Open Access

Calcium signals and potential therapy targets in ovarian cancer (Review)

  • Authors:
    • Fengying Deng
    • Mengyu Fu
    • Chenxuan Zhao
    • Jiahui Lei
    • Ting Xu
    • Bingyu Ji
    • Hongmei Ding
    • Yueming Zhang
    • Jie Chen
    • Junlan Qiu
    • Qinqin Gao
  • View Affiliations

  • Published online on: September 12, 2023     https://doi.org/10.3892/ijo.2023.5573
  • Article Number: 125
  • Copyright: © Deng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer (OC) is a deadly disease. The poor prognosis and high lethality of OC are attributed to its high degrees of aggressiveness, resistance to chemotherapy and recurrence rates. Calcium ion (Ca2+) signaling has received attention in recent years, as it appears to form an essential part of various aspects of cancer pathophysiology and is a potential therapeutic target for OC treatment. Disruption of normal Ca2+ signaling pathways can induce changes in cell cycle progression, apoptosis, proliferation and migration and invasion, leading to the development of the malignant phenotype of tumors. In the present review, the main roles of ion channel/receptor/pump‑triggered Ca2+ signaling pathways located at the plasma membrane and organelle Ca2+ transport in OC are summarized. In addition, the potential of Ca2+ signaling as a novel target for the development of effective treatment strategies for OC was discussed. Furthering the understanding into the role of Ca2+ signaling in OC is expected to facilitated the identification of novel therapeutic targets and improved clinical outcomes for patients.

1. Introduction

Ovarian cancer (OC) is considered to be the most lethal of the three major types of gynecological cancer known, which also include cervical and endometrial cancer (1). Different histological subtypes of OC can be distinguished by their unique combination of risk factors, cellular origins, molecular profile, clinical characteristics and response to treatment. Epithelial ovarian cancer (EOC) accounts for 90% of all ovarian tumors, which can then be further subdivided into the following four subtypes: Plasmacytoma, endometrioid carcinoma, clear cell carcinoma and mucinous carcinoma. In total, ~10% ovarian malignancies are classified as non-epithelial, which includes germ cell tumors, gonadal mesenchymal tumors and metastatic tumors (2,3). As the principal female reproductive organ, the ovaries are in charge of oogenesis, female sex hormone production and secretion. Previous research indicates that OC originates in the fallopian tubes rather than the ovary, as previously thought (4,5). Ovarian malignancies are difficult to detect in the early stages due to their position in the peritoneal cavity and being shielded and they are frequently discovered in the late stages. However, detecting early atypical OC remains difficult due to molecular similarities between cells in the fallopian tubes, ovaries and peritoneum (4). Ovarian cancer is exceedingly common, second only to breast cancer in terms of incidence and it has the greatest mortality rate among the three primary gynecologic malignancies, posing a substantial threat to women. OC is a very aggressive cancer that is resistant to treatment, has a high recurrence rate and has a low 5-year survival rate (6-9). In comparison to other gynecological cancers, research in this field is wanting and needs more attention and exploration. As a result, identifying early diagnostic markers and developing further focused therapy options for this cancer is critical.

Calcium ion (Ca2+) is an essential second messenger that participates in a wide range of critical physiological processes (10). Signal transduction involving Ca2+ is essential for a wide variety of biological functions, including proliferation, differentiation, growth and apoptosis. Ca2+ flow through intracellular Ca2+ ([Ca2+]i) channels are necessary for the transition from the G1/S phase to mitosis in the cell cycle. By contrast, Ca2+ deficiency can halt cell cycle at the G0/G1 and S phases (11). A number of ovarian pathologies, such as OC, typically result from the dysregulation of plasma membrane-based and organelle-based Ca2+ signaling mechanisms (12). Ca2+ levels have a profound effect on the physiology of the female reproductive system, especially the ovary. [Ca2+]i concentrations can regulate almost every cellular process currently known, from energy production and cellular metabolism to phenotypic development. In particular, novel ideas of ovarian oncogenesis involving altered Ca2+ signaling have been steadily proposed over the past decade (12). Variations in [Ca2+]i concentrations can translate through the cell to effect distant regions, to modulate Ca2+ signaling pathways that can regulate cell cycle progression, apoptosis, proliferation and metastasis. This contributes to the promotion of more malignant tumor phenotypes (13-16). The present review summarized the current knowledge on the role of Ca2+ channels and Ca2+ signaling dysfunction in OC development. In addition, how plasma membrane Ca2+ channels, [Ca2+]i channels, Ca2+ transport proteins, mitochondrial Ca2+ transport, the S100 family and extracellular factors can regulate OC development and progression are comprehensively reviewed. The potential roles of these ion channels as therapeutic targets for the diagnosis and treatment of OC are also discussed. The present review not only discussed the most recent findings in the field, but also aimed to swiftly propose entry points for developing treatment methods and future research avenues.

