impacts other transport proteins important for cancer progression, such as multidrug resistance proteins. In this review, we summarize current knowledge of the cellular and acellular components of solid tumors and their interrelationship with key ion transport proteins. We focus in particular on acid-base transport proteins with known or proposed roles in cancer development, and we discuss their relevance for novel therapeutic strategies.
Collapse
Affiliation(s)
- Line O Elingaard-Larsen
- Translational Type 2 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Michala G Rolver
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ester E Sørensen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stine F Pedersen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
274
|
Calcium Signaling in Endothelial Colony Forming Cells in Health and Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:1013-1030. [PMID: 31646543 DOI: 10.1007/978-3-030-12457-1_40] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Endothelial colony forming cells (ECFCs) represent the only known truly endothelial precursors. ECFCs are released in peripheral circulation to restore the vascular networks dismantled by an ischemic insult or to sustain the early phases of the angiogenic switch in solid tumors. A growing number of studies demonstrated that intracellular Ca2+ signaling plays a crucial role in driving ECFC proliferation, migration, homing and neovessel formation. For instance, vascular endothelial growth factor (VEGF) triggers intracellular Ca2+ oscillations and stimulates angiogenesis in healthy ECFCs, whereas stromal derived factor-1α promotes ECFC migration through a biphasic Ca2+ signal. The Ca2+ toolkit endowed to circulating ECFCs is extremely plastic and shows striking differences depending on the physiological background of the donor. For instance, inositol-1,4,5-trisphosphate-induced Ca2+ release from the endoplasmic reticulum is downregulated in tumor-derived ECFCs, while agonists-induced store-operated Ca2+ entry is up-regulated in renal cellular carcinoma and is unaltered in breast cancer and reduced in infantile hemangioma. This remodeling of the Ca2+ toolkit prevents VEGF-induced pro-angiogenic Ca2+ oscillations in tumor-derived ECFCs. An emerging theme of research is the dysregulation of the Ca2+ toolkit in primary myelofibrosis-derived ECFCs, as this myeloproliferative disorder may depend on a driver mutation in the calreticulin gene. In this chapter, I provide a comprehensive, but succinct, description on the architecture and role of the intracellular Ca2+ signaling toolkit in ECFCs derived from umbilical cord blood and from peripheral blood of healthy donors, cancer patients and subjects affected by primary myelofibrosis.
Collapse
|
275
|
Wu T, Cao H, Liu L, Peng K. Identification of Key Genes and Pathways for Enchondromas by Bioinformatics Analysis. Dose Response 2020; 18:1559325820907536. [PMID: 32284694 PMCID: PMC7137642 DOI: 10.1177/1559325820907536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/01/2020] [Accepted: 01/16/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The risk of malignant transformation of enchondromas (EC) toward central chondrosarcoma is increased up to 35%, while the exact etiology of EC is unknown. The purpose of this research was to authenticate gene signatures during EC and reveal their potential mechanisms in occurrence and development of EC. METHODS The gene expression profiles was acquired from Gene Expression Omnibus database (no. GSE22855). The gene ontology (GO), protein-protein interaction (PPI) network and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses were utilized to identify differentially expressed genes (DEGs). RESULTS Finally, 242 DEGs were appraisal, containing 200 overregulated genes and 42 downregulated genes. The outcomes of GO analysis indicated that upregulated DEGs were mainly enriched in several biological processes containing response to hypoxia, calcium ion, and negative regulation extrinsic apoptotic signaling pathway. Furthermore, the upregulated DEGs were enriched in extracellular matrix (ECM)-receptor interaction, protein processing in endoplasmic reticulum and ribosome, which was analyzed by KEGG pathway. From the PPI network, the top 10 hub genes were identified, which were related to significant pathways containing ribosome, protein processing in endoplasmic reticulum, and ECM-receptor interaction. CONCLUSION In conclusion, the present study may be helpful for understanding the diagnostic biomarkers of EC.
Collapse
Affiliation(s)
- Tianlong Wu
- Department of Sports Medicine, The First Bethune hospital of Jilin University, Changchun, Jilin, China
| | - Honghai Cao
- The Chinese PLA General Hospital, Beijing, China
| | - Lei Liu
- Department of Pain, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Kan Peng
- Department of Joint Surgery, Xi’an Hong Hui Hospital, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
276
|
Bai Y, Qiao L, Xie N, Li Y, Nie Y, Pan Y, Shi Y, Wang J, Liu N. TOB1 suppresses proliferation in K-Ras wild-type pancreatic cancer. Cancer Med 2019; 9:1503-1514. [PMID: 31891232 PMCID: PMC7013073 DOI: 10.1002/cam4.2756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/25/2022] Open
Abstract
TOB1 participates in various kinds of cancers. However, its role in pancreatic cancer has rarely been reported. In this study, we explored the expression and mechanisms of TOB1 in regulating the malignant phenotype of pancreatic cancer cells. TOB1 expression was determined by data mining and immunohistochemistry (IHC), and its localization was observed by immunofluorescence. CCK‐8 cell proliferation, colony formation, flow cytometric, transwell migration, and Western blot (WB) assays were used to examine how it impacts the malignant phenotype of pancreatic cancer. Furthermore, Foxa2 binding to TOB1 was tested by dual‐luciferase reporter assays, and RNA‐Seq was performed to identify signaling pathways. We found TOB1 was downregulated in pancreatic cancer tissues and was mainly located in the cytoplasm. TOB1 overexpression reduced the proliferation of K‐Ras wild‐type pancreatic cancer cells but made no difference to cell migration and invasion. Foxa2 overexpression significantly enhanced TOB1 promoter activity. Moreover, overexpressing TOB1 substantially enriched the calcium pathway in K‐Ras wild‐type pancreatic cancer cells. In conclusion, TOB1 may suppress the proliferation of K‐Ras wild‐type pancreatic cancer cells by regulating calcium pathway genes.
