1
|
Thaklaewphan P, Wikan N, Potikanond S, Nimlamool W. Oxyresveratrol Enhances the Anti-Cancer Effect of Cisplatin against Epithelial Ovarian Cancer Cells through Suppressing the Activation of Protein Kinase B (AKT). Biomolecules 2024; 14:1140. [PMID: 39334906 PMCID: PMC11430010 DOI: 10.3390/biom14091140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Epithelial ovarian carcinoma poses a significant challenge due to its resistance to chemotherapy and propensity for metastasis, thereby reducing the effectiveness of conventional treatments. Hence, the identification of novel compounds capable of augmenting the anti-cancer efficacy of platinum-based chemotherapy is imperative. Oxyresveratrol (OXY), a derivative of resveratrol, has been demonstrated to possess antiproliferative and apoptosis-inducing effects across various cancer cell lines. Notably, OXY appears to exert its effects by inhibiting the PI3K/AKT/mTOR signaling pathway. However, the synergistic potential of OXY in combination with cisplatin against epithelial ovarian cancer has not yet been elucidated. The current study investigated the synergistic effects of OXY and cisplatin on the ovarian cancer cell lines SKOV3 and TOV21G. We found that OXY significantly enhanced cisplatin's ability to reduce cell viability, induce apoptosis, induce cell cycle arrest, and increase the proportion of cells in the sub-G1 phase. Furthermore, OXY treatment alone dose-dependently inhibited the production of anti-apoptotic proteins including Mcl-1, Bcl-xL, and XIAP under EGF activation. Mechanistically, OXY suppressed the PI3K/AKT/mTOR signaling pathway by reducing phosphorylated AKT, while having no discernible effect on the MAPK pathway. These findings highlight OXY's potential to enhance ovarian cancer cell sensitivity to chemotherapy, suggesting its development as a pharmaceutical adjunct for clinical use in combination therapies.
Collapse
Affiliation(s)
- Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
- Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
| | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.T.); (N.W.); (S.P.)
- Lanna Rice Research Center, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
2
|
Wang KH, Chang YH, Ding DC. Bone Marrow Mesenchymal Stem Cells Promote Ovarian Cancer Cell Proliferation via Cytokine Interactions. Int J Mol Sci 2024; 25:6746. [PMID: 38928452 PMCID: PMC11203416 DOI: 10.3390/ijms25126746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are key players in promoting ovarian cancer cell proliferation, orchestrated by the dynamic interplay between cytokines and their interactions with immune cells; however, the intricate crosstalk among BMSCs and cytokines has not yet been elucidated. Here, we aimed to investigate interactions between BMSCs and ovarian cancer cells. We established BMSCs with a characterized morphology, surface marker expression, and tri-lineage differentiation potential. Ovarian cancer cells (SKOV3) cultured with conditioned medium from BMSCs showed increased migration, invasion, and colony formation, indicating the role of the tumor microenvironment in influencing cancer cell behavior. BMSCs promoted SKOV3 tumorigenesis in nonobese diabetic/severe combined immunodeficiency mice, increasing tumor growth. The co-injection of BMSCs increased the phosphorylation of p38 MAPK and GSK-3β in SKOV3 tumors. Co-culturing SKOV3 cells with BMSCs led to an increase in the expression of cytokines, especially MCP-1 and IL-6. These findings highlight the influence of BMSCs on ovarian cancer cell behavior and the potential involvement of specific cytokines in mediating these effects. Understanding these mechanisms will highlight potential therapeutic avenues that may halt ovarian cancer progression.
Collapse
Affiliation(s)
- Kai-Hung Wang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
3
|
Sweed D, Elhamed SMA, Aiad HAS, Ehsan NA, Hemida AS, Dawoud MM. STIM1/SOX2 proteins are co-expressed in the tumor and microenvironmental stromal cells of pancreatic ductal adenocarcinoma and ampullary carcinoma. World J Surg Oncol 2024; 22:84. [PMID: 38532463 DOI: 10.1186/s12957-024-03356-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) and ampullary carcinoma (AAC) are lethal malignancies with modest benefits from surgery. SOX2 and STIM1 have been linked to anticancer activity in several human malignancies. This study included 94 tumor cases: 48 primary PDAC, 25 metastatic PDAC, and 21 primary AAC with corresponding non-tumor tissue. All cases were immunohistochemically stained for STIM1 and SOX2 and results were correlated with clinicopathologic data, patient survival, and BCL2 immunostaining results. Results revealed that STIM1 and SOX2 epithelial/stromal expressions were significantly higher in PDAC and AAC in comparison to the control groups. STIM1 and SOX2 expressions were positively correlated in the primary and metastatic PDAC (P = 0.016 and, P = 0.001, respectively). However, their expressions were not significantly associated with BCL2 expression. SOX2 epithelial/stromal expressions were positively correlated with the large tumor size in the primary AAC group (P = 0.052, P = 0.044, respectively). STIM1 stromal and SOX2 epithelial over-expressions had a bad prognostic impact on the overall survival of AAC (P = 0.002 and P = 0.001, respectively). Therefore, STIM1 and SOX2 co-expression in tumor cells and intra-tumoral stroma could contribute to the development of PDAC and AAC. STIM1/SOX2 expression is linked to a bad prognosis in AAC.
Collapse
Affiliation(s)
- Dina Sweed
- Pathology Department, National Liver Institute, Menoufia University, Shibin Al Koom, Egypt
| | | | - Hayam Abdel Samie Aiad
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin Al Koom, 32511, Egypt
| | - Nermine Ahmed Ehsan
- Pathology Department, National Liver Institute, Menoufia University, Shibin Al Koom, Egypt
| | - Aiat Shaban Hemida
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin Al Koom, 32511, Egypt
| | - Marwa Mohammed Dawoud
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin Al Koom, 32511, Egypt.
| |
Collapse
|
4
|
Lai YS, Chan TW, Nguyen TMH, Lin TC, Chao YY, Wang CY, Hung LY, Tsai SJ, Chiu WT. Store-operated calcium entry inhibits primary ciliogenesis via the activation of Aurora A. FEBS J 2024; 291:1027-1042. [PMID: 38050648 DOI: 10.1111/febs.17024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/02/2023] [Accepted: 12/04/2023] [Indexed: 12/06/2023]
Abstract
The primary cilium is an antenna-like organelle protruding from the cell surface that can detect physical and chemical stimuli in the extracellular space to activate specific signaling pathways and downstream gene expressions. Calcium ion (Ca2+ ) signaling regulates a wide spectrum of cellular processes, including fertilization, proliferation, differentiation, muscle contraction, migration, and death. This study investigated the effects of the regulation of cytosolic Ca2+ levels on ciliogenesis using chemical, genetic, and optogenetic approaches. We found that ionomycin-induced Ca2+ influx inhibited ciliogenesis and Ca2+ chelator BATPA-AM-induced Ca2+ depletion promoted ciliogenesis. In addition, store-operated Ca2+ entry and the endoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1) negatively regulated ciliogenesis. Moreover, an optogenetic platform was used to create different Ca2+ oscillation patterns by manipulating lighting parameters, including density, frequency, exposure time, and duration. Light-activated Ca2+ -translocating channelrhodopsin (CatCh) is activated by 470-nm blue light to induce Ca2+ influx. Our results show that high-frequency Ca2+ oscillations decrease ciliogenesis. Furthermore, the inhibition of cilia formation induced by Ca2+ may occur via the activation of Aurora kinase A. Cilia not only induce Ca2+ signaling but also regulate cilia formation by Ca2+ signaling.
Collapse
Affiliation(s)
- Yi-Shyun Lai
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ta-Wei Chan
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Thi My Hang Nguyen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Chien Lin
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ying Chao
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, Taiwan
| | - Liang-Yi Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
5
|
Chodurek E, Orchel A, Gwiazdoń P, Kaps A, Paduszyński P, Jaworska-Kik M, Chrobak E, Bębenek E, Boryczka S, Kasperczyk J. Antiproliferative and Cytotoxic Properties of Propynoyl Betulin Derivatives against Human Ovarian Cancer Cells: In Vitro Studies. Int J Mol Sci 2023; 24:16487. [PMID: 38003677 PMCID: PMC10671498 DOI: 10.3390/ijms242216487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Due to the incidence of ovarian cancer (OC) and the limitations of available therapeutic strategies, it is necessary to search for novel therapeutic solutions. The aim of this study was to evaluate the cytotoxic effect of betulin 1 and its propynoyl derivatives 2-6 against ovarian cancer cells (SK-OV-3, OVCAR-3) and normal myofibroblasts (18Co). Paclitaxel was used as the reference compound. The propynoyl derivatives 2-6 exhibited stronger antiproliferative and cytotoxic activities compared to betulin 1. In both ovarian cancer cell lines, the most potent compound was 28-propynoylbetulin 2. In the case of compound 2, the calculated IC50 values were 0.2 µM for the SK-OV-3 cells and 0.19 µM for the OVCAR-3 cells. Under the same culture conditions, the calculated IC50 values for compound 6 were 0.26 µM and 0.59 µM, respectively. It was observed that cells treated with compounds 2 and 6 caused a decrease in the potential of the mitochondrial membrane and a significant change in cell morphology. Betulin 1, a diol from the group of pentacyclic triterpenes, has a confirmed wide spectrum of biological effects, including a significant anticancer effect. It is characterized by low bioavailability, which can be improved by introducing changes to its structure. The results showed that chemical modifications of betulin 1 only at position C-28 with the propynoyl group (compound 2) and additionally at position C-3 with the phosphate group (compound 3) or at C-29 with the phosphonate group (compound 6) allowed us to obtain compounds with greater cytotoxic activity than their parent compounds, which could be used to develop novel therapeutic systems effective in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ewa Chodurek
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| | - Arkadiusz Orchel
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| | - Paweł Gwiazdoń
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| | - Anna Kaps
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| | - Piotr Paduszyński
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| | - Marzena Jaworska-Kik
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| | - Elwira Chrobak
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland; (E.C.); (E.B.); (S.B.)
| | - Ewa Bębenek
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland; (E.C.); (E.B.); (S.B.)
| | - Stanisław Boryczka
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland; (E.C.); (E.B.); (S.B.)
| | - Janusz Kasperczyk
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 8 Jedności Str., 41-208 Sosnowiec, Poland; (A.O.); (P.G.); (A.K.); (P.P.); (M.J.-K.); (J.K.)
| |
Collapse
|
6
|
Soussi M, Hasselsweiller A, Gkika D. TRP Channels: The Neglected Culprits in Breast Cancer Chemotherapy Resistance? MEMBRANES 2023; 13:788. [PMID: 37755210 PMCID: PMC10536409 DOI: 10.3390/membranes13090788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
Breast cancer is a major health concern worldwide, and resistance to therapies remains a significant challenge in treating this disease. In breast cancer, Transient Receptor Potential (TRP) channels are well studied and constitute key players in nearly all carcinogenesis hallmarks. Recently, they have also emerged as important actors in resistance to therapy by modulating the response to various pharmaceutical agents. Targeting TRP channels may represent a promising approach to overcome resistance to therapies in breast cancer patients.