Collapse
Affiliation(s)
- Yuru Bai
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Geriatric Respiratory and Endocrinology (The Third Unit of Cadre's Ward), the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lu Qiao
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ning Xie
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yan Li
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology, the Fourth Military Medical University, Xi'an, Shaanxi, China.,Xijing Hospital of Digestive Diseases, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yan Pan
- State Key Laboratory of Cancer Biology, the Fourth Military Medical University, Xi'an, Shaanxi, China.,Xijing Hospital of Digestive Diseases, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yupeng Shi
- State Key Laboratory of Cancer Biology, the Fourth Military Medical University, Xi'an, Shaanxi, China.,Xijing Hospital of Digestive Diseases, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinhai Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Na Liu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Shaanxi Key Laboratory of Gastrointestinal Motility Disorders, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
277
|
Chen SL, Liu LL, Wang CH, Lu SX, Yang X, He YF, Zhang CZ, Yun JP. Loss of RDM1 enhances hepatocellular carcinoma progression via p53 and Ras/Raf/ERK pathways. Mol Oncol 2019; 14:373-386. [PMID: 31670863 PMCID: PMC6998392 DOI: 10.1002/1878-0261.12593] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/20/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC), with its ineffective therapeutic options and poor prognosis, represents a global threat. In the present study, we show that RAD52 motif 1 (RDM1), a key regulator of DNA double‐strand break repair and recombination, is downregulated in HCC tissues and suppresses tumor growth. In clinical HCC samples, low expression of RDM1 correlates with larger tumor size, poor tumor differentiation, and unfavorable survival. In vitro and in vivo data demonstrate that knockdown of RDM1 increases HCC cell proliferation, colony formation, and cell population at G2/M phase, whereas RDM1 overexpression results in the opposite phenotypes. Mechanistically, RDM1 binds to the tumor suppressor p53 and enhances its protein stability. In the presence of p53, RDM1 suppresses the phosphorylation of Raf and ERK. Overexpression of p53 or treatment with ERK inhibitor significantly abolishes cell proliferation induced by the depletion of RDM1. In addition, overexpression of methyltransferase‐like 3 markedly induces N6‐methyladenosine modification of RDM1 mRNA and represses its expression. Taken together, our study indicates that RDM1 functions as a tumor suppressor and may be a potential prognostic and therapeutic factor for HCC.
Collapse
Affiliation(s)
- Shi-Lu Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Li Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chun-Hua Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shi-Xun Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yang-Fan He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chris Zhiyi Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jing-Ping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
278
|
McGrath JS, Honrado C, Moore JH, Adair SJ, Varhue WB, Salahi A, Farmehini V, Goudreau BJ, Nagdas S, Blais EM, Bauer TW, Swami NS. Electrophysiology-based stratification of pancreatic tumorigenicity by label-free single-cell impedance cytometry. Anal Chim Acta 2019; 1101:90-98. [PMID: 32029124 DOI: 10.1016/j.aca.2019.12.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/13/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer lacking specific biomarkers that can be correlated to disease onset, promotion and progression. To assess whether tumor cell electrophysiology may serve as a marker for PDAC tumorigenicity, we use multi-frequency impedance cytometry at high throughput (∼350 cells/s) to measure the electrical phenotype of single PDAC tumor cells from xenografts, which are derived from primary pancreatic tumors versus those from liver metastases of different patients. A novel phase contrast metric based on variations in the high and low frequency impedance phase responses that is related to electrophysiology of the cell interior is found to be systematically altered as a function of tumorigenicity. PDAC cells of higher tumorigenicity exhibited lowered interior conductivity and enhanced permittivity, which is validated by the dielectrophoresis on the respective cell types. Using genetic analysis, we suggest the role of dysregulated Na+ transport and removal of Ca2+ ions from the cytoplasm on key oncogenic KRAS-driven processes that may be responsible for lowering of the interior cell conductivity. We envision that impedance cytometry can serve as a tool to quantify phenotypic heterogeneity for rapidly stratifying tumorigenicity. It can also aid in protocols for dielectrophoretic isolation of cells with a particular phenotype for prognostic studies on patient survival and to tailor therapy selection to specific patients.
Collapse
Affiliation(s)
- J S McGrath
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - C Honrado
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - J H Moore
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - S J Adair
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - W B Varhue
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - A Salahi
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - V Farmehini
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA
| | - B J Goudreau
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - S Nagdas
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - E M Blais
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - T W Bauer
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - N S Swami
- School of Engineering and Applied Sciences, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
279
|
Moreno-Felici J, Hyroššová P, Aragó M, Rodríguez-Arévalo S, García-Rovés PM, Escolano C, Perales JC. Phosphoenolpyruvate from Glycolysis and PEPCK Regulate Cancer Cell Fate by Altering Cytosolic Ca 2. Cells 2019; 9:E18. [PMID: 31861674 PMCID: PMC7017135 DOI: 10.3390/cells9010018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Changes in phosphoenolpyruvate (PEP) concentrations secondary to variations in glucose availability can regulate calcium signaling in T cells as this metabolite potently inhibits the sarcoplasmic reticulum Ca2+/ATPase pump (SERCA). This regulation is critical to assert immune activation in the tumor as T cells and cancer cells compete for available nutrients. We examined here whether cytosolic calcium and the activation of downstream effector pathways important for tumor biology are influenced by the presence of glucose and/or cataplerosis through the phosphoenolpyruvate carboxykinase (PEPCK) pathway, as both are hypothesized to feed the PEP pool. Our data demonstrate that cellular PEP parallels extracellular glucose in two human colon carcinoma cell lines, HCT-116 and SW480. PEP correlated with cytosolic calcium and NFAT activity, together with transcriptional up-regulation of canonical targets PTGS2 and IL6 that was fully prevented by CsA pre-treatment. Similarly, loading the metabolite directly into the cell increased cytosolic calcium and NFAT activity. PEP-stirred cytosolic calcium was also responsible for the calmodulin (CaM) dependent phosphorylation of c-Myc at Ser62, resulting in increased activity, probably through enhanced stabilization of the protein. Protein expression of several c-Myc targets also correlated with PEP levels. Finally, the participation of PEPCK in this axis was interrogated as it should directly contribute to PEP through cataplerosis from TCA cycle intermediates, especially in glucose starvation conditions. Inhibition of PEPCK activity showed the expected regulation of PEP and calcium levels and consequential downstream modulation of NFAT and c-Myc activities. Collectively, these results suggest that glucose and PEPCK can regulate NFAT and c-Myc activities through their influence on the PEP/Ca2+ axis, advancing a role for PEP as a second messenger communicating metabolism, calcium cell signaling, and tumor biology.
Collapse
Affiliation(s)
- Juan Moreno-Felici
- Department of Physiological Sciences, School of Medicine, University of Barcelona, Feixa Llarga s/n, 08907 L’Hospitalet del Llobregat, Spain; (J.M.-F.); (P.H.); (M.A.); (P.M.G.-R.)
| | - Petra Hyroššová
- Department of Physiological Sciences, School of Medicine, University of Barcelona, Feixa Llarga s/n, 08907 L’Hospitalet del Llobregat, Spain; (J.M.-F.); (P.H.); (M.A.); (P.M.G.-R.)
| | - Marc Aragó
- Department of Physiological Sciences, School of Medicine, University of Barcelona, Feixa Llarga s/n, 08907 L’Hospitalet del Llobregat, Spain; (J.M.-F.); (P.H.); (M.A.); (P.M.G.-R.)
| | - Sergio Rodríguez-Arévalo
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain; (S.R.-A.); (C.E.)
| | - Pablo M. García-Rovés
- Department of Physiological Sciences, School of Medicine, University of Barcelona, Feixa Llarga s/n, 08907 L’Hospitalet del Llobregat, Spain; (J.M.-F.); (P.H.); (M.A.); (P.M.G.-R.)
| | - Carmen Escolano
- Laboratory of Medicinal Chemistry (Associated Unit to CSIC), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain; (S.R.-A.); (C.E.)
| | - Jose C. Perales
- Department of Physiological Sciences, School of Medicine, University of Barcelona, Feixa Llarga s/n, 08907 L’Hospitalet del Llobregat, Spain; (J.M.-F.); (P.H.); (M.A.); (P.M.G.-R.)
- IDIBELL, Gran Via de l’Hospitalet 199, 08908 L’Hospitalet de Llobregat, Spain
| |
Collapse
|
280
|
Matsumoto K, Deguchi A, Motoyoshi A, Morita A, Maebashi U, Nakamoto T, Kawanishi S, Sueyoshi M, Nishimura K, Takata K, Tominaga M, Nakahara T, Kato S. Role of transient receptor potential vanilloid subtype 4 in the regulation of azoymethane/dextran sulphate sodium-induced colitis-associated cancer in mice. Eur J Pharmacol 2019; 867:172853. [PMID: 31836532 DOI: 10.1016/j.ejphar.2019.172853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023]
Abstract
Ca2+-permeable ion channels, such as transient receptor channels, are one of the potential therapeutic targets in cancer. Transient receptor potential vanilloid subtype 4 (TRPV4) is a nonselective cation channel associated with cancer progression. This study investigates the roles of TRPV4 in the pathogenesis of colitis-associated cancer (CAC) in mice. The role of TRPV4 was examined in azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced murine CAC model. The formation of colon tumours induced by AOM/DSS treatment was significantly attenuated in TRPV4-deficient mice (TRPV4KO). TRPV4 was co-localised with markers of angiogenesis and macrophages. AOM/DSS treatment upregulated the expression of CD105, vascular endothelial growth factor receptor 2, and TRPV4 in wildtype, but the upregulation of CD105 was significantly attenuated in TRPV4KO. Bone marrow chimera experiments indicated that TRPV4, expressed in both vascular endothelial cells and bone marrow-derived macrophages, played a significant role in colitis-associated tumorigenesis. There was no significant difference in the population of hematopoietic cells, neutrophils, and monocytes between untreated and AOM/DSS-treated WT and TRPV4KO on flow cytometric analysis. TRPV4 activation by a selective agonist induced TNF-α and CXCL2 release in macrophages. Furthermore, TRPV4 activation enhanced the proliferation of human umbilical vein endothelial cells. These results suggest that TRPV4 expressed in neovascular endothelial cells and bone marrow-derived macrophages contributes to the progression of CAC in mice.
Collapse
Affiliation(s)
- Kenjiro Matsumoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan.
| | - Ayuka Deguchi
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Aoi Motoyoshi
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Akane Morita
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo, 1070072, Japan
| | - Urara Maebashi
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Tomohiro Nakamoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Shohei Kawanishi
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan; Division of Biological Sciences, Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Mari Sueyoshi
- Division of Biological Sciences, Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Kaneyasu Nishimura
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Kazuyuki Takata
- Division of Integrated Pharmaceutical Sciences, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan; Division of Biological Sciences, Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, National Institute for Physiological Sciences, Okazaki, 4440864, Japan; Thermal Biology Group, Exploratory Research Center on Life and Living Systems, Okazaki, 4440864, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo, 1070072, Japan
| | - Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University, Kyoto, 6078414, Japan
| |
Collapse
|
281
|
Brossa A, Buono L, Fallo S, Fiorio Pla A, Munaron L, Bussolati B. Alternative Strategies to Inhibit Tumor Vascularization. Int J Mol Sci 2019; 20:E6180. [PMID: 31817884 PMCID: PMC6940973 DOI: 10.3390/ijms20246180] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells present in tumors show different origin, phenotype, and genotype with respect to the normal counterpart. Various mechanisms of intra-tumor vasculogenesis sustain the complexity of tumor vasculature, which can be further modified by signals deriving from the tumor microenvironment. As a result, resistance to anti-VEGF therapy and activation of compensatory pathways remain a challenge in the treatment of cancer patients, revealing the need to explore alternative strategies to the classical anti-angiogenic drugs. In this review, we will describe some alternative strategies to inhibit tumor vascularization, including targeting of antigens and signaling pathways overexpressed by tumor endothelial cells, the development of endothelial vaccinations, and the use of extracellular vesicles. In addition, anti-angiogenic drugs with normalizing effects on tumor vessels will be discussed. Finally, we will present the concept of endothelial demesenchymalization as an alternative approach to restore normal endothelial cell phenotype.
Collapse
Affiliation(s)
- Alessia Brossa
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Lola Buono
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Sofia Fallo
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| | - Alessandra Fiorio Pla
- Department of Life Science and Systems Biology, University of Torino, 10126 Torino, Italy; (A.F.P.); (L.M.)
| | - Luca Munaron
- Department of Life Science and Systems Biology, University of Torino, 10126 Torino, Italy; (A.F.P.); (L.M.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, Universitty of Torino, 10126 Torino, Italy; (A.B.); (L.B.); (S.F.)
| |
Collapse
|
282
|
Yang H, Li W, Lv Y, Fan Q, Mao X, Long T, Xie L, Dong C, Yang R, Zhang H. Exploring the mechanism of clear cell renal cell carcinoma metastasis and key genes based on multi-tool joint analysis. Gene 2019; 720:144103. [DOI: 10.1016/j.gene.2019.144103] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/16/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022]
|
283
|
Affiliation(s)
- Mathieu Gautier
- Laboratoire de Physiologie Cellulaire et Moléculaire - EA4667, UFR Sciences, Université de Picardie Jules Verne (UPJV), F-80039, Amiens, France.