Collapse
Affiliation(s)
| | | | - Dimitra Gkika
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (M.S.); (A.H.)
| |
Collapse
|
7
|
Hankittichai P, Thaklaewphan P, Wikan N, Ruttanapattanakul J, Potikanond S, Smith DR, Nimlamool W. Resveratrol Enhances Cytotoxic Effects of Cisplatin by Inducing Cell Cycle Arrest and Apoptosis in Ovarian Adenocarcinoma SKOV-3 Cells through Activating the p38 MAPK and Suppressing AKT. Pharmaceuticals (Basel) 2023; 16:ph16050755. [PMID: 37242538 DOI: 10.3390/ph16050755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
In the current study, we identified a mechanism of resveratrol (RES) underlying its anti-cancer properties against human ovarian adenocarcinoma SKOV-3 cells. We investigated its anti-proliferative and apoptosis-inducing effects in combination with cisplatin, using cell viability assay, flow cytometry, immunofluorescence study and Western blot analysis. We discovered that RES suppressed cancer cell proliferation and stimulated apoptosis, especially when combined with cisplatin. This compound also inhibited SKOV-3 cell survival, which may partly be due to its potential to inhibit protein kinase B (AKT) phosphorylation and induce the S-phase cell cycle arrest. RES in combination with cisplatin strongly induced cancer cell apoptosis through activating the caspase-dependent cascade, which was associated with its ability to stimulate nuclear phosphorylation of p38 mitogen-activated protein kinase (MAPK), well recognized to be involved in transducing environmental stress signals. RES-induced p38 phosphorylation was very specific, and the activation status of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) was not mainly affected. Taken together, our study provides accumulated evidence that RES represses proliferation and promotes apoptosis in SKOV-3 ovarian cancer cells through activating the p38 MAPK pathway. It is interesting that this active compound may be used as an effective agent to sensitize ovarian cancer to apoptosis induced by standard chemotherapies.
Collapse
Affiliation(s)
- Phateep Hankittichai
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phatarawat Thaklaewphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nitwara Wikan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Duncan R Smith
- Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
8
|
Alhamed AS, Alqinyah M, Alsufayan MA, Alhaydan IA, Alassmrry YA, Alnefaie HO, Algahtani MM, Alghaith AF, Alhamami HN, Albogami AM, Alhazzani K, AZ A. Blockade of store-operated calcium entry sensitizes breast cancer cells to cisplatin therapy via modulating inflammatory response. Saudi Pharm J 2023; 31:245-254. [PMID: 36942275 PMCID: PMC10023550 DOI: 10.1016/j.jsps.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
Store-operated calcium entry (SOCE) is an important pathway for calcium signaling that regulates calcium influx across the plasma membrane upon the depletion of calcium stores in the endoplasmic reticulum. SOCE participates in regulating a number of physiological processes including cell proliferation and migration while SOCE dysregulation has been linked with pathophysiological conditions such as inflammation and cancer. The crosslink between cancer and inflammation has been well-established where abundant evidence demonstrate that inflammation plays a role in cancer pathophysiology and the response of cancer cells to chemotherapeutic agents including cisplatin. Indeed, the efficacy of cisplatin against cancer cells is reduced by inflammation. Interestingly, it was shown that SOCE enhances inflammatory signaling in immune cells. Therefore, the main objectives of this study are to examine the impact of SOCE inhibition on the cisplatin sensitivity of breast cancer cells and to explore its related mechanism in modulating the inflammatory response in breast cancer cells. Our findings showed that SOCE inhibitor (BTP2) enhanced cisplatin cytotoxicity against resistant breast cancer cells via inhibition of cell proliferation and migration as well as induction of apoptosis. We also found an upregulation in the gene expression of two major components of SOCE, STIM1 and ORAI1, in cisplatin-resistant breast cancer cells compared to cisplatin-sensitive breast cancer cells. In addition, cisplatin treatment increased the gene expression of STIM1 and ORAI1 in cisplatin-resistant breast cancer cells. Finally, this study also demonstrated that cisplatin therapy caused an increase in the gene expression of inflammatory mediators COX2, IL-8, and TNF-α as well as COX2 protein and upon SOCE inhibition using BTP2, the effect of cisplatin on the inflammatory mediators was reversed. Altogether, this study has proven the pivotal role of SOCE in cisplatin resistance of breast cancer cells and showed the importance of targeting this pathway in improving breast cancer therapy.
Collapse
Affiliation(s)
- Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Corresponding author.
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Musab A. Alsufayan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ibrahim A. Alhaydan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Yasseen A. Alassmrry
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Hajar O. Alnefaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad M. Algahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Adel F. Alghaith
- Department of pharmaceutics, College of pharmacy, king Saud university, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hussain N. Alhamami
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah M. Albogami
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alanazi AZ
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
9
|
Rubaiy HN. ORAI Calcium Channels: Regulation, Function, Pharmacology, and Therapeutic Targets. Pharmaceuticals (Basel) 2023; 16:162. [PMID: 37259313 PMCID: PMC9967976 DOI: 10.3390/ph16020162] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 11/25/2023] Open
Abstract
The changes in intracellular free calcium (Ca2+) levels are one of the most widely regulators of cell function; therefore, calcium as a universal intracellular mediator is involved in very important human diseases and disorders. In many cells, Ca2+ inflow is mediated by store-operated calcium channels, and it is recognized that the store-operated calcium entry (SOCE) is mediated by the two partners: the pore-forming proteins Orai (Orai1-3) and the calcium store sensor, stromal interaction molecule (STIM1-2). Importantly, the Orai/STIM channels are involved in crucial cell signalling processes such as growth factors, neurotransmitters, and cytokines via interaction with protein tyrosine kinase coupled receptors and G protein-coupled receptors. Therefore, in recent years, the issue of Orai/STIM channels as a drug target in human diseases has received considerable attention. This review summarizes and highlights our current knowledge of the Orai/STIM channels in human diseases and disorders, including immunodeficiency, myopathy, tubular aggregate, Stormorken syndrome, York platelet syndrome, cardiovascular and metabolic disorders, and cancers, as well as suggesting these channels as drug targets for pharmacological therapeutic intervention. Moreover, this work will also focus on the pharmacological modulators of Orai/STIM channel complexes. Together, our thoughtful of the biology and physiology of the Orai/STIM channels have grown remarkably during the past three decades, and the next important milestone in the field of store-operated calcium entry will be to identify potent and selective small molecules as a therapeutic agent with the purpose to target human diseases and disorders for patient benefit.