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology and Penn State Cancer Institute (Mechanisms of Carcinogenesis), Penn State University College of Medicine, H166, 500 University Drive, Hershey, PA, 17033, USA.
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center, Honolulu, HI 96813, USA; University of Hawaii Cancer Center and John A. Burns School of Medicine, Honolulu, HI 96813, USA
| | - Christophe Vandier
- Nutrition-Growth and Cancer-INSERM UMR 1069, Université de Tours, F-37000, Tours, France
| | - Halima Ouadid-Ahidouch
- Laboratoire de Physiologie Cellulaire et Moléculaire - EA4667, UFR Sciences, Université de Picardie Jules Verne (UPJV), F-80039, Amiens, France
| |
Collapse
|
284
|
Leslie TK, James AD, Zaccagna F, Grist JT, Deen S, Kennerley A, Riemer F, Kaggie JD, Gallagher FA, Gilbert FJ, Brackenbury WJ. Sodium homeostasis in the tumour microenvironment. Biochim Biophys Acta Rev Cancer 2019; 1872:188304. [PMID: 31348974 PMCID: PMC7115894 DOI: 10.1016/j.bbcan.2019.07.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/11/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
The concentration of sodium ions (Na+) is raised in solid tumours and can be measured at the cellular, tissue and patient levels. At the cellular level, the Na+ gradient across the membrane powers the transport of H+ ions and essential nutrients for normal activity. The maintenance of the Na+ gradient requires a large proportion of the cell's ATP. Na+ is a major contributor to the osmolarity of the tumour microenvironment, which affects cell volume and metabolism as well as immune function. Here, we review evidence indicating that Na+ handling is altered in tumours, explore our current understanding of the mechanisms that may underlie these alterations and consider the potential consequences for cancer progression. Dysregulated Na+ balance in tumours may open opportunities for new imaging biomarkers and re-purposing of drugs for treatment.
Collapse
Affiliation(s)
- Theresa K Leslie
- Department of Biology, University of York, Heslington, York YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Andrew D James
- Department of Biology, University of York, Heslington, York YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK
| | - Fulvio Zaccagna
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - James T Grist
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Surrin Deen
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Aneurin Kennerley
- York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK; Department of Chemistry, University of York, Heslington, York YO10 5DD, UK
| | - Frank Riemer
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Joshua D Kaggie
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Fiona J Gilbert
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - William J Brackenbury
- Department of Biology, University of York, Heslington, York YO10 5DD, UK; York Biomedical Research Institute, University of York, Heslington, York YO10 5DD, UK.
| |
Collapse
|
285
|
Assessment of doxorubicin-induced remodeling of Ca 2+ signaling and associated Ca 2+ regulating proteins in MDA-MB-231 breast cancer cells. Biochem Biophys Res Commun 2019; 522:532-538. [PMID: 31780263 DOI: 10.1016/j.bbrc.2019.11.136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/20/2019] [Indexed: 12/22/2022]
Abstract
Triple-negative breast cancers (TNBC) are often associated with high relapse rates, despite treatment with chemotherapy agents such as doxorubicin. A better understanding of the signaling and molecular changes associated with doxorubicin may provide novel insights into strategies to enhance treatment efficacy. Calcium signaling is involved in many pathways influencing the efficacy of chemotherapy agents such as proliferation and cell death. However, there are a limited number of studies exploring the effect of doxorubicin on calcium signaling in TNBC. In this study, MDA-MB-231 triple-negative, basal breast cancer cells stably expressing the genetically-encoded calcium indicator GCaMP6m (GCaMP6m-MDA-MB-231) were used to define alterations in calcium signaling. The effects of doxorubicin in GCaMP6m-MDA-MB-231 cells were determined using live cell imaging and fluorescence microscopy. Changes in mRNA levels of specific calcium regulating proteins as a result of doxorubicin treatment were also assessed using real time qPCR. Doxorubicin (1 μM) produced alterations in intracellular calcium signaling, including enhancing the sensitivity of MDA-MB-231 cells to ATP stimulation and prolonging the recovery time after store-operated calcium entry. Upregulation in mRNA levels of ORAI1, TRPC1, SERCA1, IP3R2 and PMCA2 with doxorubicin 1 μM treatment was also observed. Doxorubicin treatment is associated with specific remodeling in calcium signaling in MDA-MB-231 cells, with associated changes in mRNA levels of specific calcium-regulating proteins.
Collapse
|
286
|
A Novel Calcium-Mediated EMT Pathway Controlled by Lipids: An Opportunity for Prostate Cancer Adjuvant Therapy. Cancers (Basel) 2019; 11:cancers11111814. [PMID: 31752242 PMCID: PMC6896176 DOI: 10.3390/cancers11111814] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 01/26/2023] Open
Abstract
The composition of periprostatic adipose tissue (PPAT) has been shown to play a role in prostate cancer (PCa) progression. We recently reported an inverse association between PCa aggressiveness and elevated PPAT linoleic acid (LA) and eicosapentaenoic acid (EPA) content. In the present study, we identified a new signaling pathway with a positive feedback loop between the epithelial-to-mesenchymal transition (EMT) transcription factor Zeb1 and the Ca2+-activated K+ channel SK3, which leads to an amplification of Ca2+ entry and cellular migration. Using in vitro experiments and ex vivo cultures of human PCa slices, we demonstrated that LA and EPA exert anticancer effects, by modulating Ca2+ entry, which was involved in Zeb1 regulation and cancer cellular migration. This functional approach using human prostate tumors highlights the clinical relevance of our observations, and may allow us to consider the possibility of targeting cancer spread by altering the lipid microenvironment.
Collapse
|
287
|
Yang L, Li D, Tang P, Zuo Y. Curcumin increases the sensitivity of K562/DOX cells to doxorubicin by targeting S100 calcium-binding protein A8 and P-glycoprotein. Oncol Lett 2019; 19:83-92. [PMID: 31897118 PMCID: PMC6924120 DOI: 10.3892/ol.2019.11083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 06/04/2019] [Indexed: 12/17/2022] Open
Abstract
The development of multidrug resistance (MDR) has seriously impeded the efficacy of drug treatment of chronic myeloid leukemia (CML). Recent studies have indicated that S100 calcium-binding protein A8 (S100A8) is associated with the occurrence and development of MDR. Traditional Chinese medicine may provide drugs with the potential to be used as multidrug resistance reversal agents with low toxicity and multi-target characteristics. The present study selected K562/DOX cells, a CML drug-resistant cell line, as a research model, and aimed to examine whether curcumin was able to reverse the resistance to doxorubicin (DOX), and elucidate the underlying molecular mechanisms. An MTT cytotoxicity assay indicated that curcumin at 0.5–2 µM reversed DOX resistance with a reversal index of 1.3–9.3. Western blot analysis revealed that curcumin treatment caused a downregulation of the expression of P-glycoprotein (P-gp) and S100A8 in a dose- and time-dependent manner. To study the internal association between S100A8 and P-gp, and the S100A8 role in drug resistance reversal, an RNA knockdown assay was conducted; however, S100A8 did not regulate the expression of P-gp or vice versa. After inhibiting the expression of S100A8 with specific small interfering RNA (si-S100A8), the sensitivity of K562/DOX cells to DOX was enhanced. In addition, si-S100A8 did not increase the intracellular accumulation of DOX, but increased the intracellular free calcium ion content, and the expression and activity of apoptosis-associated proteins, thereby inducing apoptosis. In conclusion, the present study suggested that inhibition of S100A8 expression increased DOX-induced apoptosis, and curcumin acted independently on S100A8 and P-gp to exert its drug resistance reversal effects
Collapse
Affiliation(s)
- Liu Yang
- Center for Post-doctoral Research, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Clinical Biochemistry, School of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Duo Li
- College of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Peiyan Tang
- College of Stomatology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yunfei Zuo
- Center for Post-doctoral Research, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Department of Clinical Biochemistry, School of Laboratory Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
288
|
Yang G, Zhang Y, Yang J. A Five-microRNA Signature as Prognostic Biomarker in Colorectal Cancer by Bioinformatics Analysis. Front Oncol 2019; 9:1207. [PMID: 31799184 PMCID: PMC6863365 DOI: 10.3389/fonc.2019.01207] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/23/2019] [Indexed: 01/09/2023] Open
Abstract
Mounting evidence has demonstrated that a lot of miRNAs are overexpressed or downregulated in colorectal cancer (CRC) tissues and play a crucial role in tumorigenesis, invasion, and migration. The aim of our study was to screen new biomarkers related to CRC prognosis by bioinformatics analysis. By using the R language edgeR package for the differential analysis and standardization of miRNA expression profiles from The Cancer Genome Atlas (TCGA), 502 differentially expressed miRNAs (343 up-regulated, 159 down-regulated) were screened based on the cut-off criteria of p < 0.05 and |log2FC|>1, then all the patients (421) with differentially expressed miRNAs and complete survival time, status were then randomly divided into train group (212) and the test group (209). Eight miRNAs with p < 0.005 were revealed in univariate cox regression analysis of train group, then stepwise multivariate cox regression was applied for constituting a five-miRNA (hsa-miR-5091, hsa-miR-10b-3p, hsa-miR-9-5p, hsa-miR-187-3p, hsa-miR-32-5p) signature prognostic biomarkers with obviously different overall survival. Test group and entire group shown the same results utilizing the same prescient miRNA signature. The area under curve (AUC) of receiver operating characteristic (ROC) curve for predicting 5 years survival in train group, test group, and whole cohort were 0.79, 0.679, and 0.744, respectively, which demonstrated better predictive power of prognostic model. Furthermore, Univariate cox regression and multivariate cox regression considering other clinical factors displayed that the five-miRNA signature could serve as an independent prognostic factor. In order to predict the potential biological functions of five-miRNA signature, target genes of these five miRNAs were analyzed by Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway and Gene Ontology (GO) enrichment analysis. The top 10 hub genes (ESR1, ADCY9, MEF2C, NRXN1, ADCY5, FGF2, KITLG, GATA1, GRIA1, KAT2B) of target genes in protein protein interaction (PPI) network were screened by string database and Cytoscape 3.6.1 (plug-in cytoHubba). In addition, 19 of target genes were associated with survival prognosis. Taken together, the current study showed the model of five-miRNA signature could efficiently function as a novel and independent prognosis biomarker and therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Guodong Yang
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Yujiao Zhang
- Respiratory Medicine, Huanggang Central Hospital Affiliated to Yangtze University, Huanggang, China
| | - Jiyuan Yang
- Department of Oncology, The First People's Hospital Affiliated to Yangtze University, Jingzhou, China
| |
Collapse
|
289
|
Soriani O, Kourrich S. The Sigma-1 Receptor: When Adaptive Regulation of Cell Electrical Activity Contributes to Stimulant Addiction and Cancer. Front Neurosci 2019; 13:1186. [PMID: 31780884 PMCID: PMC6861184 DOI: 10.3389/fnins.2019.01186] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
The sigma-1 receptor (σ1R) is an endoplasmic reticulum (ER)-resident chaperone protein that acts like an inter-organelle signaling modulator. Among its several functions such as ER lipid metabolisms/transports and indirect regulation of genes transcription, one of its most intriguing feature is the ability to regulate the function and trafficking of a variety of functional proteins. To date, and directly relevant to the present review, σ1R has been found to regulate both voltage-gated ion channels (VGICs) belonging to distinct superfamilies (i.e., sodium, Na+; potassium, K+; and calcium, Ca2+ channels) and non-voltage-gated ion channels. This regulatory function endows σ1R with a powerful capability to fine tune cells’ electrical activity and calcium homeostasis—a regulatory power that appears to favor cell survival in pathological contexts such as stroke or neurodegenerative diseases. In this review, we present the current state of knowledge on σ1R’s role in the regulation of cellular electrical activity, and how this seemingly adaptive function can shift cell homeostasis and contribute to the development of very distinct chronic pathologies such as psychostimulant abuse and tumor cell growth in cancers.