Collapse
Affiliation(s)
- Hussein N Rubaiy
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska Institute and Karolinska University Hospital, C1:68, 141 86 Stockholm, Sweden
| |
Collapse
|
10
|
Romito O, Guéguinou M, Raoul W, Champion O, Robert A, Trebak M, Goupille C, Potier-Cartereau M. Calcium signaling: A therapeutic target to overcome resistance to therapies in cancer. Cell Calcium 2022; 108:102673. [PMID: 36410063 DOI: 10.1016/j.ceca.2022.102673] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Innate and acquired resistances to therapeutic agents are responsible for the failure of cancer treatments. Due to the multifactorial nature of resistance, the identification of new therapeutic targets is required to improve cancer treatment. Calcium is a universal second messenger that regulates many cellular functions such as proliferation, migration, and survival. Calcium channels, pumps and exchangers tightly regulate the duration, location and magnitude of calcium signals. Many studies have implicated dysregulation of calcium signaling in several pathologies, including cancer. Abnormal calcium fluxes due to altered channel expression or activation contribute to carcinogenesis and promote tumor development. However, there is limited information on the role of calcium signaling in cancer resistance to therapeutic drugs. This review discusses the role of calcium signaling as a mediator of cancer resistance, and assesses the potential value of combining anticancer therapy with calcium signaling modulators to improve the effectiveness of current treatments.
Collapse
Affiliation(s)
- Olivier Romito
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Maxime Guéguinou
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - William Raoul
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Ophélie Champion
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Alison Robert
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Mohamed Trebak
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Caroline Goupille
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France; CHRU de Tours, hôpital Bretonneau, Tours, France.
| | - Marie Potier-Cartereau
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| |
Collapse
|
11
|
Omics Analysis of Chemoresistant Triple Negative Breast Cancer Cells Reveals Novel Metabolic Vulnerabilities. Cells 2022; 11:cells11172719. [PMID: 36078127 PMCID: PMC9454761 DOI: 10.3390/cells11172719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
The emergence of drug resistance in cancer poses the greatest hurdle for successful therapeutic results and is associated with most cancer deaths. In triple negative breast cancer (TNBC), due to the lack of specific therapeutic targets, systemic chemotherapy is at the forefront of treatments, but it only benefits a fraction of patients because of the development of resistance. Cancer cells may possess an innate resistance to chemotherapeutic agents or develop new mechanisms of acquired resistance after long-term drug exposure. Such mechanisms involve an interplay between genetic, epigenetic and metabolic alterations that enable cancer cells to evade therapy. In this work, we generated and characterized a chemoresistant TNBC cell line to be used for the investigation of mechanisms that drive resistance to paclitaxel. Transcriptomic analysis highlighted the important role of metabolic-associated pathways in the resistant cells, prompting us to employ 1H-NMR to explore the metabolome and lipidome of these cells. We identified and described herein numerous metabolites and lipids that were significantly altered in the resistant cells. Integrated analysis of our omics data revealed MSMO1, an intermediate enzyme of cholesterol biosynthesis, as a novel mediator of chemoresistance in TNBC. Overall, our data provide a critical insight into the metabolic adaptations that accompany acquired resistance in TNBC and pinpoint potential new targets.
Collapse
|
12
|
Characterization of Vemurafenib-Resistant Melanoma Cell Lines Reveals Novel Hallmarks of Targeted Therapy Resistance. Int J Mol Sci 2022; 23:ijms23179910. [PMID: 36077308 PMCID: PMC9455970 DOI: 10.3390/ijms23179910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Regardless of the significant improvements in treatment of melanoma, the majority of patients develop resistance whose mechanisms are still not completely understood. Hence, we generated and characterized two melanoma-derived cell lines, primary WM793B and metastatic A375M, with acquired resistance to the RAF inhibitor vemurafenib. The morphology of the resistant primary WM793B melanoma cells showed EMT-like features and exhibited a hybrid phenotype with both epithelial and mesenchymal characteristics. Surprisingly, the vemurafenib-resistant melanoma cells showed a decreased migration ability but also displayed a tendency to collective migration. Signaling pathway analysis revealed the reactivation of MAPK and the activation of the PI3K/AKT pathway depending on the vemurafenib-resistant cell line. The acquired resistance to vemurafenib caused resistance to chemotherapy in primary WM793B melanoma cells. Furthermore, the cell-cycle analysis and altered levels of cell-cycle regulators revealed that resistant cells likely transiently enter into cell cycle arrest at the G0/G1 phase and gain slow-cycling cell features. A decreased level of NME1 and NME2 metastasis suppressor proteins were found in WM793B-resistant primary melanoma, which is possibly the result of vemurafenib-acquired resistance and is one of the causes of increased PI3K/AKT signaling. Further studies are needed to reveal the vemurafenib-dependent negative regulators of NME proteins, their role in PI3K/AKT signaling, and their influence on vemurafenib-resistant melanoma cell characteristics.
Collapse
|
13
|
Audero MM, Prevarskaya N, Fiorio Pla A. Ca 2+ Signalling and Hypoxia/Acidic Tumour Microenvironment Interplay in Tumour Progression. Int J Mol Sci 2022; 23:7377. [PMID: 35806388 PMCID: PMC9266881 DOI: 10.3390/ijms23137377] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/18/2023] Open
Abstract
Solid tumours are characterised by an altered microenvironment (TME) from the physicochemical point of view, displaying a highly hypoxic and acidic interstitial fluid. Hypoxia results from uncontrolled proliferation, aberrant vascularization and altered cancer cell metabolism. Tumour cellular apparatus adapts to hypoxia by altering its metabolism and behaviour, increasing its migratory and metastatic abilities by the acquisition of a mesenchymal phenotype and selection of aggressive tumour cell clones. Extracellular acidosis is considered a cancer hallmark, acting as a driver of cancer aggressiveness by promoting tumour metastasis and chemoresistance via the selection of more aggressive cell phenotypes, although the underlying mechanism is still not clear. In this context, Ca2+ channels represent good target candidates due to their ability to integrate signals from the TME. Ca2+ channels are pH and hypoxia sensors and alterations in Ca2+ homeostasis in cancer progression and vascularization have been extensively reported. In the present review, we present an up-to-date and critical view on Ca2+ permeable ion channels, with a major focus on TRPs, SOCs and PIEZO channels, which are modulated by tumour hypoxia and acidosis, as well as the consequent role of the altered Ca2+ signals on cancer progression hallmarks. We believe that a deeper comprehension of the Ca2+ signalling and acidic pH/hypoxia interplay will break new ground for the discovery of alternative and attractive therapeutic targets.