Collapse
Affiliation(s)
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
290
|
Jiang SH, Zhu LL, Zhang M, Li RK, Yang Q, Yan JY, Zhang C, Yang JY, Dong FY, Dai M, Hu LP, Li J, Li Q, Wang YH, Yang XM, Zhang YL, Nie HZ, Zhu L, Zhang XL, Tian GA, Zhang XX, Cao XY, Tao LY, Huang S, Jiang YS, Hua R, Qian Luo K, Gu JR, Sun YW, Hou S, Zhang ZG. GABRP regulates chemokine signalling, macrophage recruitment and tumour progression in pancreatic cancer through tuning KCNN4-mediated Ca 2+ signalling in a GABA-independent manner. Gut 2019; 68:1994-2006. [PMID: 30826748 DOI: 10.1136/gutjnl-2018-317479] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 02/04/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer-related death worldwide. Neurotransmitter-initiated signalling pathway is profoundly implicated in tumour initiation and progression. Here, we investigated whether dysregulated neurotransmitter receptors play a role during pancreatic tumourigenesis. METHODS The Cancer Genome Atlas and Gene Expression Omnibus datasets were used to identify differentially expressed neurotransmitter receptors. The expression pattern of gamma-aminobutyric acid type A receptor pi subunit (GABRP) in human and mouse PDAC tissues and cells was studied by immunohistochemistry and western blot analysis. The in vivo implications of GABRP in PDAC were tested by subcutaneous xenograft model and lung metastasis model. Bioinformatics analysis, transwell experiment and orthotopic xenograft model were used to identify the in vitro and in vivo effects of GABRP on macrophages in PDAC. ELISA, co-immunoprecipitation, proximity ligation assay, electrophysiology, promoter luciferase activity and quantitative real-time PCR analyses were used to identify molecular mechanism. RESULTS GABRP expression was remarkably increased in PDAC tissues and associated with poor prognosis, contributed to tumour growth and metastasis. GABRP was correlated with macrophage infiltration in PDAC and pharmacological deletion of macrophages largely abrogated the oncogenic functions of GABRP in PDAC. Mechanistically, GABRP interacted with KCNN4 to induce Ca2+ entry, which leads to activation of nuclear factor κB signalling and ultimately facilitates macrophage infiltration by inducing CXCL5 and CCL20 expression. CONCLUSIONS Overexpressed GABRP exhibits an immunomodulatory role in PDAC in a neurotransmitter-independent manner. Targeting GABRP or its interaction partner KCNN4 may be an effective therapeutic strategy for PDAC.
Collapse
Affiliation(s)
- Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Departmentof Biliary-Pancreatic Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Li Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Man Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rong-Kun Li
- Department of Interventional Radiology, Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiang-Yu Yan
- Key Laboratory for Cellular Physiology of Ministry of Education, Department of Neurobiology, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | | | - Jian-Yu Yang
- Departmentof Biliary-Pancreatic Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fang-Yuan Dong
- Department of Gastroenterology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Miao Dai
- Department of Gynecologic Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Mei Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui-Zhen Nie
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guang-Ang Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Yan Cao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ling-Ye Tao
- Departmentof Biliary-Pancreatic Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shan Huang
- College of Animal Science, Jilin University, Changchun, China
| | - Yong-Sheng Jiang
- Departmentof Biliary-Pancreatic Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rong Hua
- Departmentof Biliary-Pancreatic Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jian-Ren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Wei Sun
- Departmentof Biliary-Pancreatic Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shangwei Hou
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
291
|
Dong W, Cao Z, Pang Y, Feng T, Tian H. CARF, As An Oncogene, Promotes Colorectal Cancer Stemness By Activating ERBB Signaling Pathway. Onco Targets Ther 2019; 12:9041-9051. [PMID: 31802911 PMCID: PMC6830361 DOI: 10.2147/ott.s225733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/14/2019] [Indexed: 12/27/2022] Open
Abstract
Introduction The role of CARF, a calcium-responsive transcription factor, in colorectal cancer initiation and development is still unknown. Here, we report that CARF promotes colorectal cancer stemness through ERBB signaling pathway. Materials and methods Both colorectal cancer cell lines and primary cells were used in this study. The levels of target mRNA and protein in the cells were examined by qRT-PCR and Western blot. Gene manipulation was achieved by the lentivirus delivery system. Luciferase reporter gene assay was employed to analyze the transcriptional activity of the promoter. ChIP assay was performed for the examination of the binding between CARF and the promoters of MAPK8 and JUN. Kaplan-Meier survival curve was generated by the R2 program. Correlation analysis was performed using Spearman correlation analysis. Results Aberrant upregulation of CARF has been found in tumor tissues of colorectal cancer patients and associated with poor prognosis. Ectopic expression of CARF promoted the sphere-formation activities, as well as the expression of stem cell markers in colorectal cancer cells and knockdown of CARF, inhibited these activities. The mechanistic analysis showed that CARF directly binds to the promoter of MAPK8 and JUN, promotes the expression of MAPK8 and JUN, activates the ERBB signaling pathway, and thereby promotes the maintenance of the stemness in colorectal cancer cells. Conclusion CARF, as an oncogene, promotes colorectal cancer stemness by activating ERBB signaling pathway. The ERBB signaling pathway that serves as the main downstream effector of CARF could be an efficient drug target for colorectal cancer caused by aberrant expression of CARF.
Collapse
Affiliation(s)
- Weiyi Dong
- Department of Pathology, Heze Municipal Hospital, Heze City, Shandong 274031, People's Republic of China
| | - Zheng Cao
- Department of Pathology, Juye County People's Hospital, Heze City, Shandong 274900, People's Republic of China
| | - Yanmin Pang
- Department of Intensive Care Unit, Heze Municipal Hospital, Heze City, Shandong 274031, People's Republic of China
| | - Teng Feng
- Department of Pathology, Heze Municipal Hospital, Heze City, Shandong 274031, People's Republic of China
| | - Hongtao Tian
- Department of Pathology, Heze Municipal Hospital, Heze City, Shandong 274031, People's Republic of China
| |
Collapse
|
292
|
The regulatory roles of calcium channels in tumors. Biochem Pharmacol 2019; 169:113603. [DOI: 10.1016/j.bcp.2019.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023]
|
293
|
Xie R, Tuo B, Yang S, Chen XQ, Xu J. Calcium-sensing receptor bridges calcium and telomerase reverse transcriptase in gastric cancers via Akt. Clin Transl Oncol 2019; 22:1023-1032. [PMID: 31650467 DOI: 10.1007/s12094-019-02226-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 10/03/2019] [Indexed: 01/27/2023]
Abstract
PURPOSE Human telomerase reverse transcriptase (hTERT) and calcium-sensing receptor (CaSR) act as an oncogene in gastric cancers, however, their relationship in the progression of gastric cancers is yet to be elucidated. Herein, we aimed to access the potential interaction between hTERT and CaSR in the development of gastric cancers. METHODS The clinical data of 41 patients with gastric cancers were analyzed regarding the expressions of hTERT and CaSR by immunohistochemistry. Among them, five patients' specimens were also analyzed by Western blotting. The regulation of calcium on the expression level of hTERT and the possible underlying mechanism via CaSR were explored in gastric cancer cell lines MKN45 and SGC-7901. RESULTS Both hTERT and CaSR were increased and positively correlated in human gastric cancers, which also occurs in gastric cancer cells MKN45 and SGC-7901. Calcium induced hTERT expression at the transcriptional level in a CaSR-dependent manner followed by an increase in telomerase activity, as either a CaSR shRNA or the CaSR antagonist NPS2143 abolished the calcium-mediated regulation of hTERT and telomerase activity. Further studies showed that CaSR-mediated cytosolic calcium rise followed by Akt activation was involved in the regulation of hTERT by extracellular calcium. Finally, neither CaSR overexpression nor shRNA-mediated CaSR downregulation had an effect on the expression level of hTERT. CONCLUSIONS Our findings established a functional linkage between CaSR and hTERT in the development of gastric cancers and CaSR-hTERT coupling might serve as a novel target for therapeutic strategy against human gastric cancers.
Collapse
Affiliation(s)
- R Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - B Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - S Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - X-Q Chen
- Department of Neurosciences, School of Medicine, University of California, San Diego, CA, 92093, USA.
| | - J Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
294
|
Ma X, Lu JY, Moraru A, Teleman AA, Fang J, Qiu Y, Liu P, Xu T. A novel regulator of ER Ca 2+ drives Hippo-mediated tumorigenesis. Oncogene 2019; 39:1378-1387. [PMID: 31649333 DOI: 10.1038/s41388-019-1076-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 02/07/2023]
Abstract
Calcium ion (Ca2+) is a versatile second messenger that regulates various cellular and physiological functions. However, the in vivo molecular mechanisms by which Ca2+ alterations contribute to tumor growth remain poorly explored. Here we show that Emei is a novel ER Ca2+ regulator that synergizes with RasV12 to induce tumor growth via JNK-mediated Hippo signaling. Emei disruption reduces ER Ca2+ level and subsequently leads to JNK activation and Hippo inactivation. Importantly, genetically increasing cytosolic Ca2+ concentration cooperates with RasV12 to drive tumor growth via inactivating the Hippo pathway. Finally, we identify POSH as a crucial link that bridges cytosolic Ca2+ alteration with JNK activation and Hippo-mediated tumor growth. Together, our findings provide a novel mechanism of tumor growth that acts through intracellular Ca2+ levels to modulate JNK-mediated Hippo signaling.
Collapse
Affiliation(s)
- Xianjue Ma
- School of Life Sciences, Westlake University, Hangzhou, China. .,Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| | - Jin-Yu Lu
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.,Baylor College of Medicine, Hematology & Oncology, Houston, TX, USA
| | | | - Aurelio A Teleman
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg University, 69120, Heidelberg, Germany.,CellNetworks - Cluster of Excellence, Heidelberg University, Heidelberg, Germany
| | - Jinan Fang
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Yue Qiu
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Peng Liu
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Tian Xu
- School of Life Sciences, Westlake University, Hangzhou, China. .,Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
295
|
Almeida N, Carrara G, Palmeira CM, Fernandes AS, Parsons M, Smith GL, Saraiva N. Stimulation of cell invasion by the Golgi Ion Channel GAAP/TMBIM4 via an H 2O 2-Dependent Mechanism. Redox Biol 2019; 28:101361. [PMID: 31693977 PMCID: PMC6838802 DOI: 10.1016/j.redox.2019.101361] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
The mechanisms by which the Golgi apparatus (GA) impacts on cell invasion are poorly understood. The human Golgi Anti-Apoptotic Protein (hGAAP, also known as TMBIM4) is a highly conserved Golgi cation channel that modulates intracellular Ca2+ fluxes. Human GAAP is expressed in all human tissues, is essential for cell viability and provides resistance against a range of apoptotic stresses. Furthermore, hGAAP enhances adhesion and cell migration by increasing the turnover of focal adhesions due to activation of store-operated Ca2+ entry. Here, we describe a GA-derived mechanism that controls cell invasion. The overexpression of hGAAP stimulates 3-dimensional proteolytic cell invasion by a mechanism that is dependent on the accumulation of intracellular hydrogen peroxide, which might be produced by the hGAAP-dependent stimulation of mitochondrial respiration. These findings provide new insight into the complex mechanisms by which Ca2+ and reactive oxygen species signaling contribute to cell invasion and to the role of the GA in these processes.
Collapse
Affiliation(s)
- Nuno Almeida
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisbon, 1749-024, Portugal
| | - Guia Carrara
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Carlos M Palmeira
- Department of Life Sciences, University of Coimbra, Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ana S Fernandes
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisbon, 1749-024, Portugal
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guys Campus, London, SE1 1UL, UK
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Campo Grande 376, Lisbon, 1749-024, Portugal.