Collapse
Affiliation(s)
- Madelaine Magalì Audero
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| | - Natalia Prevarskaya
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
| | - Alessandra Fiorio Pla
- U1003—PHYCEL—Laboratoire de Physiologie Cellulaire, Inserm, University of Lille, Villeneuve d’Ascq, 59000 Lille, France; (M.M.A.); (N.P.)
- Laboratory of Cellular and Molecular Angiogenesis, Department of Life Sciences and Systems Biology, University of Turin, 10123 Turin, Italy
| |
Collapse
|
14
|
Chen H, He Y, Wen X, Shao S, Liu Y, Wang J. SOX9: Advances in Gynecological Malignancies. Front Oncol 2021; 11:768264. [PMID: 34881182 PMCID: PMC8645898 DOI: 10.3389/fonc.2021.768264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/05/2021] [Indexed: 01/10/2023] Open
Abstract
Transcription factors of the SOX family were first discovered in mammals in 1990. The sex-determining region Y box 9 belongs to the SOX transcription factor family. It plays an important role in inducing tissue and cell morphogenesis, survival, and many developmental processes. Furthermore, it has been shown to be an oncogene in many tumors. Gynecological malignancies are tumors that occur in the female reproductive system and seriously threaten the lives of patients. Common gynecological malignancies include ovarian cancer, cervical cancer, and endometrial cancer. So far, the molecular mechanisms related to the incidence and development of gynecological malignancies remain unclear. This makes it particularly important to discover their common causative molecule and thus provide an effective therapeutic target. In recent years, studies have found that multiple mechanisms are involved in regulating the expression of the sex-determining region Y box 9, leading to the occurrence and development of gynecological malignancies. In this review, we discuss the prognostic value of SOX9 expression and the potential of targeting SOX9 for gynecological malignancy treatment. We also discuss progress regarding the role of SOX9 in gynecological malignancy pathogenesis through its mediation of important mechanisms, including tumor initiation and proliferation, apoptosis, migration, invasion, chemoresistance, and stem cell maintenance.
Collapse
Affiliation(s)
- Huan Chen
- Department of Obstetrics and Gynecology, Zhu Zhou Central Hospital, Zhuzhou, China
| | - Yujie He
- Designated Ward, Zhu Zhou Central Hospital, Zhuzhou, China
| | - Xiangping Wen
- Department of Operation, Zhu Zhou Central Hospital, Zhuzhou, China
| | - Shihong Shao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujie Liu
- Department of Obstetrics and Gynecology, Zhu Zhou Central Hospital, Zhuzhou, China
| | - Jinjin Wang
- Department of Obstetrics and Gynecology, Zhu Zhou Central Hospital, Zhuzhou, China
| |
Collapse
|
15
|
Nunes M, Silva PMA, Coelho R, Pinto C, Resende A, Bousbaa H, Almeida GM, Ricardo S. Generation of Two Paclitaxel-Resistant High-Grade Serous Carcinoma Cell Lines With Increased Expression of P-Glycoprotein. Front Oncol 2021; 11:752127. [PMID: 34745981 PMCID: PMC8566917 DOI: 10.3389/fonc.2021.752127] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Debulking surgery followed by chemotherapy are the standard of care for high-grade serous carcinoma. After an initial good response to treatment, the majority of patients relapse with a chemoresistant profile, leading to a poor overall survival. Chemotherapy regimens used in high-grade serous carcinomas are based in a combination of classical chemotherapeutic drugs, namely, Carboplatin and Paclitaxel. The mechanisms underlying drug resistance and new drug discovery are crucial to improve patients’ survival. To uncover the molecular mechanisms of chemoresistance and test drugs capable of overcoming this resistant profile, it is fundamental to use good cellular models capable of mimicking the chemoresistant disease. Herein, we established two high-grade serous carcinoma cell lines with intrinsic resistance to Carboplatin and induced Paclitaxel resistance (OVCAR8 PTX R C and OVCAR8 PTX R P) derived from the OVCAR8 cell line. These two chemoresistant cell line variants acquired an enhanced resistance to Paclitaxel-induced cell death by increasing the drug efflux capacity, and this resistance was stable in long-term culture and following freeze/thaw cycles. The mechanism underlying Paclitaxel resistance resides in a significant increase in P-glycoprotein expression and, when this drug efflux pump was blocked with Verapamil, cells re-acquired Paclitaxel sensitivity. We generated two high-grade serous carcinoma cell lines, with a double-chemoresistant (Carboplatin and Paclitaxel) phenotype that mimics the majority of tumor recurrences in ovarian cancer context. This robust tool is suitable for preliminary drug testing towards the development of therapeutic strategies to overcome chemoresistance.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Patrícia M A Silva
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal.,TOXRUN, Toxicology Research Unit, University Institute of Health Sciences, Advanced Polytechnic and University Cooperative (CESPU), Gandra, Portugal
| | - Ricardo Coelho
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Carla Pinto
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal.,Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
| | - Albina Resende
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal.,Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
| | - Hassan Bousbaa
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal
| | - Gabriela M Almeida
- Expression Regulation in Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,Faculty of Medicine from University of Porto (FMUP), Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,TOXRUN, Toxicology Research Unit, University Institute of Health Sciences, Advanced Polytechnic and University Cooperative (CESPU), Gandra, Portugal.,Faculty of Medicine from University of Porto (FMUP), Porto, Portugal
| |
Collapse
|
16
|