| |
Collapse
|
296
|
So CL, Saunus JM, Roberts-Thomson SJ, Monteith GR. Calcium signalling and breast cancer. Semin Cell Dev Biol 2019; 94:74-83. [DOI: 10.1016/j.semcdb.2018.11.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 12/11/2022]
|
297
|
Matsumoto K, Kato S. [Physiological and pathophysiological roles of TRPV4 channel in gastrointestinal tract]. Nihon Yakurigaku Zasshi 2019; 154:92-96. [PMID: 31527366 DOI: 10.1254/fpj.154.92] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective cation channel that responds to mechanical, thermal, and chemical stimuli in addition to various endogenous ligands, such as arachidonic acid metabolites. The present study aimed to elucidate the expression of TRPV4 in the gastrointestinal tract and the pathogenic roles of TRPV4 in dextran sulphate sodium (DSS)-induced colitis. TRPV4-immunoreactivity was detected in epithelial-like cells of the mouse tongue, esophagus, stomach, ileum, and colon; TRPV4 expression in the tongue was higher than other gastrointestinal tracts. TRPV4 colocalized with a type IV cell marker sonic hedgehog in circumvallate papillae. These findings suggest that TRPV4 contributes to sour taste sensing by regulating type III taste cell differentiation in mice. DSS-induced colitis was significantly attenuated in TRPV4-knockout (TRPV4KO) mice when compared to wild-type mice. DSS treatment upregulated TRPV4 expression in vascular endothelia of colonic mucosa and submucosa. DSS treatment increased vascular permeability, which was abolished in TRPV4KO mice. The activation of TRPV4 decreased VE-cadherin expression in mouse aortic endothelial cells exposed to TNF-α. These findings indicate that the upregulation of TRPV4 in vascular endothelial cells contributes to the progression of colonic inflammation via the activation of vascular permeability. Thus, TRPV4 is an attractive target for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Kenjiro Matsumoto
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University
| | - Shinichi Kato
- Division of Pathological Sciences, Department of Pharmacology and Experimental Therapeutics, Kyoto Pharmaceutical University
| |
Collapse
|
298
|
Wang C, Yu F, Liu X, Chen S, Wu R, Zhao R, Hu F, Yuan H. Cancer-Specific Therapy by Artificial Modulation of Intracellular Calcium Concentration. Adv Healthc Mater 2019; 8:e1900501. [PMID: 31368208 DOI: 10.1002/adhm.201900501] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/18/2019] [Indexed: 01/16/2023]
Abstract
Calcium (Ca2+ ) hemeostasis is crucial for the normal function of cellular biochemistry. The abnormal frequency of Ca2+ signaling in cancer cells makes them more vulnerable to Ca2+ modulation than normal cells. Here in this study, a novel cancer-specific therapy by artificially triggering Ca2+ overload in cancer cells is proposed. The feasibility of this therapy is illustrated by successful coupling of selective extrusion (Ca2+ ) inhibition effect of Curcumin (Cur) and the effective Ca2+ generating capability of amorphous calcium carbonate (ACC) into a facilely prepared water responsive phospholipid (PL)-ACC hybrid platform (PL/ACC-Cur). The obtained results demonstrate that PL/ACC-Cur can specifically boost the intracellular Ca2+ concentration to cause Ca2+ overload and to trigger mitochondria-related apoptosis in MCF-7 cells while sparing normal hepatocyte (L02), which might be a promising approach for effective cancer therapy.
Collapse
Affiliation(s)
- Cheng Wang
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
- School of Food Science and Pharmaceutical EngineeringNanjing Normal University No. 1 Wenyuan Road Nanjing 210046 China
| | - Fangying Yu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Xuerong Liu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Shaoqing Chen
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Rui Wu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Rui Zhao
- Sir Run Run Shaw HospitalSchool of MedicineZhejiang University No. 3 Qingchun East Road Hangzhou 310016 China
| | - Fuqiang Hu
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| | - Hong Yuan
- College of Pharmaceutical SciencesZhejiang University 866 Yuhangtang Road Hangzhou 310058 China
| |
Collapse
|
299
|
Silvestri R, Pucci P, Venalainen E, Matheou C, Mather R, Chandler S, Aceto R, Rigas SH, Wang Y, Rietdorf K, Bootman MD, Crea F. T-type calcium channels drive the proliferation of androgen-receptor negative prostate cancer cells. Prostate 2019; 79:1580-1586. [PMID: 31334879 DOI: 10.1002/pros.23879] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) is the treatment of choice for metastatic prostate cancer (PCa). After an initial response to ADT, PCa cells can generate castration resistant (CRPC) or neuroendocrine (NEPC) malignancies, which are incurable. T-type calcium channels (TTCCs) are emerging as promising therapeutic targets for several cancers, but their role in PCa progression has never been investigated. METHODS To examine the role of TTCCs in PCa, we analyzed their expression level, copy number variants (CNV) and prognostic significance using clinical datasets (Oncomine and cBioPortal). We then evaluated TTCC expression in a panel of PCa cell lines and measured the effect of their inhibition on cell proliferation and survival using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and caspase assays. RESULTS TTCCs were upregulated in PCas harboring androgen receptor (AR) mutations; CNV rate was positively associated with PCa progression. Higher expression of one TTCC isoform (CACNA1G) predicted poorer postoperative prognosis in early stage PCa samples. Pharmacological or small interfering RNA (siRNA)-based inhibition of TTCCs caused a decrease in PC-3 cell survival and proliferation. CONCLUSIONS Our results show that TTCCs are overexpressed in advanced forms of PCa and correlate with a poorer prognosis. TTCC inhibition reduces cell proliferation and survival, suggesting that there may be possible value in the therapeutic targeting of TTCCs in advanced PCa.
Collapse
Affiliation(s)
- Roberto Silvestri
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
- Department of Biology, University of Pisa, Pisa, Italy
| | - Perla Pucci
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Erik Venalainen
- Department of Experimental Therapeutics, BCCA Cancer Research Centre, Vancouver, Canada
| | - Chrysanthi Matheou
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Rebecca Mather
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Stephen Chandler
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Romina Aceto
- Department of Biology, University of Pisa, Pisa, Italy
| | - Sushilaben H Rigas
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Yuzhuo Wang
- Department of Biology, University of Pisa, Pisa, Italy
| | - Katja Rietdorf
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Martin David Bootman
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Francesco Crea
- School of Life Health and Chemical Sciences, The Open University, Milton Keynes, UK
| |
Collapse
|
300
|
Abstract
As the leading cause of death in cancer, there is an urgent need to develop treatments to target the dissemination of primary tumor cells to secondary organs, known as metastasis. Bioelectric signaling has emerged in the last century as an important controller of cell growth, and with the development of current molecular tools we are now beginning to identify its role in driving cell migration and metastasis in a variety of cancer types. This review summarizes the currently available research for bioelectric signaling in solid tumor metastasis. We review the steps of metastasis and discuss how these can be controlled by bioelectric cues at the level of a cell, a population of cells, and the tissue. The role of ion channel, pump, and exchanger activity and ion flux is discussed, along with the importance of the membrane potential and the relationship between ion flux and membrane potential. We also provide an overview of the evidence for control of metastasis by external electric fields (EFs) and draw from examples in embryogenesis and regeneration to discuss the implications for endogenous EFs. By increasing our understanding of the dynamic properties of bioelectric signaling, we can develop new strategies that target metastasis to be translated into the clinic.
Collapse
Affiliation(s)
- Samantha L. Payne
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, Massachusetts
| | - Madeleine J. Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|