Xu X, Chen F, Zhang L, Liu L, Zhang C, Zhang Z, Li W. Exploring the mechanisms of anti-ovarian cancer of Hedyotis diffusa Willd and Scutellaria barbata D. Don through focal adhesion pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114343. [PMID: 34147618 DOI: 10.1016/j.jep.2021.114343] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/28/2021] [Accepted: 06/15/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa Willd and Scutellaria barbata D.Don (HD-SB) pairing were widely used as traditional medicine known for their anti-tumor effects. However, the inhibitory effect of HD-SB on ovarian cancer and its potential mechanisms were still not clear. AIM OF THE STUDY Our study identified the anti-tumor effect of HD-SB on ovarian cancer and analyzed the potential mechanisms by the network pharmacology and molecular docking method. MATERIALS AND METHODS The inhibitory effect of HD-SB combination on the growth and migration of ovarian cancer was detected by MTT and transwell assays. The effective ingredients of HD-SB and their potential targets were obtained from the Traditional Chinese Medicines for Systems Pharmacology Database (TCMSP), the GeneCards database, and the UniProt database. The relationships between active ingredients of HD-SB and potential targets or pathways of ovarian cancer were analyzed by String database, Cytoscape 3.7.2 software, and David 6.7 online database. The anti-ovarian cancer targets of HD-SB in the focal adhesion pathway were identified by RT-qPCR and molecular docking. RESULTS HD-SB combination significantly inhibited the proliferation and migration of ovarian cancer cells. We observed that the 1:2 ratio of HD-SB had the lowest IC50 value. 60 gene targets of 33 active ingredients in HD-SB were selected by pharmacokinetic parameters. The network pharmacological analysis showed that quercetin, luteolin, and baicalein might be the important anti-ovarian cancer ingredients in HD-SB, and the inhibitory effects of these three ingredients on the proliferation of ovarian cancer cells were verified respectively. Functional enrichment results suggested that HD-SB inhibited ovarian cancer growth and migration mainly through the focal adhesion pathway and the potential targets were EGFR, MAPK1, VEGFA, and PIK3CG. CONCLUSIONS HD-SB pairing significantly inhibited the proliferation and migration of ovarian cancer. Using network pharmacological methods and validation experiments, we found that HD-SB might, at least partially, inhibit ovarian cancer through the focal adhesion pathway. We believed that the HD-SB combination could be a potential therapeutic drug for the treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Fenglin Chen
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Cuili Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Zhiwei Zhang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| |
Collapse
|
17
|
Cui C, Zhang Y, Liu G, Zhang S, Zhang J, Wang X. Advances in the study of cancer metastasis and calcium signaling as potential therapeutic targets. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:266-291. [PMID: 36046433 PMCID: PMC9400724 DOI: 10.37349/etat.2021.00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/21/2021] [Indexed: 11/19/2022] Open
Abstract
Metastasis is still the primary cause of cancer-related mortality. However, the underlying mechanisms of cancer metastasis are not yet fully understood. Currently, the epithelial-mesenchymal transition, metabolic remodeling, cancer cell intercommunication and the tumor microenvironment including diverse stromal cells, are reported to affect the metastatic process of cancer cells. Calcium ions (Ca2+) are ubiquitous second messengers that manipulate cancer metastasis by affecting signaling pathways. Diverse transporter/pump/channel-mediated Ca2+ currents form Ca2+ oscillations that can be decoded by Ca2+-binding proteins, which are promising prognostic biomarkers and therapeutic targets of cancer metastasis. This paper presents a review of the advances in research on the mechanisms underlying cancer metastasis and the roles of Ca2+-related signals in these events.
Collapse
Affiliation(s)
- Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Gang Liu
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Shuhong Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Jinghang Zhang
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, Henan, China
| |
Collapse
|
18
|
Zhu M, Xiao S. Expression profiles and prognostic values of BolA family members in ovarian cancer. J Ovarian Res 2021; 14:75. [PMID: 34078439 PMCID: PMC8170995 DOI: 10.1186/s13048-021-00821-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/07/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The BOLA gene family, comprising three members, is mainly involved in regulating intracellular iron homeostasis. Emerging evidence suggests that BolA family member 2 plays a vital role in tumorigenesis and hepatic cellular carcinoma progression. However, there was less known about its role in ovarian cancer. METHODS In the present study, we investigated the expression profiles, prognostic roles, and genetic alterations of three BolA family members in patients with ovarian cancer through several public databases, containing Oncomine and Gene Expression Profiling Interactive Analysis, Human Protein Atlas, Kaplan-Meier plotter and cBioPortal. Then, we constructed the protein-protein interaction networks of BOLA proteins and their interactors by using the String database and Cytoscape software. In addition, we performed the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment by the Annotation, Visualization, and Integrated Discovery database. Finally, we explored the mechanisms underlying BolA family members' involvement in OC by using gene set enrichment analysis. RESULTS The mRNA and protein expression levels of BOLA2 and BOLA3 were heavily higher in ovarian cancer tissues than in normal ovarian tissues. Dysregulated mRNA expressions of three BolA family members were significantly associated with prognosis in overall or subgroup analysis. Moreover, genetic alterations also occurred in three BolA family members in ovarian cancer. GO analysis indicated that BolA family members might regulate the function of metal ion binding and protein disulfide oxidoreductase activity. Gene set enrichment analysis indicated that BolA family members were mainly associated with oxidative phosphorylation, proteasome, protein export, and glutathione metabolism in ovarian cancer. CONCLUSION In brief, our finding may contribute to increasing currently limited prognostic biomarkers and treatment options for ovarian cancer.
Collapse
Affiliation(s)
- Mingyang Zhu
- Department of Nursing, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, 110004, People's Republic of China
| | - Shiqi Xiao
- Department of Nursing, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, 110004, People's Republic of China.
| |
Collapse
|
19
|
Store-operated Ca 2+ entry as a key oncogenic Ca 2+ signaling driving tumor invasion-metastasis cascade and its translational potential. Cancer Lett 2021; 516:64-72. [PMID: 34089807 DOI: 10.1016/j.canlet.2021.05.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 12/25/2022]
Abstract
Tumor metastasis is the primary cause of treatment failure and cancer-related deaths. Store-operated Ca2+ entry (SOCE), which is mediated by stromal interaction molecules (STIM) and ORAI proteins, has been implicated in the tumor invasion-metastasis cascade. Epithelial-mesenchymal transition (EMT) is a cellular program that enables tumor cells to acquire the capacities needed for migration and invasion and the formation of distal metastases. Tumor-associated angiogenesis contributes to metastasis because aberrantly developed vessels offer a path for tumor cell dissemination as well as supply sufficient nutrients for the metastatic colony to develop into metastasis. Recently, increasing evidence has indicated that SOCE alterations actively participate in the multi-step process of tumor metastasis. In addition, the dysregulated expression of STIM/ORAI has been reported to be a predictor of poor prognosis. Herein, we review the latest advances about the critical role of SOCE in the tumor metastasis cascade and the underlying regulatory mechanisms. We emphasize the contributions of SOCE to the EMT program, tumor cell migration and invasion, and angiogenesis. We further discuss the possibility of modulating SOCE or intervening in the downstream signaling pathways as a feasible targeting therapy for cancer treatment.
Collapse
|
20
|
Liang X, Zhang N, Pan H, Xie J, Han W. Development of Store-Operated Calcium Entry-Targeted Compounds in Cancer. Front Pharmacol 2021; 12:688244. [PMID: 34122115 PMCID: PMC8194303 DOI: 10.3389/fphar.2021.688244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is the major pathway of Ca2+ entry in mammalian cells, and regulates a variety of cellular functions including proliferation, motility, apoptosis, and death. Accumulating evidence has indicated that augmented SOCE is related to the generation and development of cancer, including tumor formation, proliferation, angiogenesis, metastasis, and antitumor immunity. Therefore, the development of compounds targeting SOCE has been proposed as a potential and effective strategy for use in cancer therapy. In this review, we summarize the current research on SOCE inhibitors and blockers, discuss their effects and possible mechanisms of action in cancer therapy, and induce a new perspective on the treatment of cancer.
Collapse
Affiliation(s)
- Xiaojing Liang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ningxia Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiansheng Xie
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
21
|
Store Operated Calcium Entry in Cell Migration and Cancer Metastasis. Cells 2021; 10:cells10051246. [PMID: 34069353 PMCID: PMC8158756 DOI: 10.3390/cells10051246] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023] Open
Abstract
Ca2+ signaling is ubiquitous in eukaryotic cells and modulates many cellular events including cell migration. Directional cell migration requires the polarization of both signaling and structural elements. This polarization is reflected in various Ca2+ signaling pathways that impinge on cell movement. In particular, store-operated Ca2+ entry (SOCE) plays important roles in regulating cell movement at both the front and rear of migrating cells. SOCE represents a predominant Ca2+ influx pathway in non-excitable cells, which are the primary migrating cells in multicellular organisms. In this review, we summarize the role of Ca2+ signaling in cell migration with a focus on SOCE and its diverse functions in migrating cells and cancer metastasis. SOCE has been implicated in regulating focal adhesion turnover in a polarized fashion and the mechanisms involved are beginning to be elucidated. However, SOCE is also involved is other aspects of cell migration with a less well-defined mechanistic understanding. Therefore, much remains to be learned regarding the role and regulation of SOCE in migrating cells.
Collapse
|
22
|
Chang Y, Roy S, Pan Z. Store-Operated Calcium Channels as Drug Target in Gastroesophageal Cancers. Front Pharmacol 2021; 12:668730. [PMID: 34012400 PMCID: PMC8126661 DOI: 10.3389/fphar.2021.668730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Gastroesophageal cancers, including tumors occurring in esophagus and stomach, usually have poor prognosis and lack effective chemotherapeutic drugs for treatment. The association between dysregulated store-operated calcium entry (SOCE), a key intracellular Ca2+ signaling pathway and gastroesophageal cancers are emerging. This review summarizes the recent advances in understanding the contribution of SOCE-mediated intracellular Ca2+ signaling to gastroesophageal cancers. It assesses the pathophysiological role of each component in SOCE machinery, such as Orais and STIMs in the cancer cell proliferation, migration, and invasion as well as stemness maintenance. Lastly, it discusses efforts towards development of more specific and potent SOCE inhibitors, which may be a new set of chemotherapeutic drugs appearing at the horizon, to provide either targeted therapy or adjuvant treatment to overcome drug resistance for gastroesophageal cancers.
Collapse
Affiliation(s)
- Yan Chang
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| | - Souvik Roy
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX, United States
| | - Zui Pan
- College of Nursing and Health Innovation, The University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
23
|
Hossian AKMN, Zahra FT, Poudel S, Abshire CF, Polk P, Garai J, Zabaleta J, Mikelis CM, Mattheolabakis G. Advanced bioinformatic analysis and pathway prediction of NSCLC cells upon cisplatin resistance. Sci Rep 2021; 11:6520. [PMID: 33753779 PMCID: PMC7985311 DOI: 10.1038/s41598-021-85930-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 03/05/2021] [Indexed: 11/08/2022] Open
Abstract
This study aims to identify pathway involvement in the development of cisplatin (cis-diamminedichloroplatinum (II); CDDP) resistance in A549 lung cancer (LC) cells by utilizing advanced bioinformatics software. We developed CDDP-resistant A549 (A549/DDP) cells through prolonged incubation with the drug and performed RNA-seq on RNA extracts to determine differential mRNA and miRNA expression between A549/DDP and A549 cells. We analyzed the gene dysregulation with Ingenuity Pathway Analysis (IPA; QIAGEN) software. In contrast to prior research, which relied on the clustering of dysregulated genes to pathways as an indication of pathway activity, we utilized the IPA software for the dynamic evaluation of pathway activity depending on the gene dysregulation levels. We predicted 15 pathways significantly contributing to the chemoresistance, with several of them to have not been previously reported or analyzed in detail. Among them, the PKR signaling, cholesterol biosynthesis, and TEC signaling pathways are included, as well as genes, such as PIK3R3, miR-34c-5p, and MDM2, among others. We also provide a preliminary analysis of SNPs and indels, present exclusively in A549/DDP cells. This study's results provide novel potential mechanisms and molecular targets that can be explored in future studies and assist in improving the understanding of the chemoresistance phenotype.
Collapse
Affiliation(s)
- A K M Nawshad Hossian
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Sagun Poudel
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA
| | - Camille F Abshire
- Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paula Polk
- Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Jone Garai
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Jovanny Zabaleta
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, USA.
| |
Collapse
|
24
|
Naffa R, Padányi R, Ignácz A, Hegyi Z, Jezsó B, Tóth S, Varga K, Homolya L, Hegedűs L, Schlett K, Enyedi A. The Plasma Membrane Ca 2+ Pump PMCA4b Regulates Melanoma Cell Migration through Remodeling of the Actin Cytoskeleton. Cancers (Basel) 2021; 13:cancers13061354. [PMID: 33802790 PMCID: PMC8002435 DOI: 10.3390/cancers13061354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/08/2021] [Accepted: 03/14/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Earlier we demonstrated that the plasma membrane Ca2+ pump PMCA4b inhibits migration and metastatic activity of BRAF mutant melanoma cells, however, the exact mechanism has not been fully understood. Here we demonstrate that PMCA4b acted through actin cytoskeleton remodeling in generating a low migratory melanoma cell phenotype resulting in increased cell–cell connections, lamellipodia and stress fiber formation. Both proper trafficking and calcium transporting activity of the pump were essential to complete these tasks indicating that controlling Ca2+ concentration levels at specific plasma membrane locations such as the cell front played a role. Our findings suggest that PMCA4b downregulation is likely one of the mechanisms that leads to the perturbed cancer cell cytoskeleton organization resulting in enhanced melanoma cell migration and metastasis. Abstract We demonstrated that the plasma membrane Ca2+ ATPase PMCA4b inhibits migration and metastatic activity of BRAF mutant melanoma cells. Actin dynamics are essential for cells to move, invade and metastasize, therefore, we hypothesized that PMCA4b affected cell migration through remodeling of the actin cytoskeleton. We found that expression of PMCA4b in A375 BRAF mutant melanoma cells induced a profound change in cell shape, cell culture morphology, and displayed a polarized migratory character. Along with these changes the cells became more rounded with increased cell–cell connections, lamellipodia and stress fiber formation. Silencing PMCA4b in MCF-7 breast cancer cells had a similar effect, resulting in a dramatic loss of stress fibers. In addition, the PMCA4b expressing A375 cells maintained front-to-rear Ca2+ concentration gradient with the actin severing protein cofilin localizing to the lamellipodia, and preserved the integrity of the actin cytoskeleton from a destructive Ca2+ overload. We showed that both PMCA4b activity and trafficking were essential for the observed morphology and motility changes. In conclusion, our data suggest that PMCA4b plays a critical role in adopting front-to-rear polarity in a normally spindle-shaped cell type through F-actin rearrangement resulting in a less aggressive melanoma cell phenotype.
Collapse
Affiliation(s)
- Randa Naffa
- Department of Transfusiology, Semmelweis University, H-1089 Budapest, Hungary; (R.N.); (S.T.)
| | - Rita Padányi
- Department of Biophysics and Radiation Biology, Semmelweis University, H-1094 Budapest, Hungary;
| | - Attila Ignácz
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary; (A.I.); (K.S.)
| | - Zoltán Hegyi
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudosok krt.2, H-1117 Budapest, Hungary; (Z.H.); (B.J.); (L.H.)
| | - Bálint Jezsó
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudosok krt.2, H-1117 Budapest, Hungary; (Z.H.); (B.J.); (L.H.)
| | - Sarolta Tóth
- Department of Transfusiology, Semmelweis University, H-1089 Budapest, Hungary; (R.N.); (S.T.)
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | | | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Magyar Tudosok krt.2, H-1117 Budapest, Hungary; (Z.H.); (B.J.); (L.H.)
| | - Luca Hegedűs
- Department of Thoracic Surgery, Ruhrlandklinik, University Clinic Essen, 45239 Essen, Germany;
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary; (A.I.); (K.S.)
| | - Agnes Enyedi
- Department of Transfusiology, Semmelweis University, H-1089 Budapest, Hungary; (R.N.); (S.T.)
- Correspondence:
| |
Collapse
|
25
|
Azizi L, Cowell AR, Mykuliak VV, Goult BT, Turkki P, Hytönen VP. Cancer associated talin point mutations disorganise cell adhesion and migration. Sci Rep 2021; 11:347. [PMID: 33431906 PMCID: PMC7801617 DOI: 10.1038/s41598-020-77911-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Talin-1 is a key component of the multiprotein adhesion complexes which mediate cell migration, adhesion and integrin signalling and has been linked to cancer in several studies. We analysed talin-1 mutations reported in the Catalogue of Somatic Mutations in Cancer database and developed a bioinformatics pipeline to predict the severity of each mutation. These predictions were then assessed using biochemistry and cell biology experiments. With this approach we were able to identify several talin-1 mutations affecting integrin activity, actin recruitment and Deleted in Liver Cancer 1 localization. We explored potential changes in talin-1 signalling responses by assessing impact on migration, invasion and proliferation. Altogether, this study describes a pipeline approach of experiments for crude characterization of talin-1 mutants in order to evaluate their functional effects and potential pathogenicity. Our findings suggest that cancer related point mutations in talin-1 can affect cell behaviour and so may contribute to cancer progression.
Collapse
Affiliation(s)
- Latifeh Azizi
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alana R Cowell
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, Kent, UK
| | - Vasyl V Mykuliak
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, CT2 7NJ, Kent, UK.
| | - Paula Turkki
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
| | - Vesa P Hytönen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
| |
Collapse
|