2. Plasma membrane Ca2+ channels

Transient receptor potential (TRP) channels

TRP channels form a group of non-selective cation channels that allow Ca2+ to permeate. These channels can be categorized into seven groups based on the similarity of their amino acid sequences: TRP canonical (C), TRP vanilloid (V), TRP mestatin (M), TR polycystic protein (PP), TRP mucin (ML), TRP anchor protein (A) and TRP no mechanoreceptor potential C (N) (17). Studies have shown that mutations in the TRP gene can affect the spatial and temporal distribution of Ca2+, which can in turn promote the proliferation and spread of cancer cells (18-22). In OC, the TRPC, TRPM and TRPV families of TRP channels have received the most attention.

TRPC is a subfamily of TRP channels that can be activated by hormones and growth factors, which can mediate Ca2+ transport (19). TRPC1, which is widely expressed, is involved in various physiological processes, including cancer development (20), cell proliferation, differentiation, migration, membrane permeability, fluid secretion and apoptosis (21). In OC, the mRNA expression levels of TRPC1 have been reported to be significantly decreased, especially in drug-resistant cases. In addition, this decrease may be associated with higher histological tumor grades and drug resistance (22).

TRPC3 is an important member of the TRPC family and has been shown to be involved in tumor proliferation, metastasis and invasion in OC (23). The protein levels of TRPC3 are considerably higher in human OC samples compared with those in normal ovarian tissue (23-26). Relapse, metastasis and a poor prognosis in human OC have all been associated with high TRPC3 expression (24-26). Downregulating TRPC3 expression in human OC cells leads to a reduction in cell proliferation through the suppression of epidermal growth factor-induced Ca2+ influx, dephosphorylation of cell division cycle 2 and Ca2+/calmodulin (CaM)-dependent protein kinase IIα, in addition to prolonged M phase progression (23). By contrast, follicle-stimulating hormone (FSH), estrogen and long chain noncoding RNA (lncRNA) small nucleolar RNA host gene (SNHG)3 can upregulate TRPC3 expression, which contributes to the progression of human OC (24-26). Additionally, phospholipase A2-activated protein has been found to inhibit OC cell invasion and tumor metastasis via decreasing the levels of m6A-modified TRPC3 mRNA by inhibiting methyltransferase-like 3 expressions (27). Therefore, TRPC3 probably serves a significant role in the progression of human OC, rendering it a potential diagnostic and therapeutic target for this malignancy. In particular, TRPC3 downregulation in aging fibroblasts has been documented to increase endoplasmic-reticulum (ER)-mitochondrial Ca2+ transfer, which enhances oxidative phosphorylation in mitochondria and promotes the release of tumor-promoting molecules such as interleukin-8 and matrix metalloproteinase 1 (28). However, it remains unclear whether this mechanism would have a counteracting effect on the downregulation of TRPC3 in the treatment pathway for OC, for which further research is required.

TRPV1 is a non-selective cation channel that belongs to the TRP channels family. It is particularly sensitive to capsaicin, heat, protons, lipids, phorbols and phosphorylation (29,30). Aberrant expression of TRPV1 has been associated with malignant tumors in the female reproductive system, including breast, ovarian and cervical cancer (31-33). During the development and progression of OC, Han et al (33) previously found that high TRPV1 expression was present in the tissues of ovarian malignancies, particularly in the plasma-type EOC. Therefore, it was proposed that high TRPV1 expression can be applied as an independent prognostic factor for the overall survival of patients with OC. In addition, Han et al (33) found that the expression of PTEN, a dual-lipoprotein phosphatase, was negatively correlated with that of TRPV1 expression in late-stage OC, whereby high TRPV1/low PTEN was confirmed by Cox regression analysis to be a significant predictor of prognosis in patients with OC. Subsequent in vitro functional studies revealed that inhibiting TRPV1 can prevent the development of OC cells (33). In another previous study, Wang et al (34) found that the TRPV1 antagonist DWP05195 significantly suppressed the proliferation of five human OC cell lines A2780, SKOV3, OVCAR3, TOV-21G and Hey8A by inducing C/EBP homologous protein expression, ER stress and apoptosis through the accumulation of reactive oxygen species (ROS). Cisplatin, which is used to treat OC, is known to increase the risk of cytotoxicity. Ursolic acid treatment has been reported to effectively prevent the development of cytotoxicity by inhibiting the TRPV1/-Ca2+/calpain signaling pathway in the cochlea (35). At present, TRPV1 is one of the most extensively researched TRP channels. However, the mechanism underlying its involvement in the development of OC requires further study. These promising findings provide a path for the future investigation of TRPV1 as a possible therapeutic target for OC.

TRPV2 is typically found inside the cell membrane and has been shown to regulate a number of pathological processes, including cancer, through a signaling route that occurs outside the membrane (36). TRPV2 activation has been found to promote cell migration and cell invasiveness, while the absence or modification of TRPV2-mediated signaling can lead to uncontrolled proliferation and apoptotic (37). Cannabidiol (CBD) has been shown to bind to the TRPV2 channel and has been associated with the dysregulation of proliferation, cell differentiation and invasion in a variety of cancer cell lines and animal models (36). CBD treatment of endometrial cancer has been reported to reverse the cytotoxic effects of chemotherapeutic agents, which is also enhanced by TRPV2 overexpression. Antitumor effects of CBD on OC have been previously observed, both as a potential monotherapy and in combination with conventional chemotherapeutic agents (36). Using PLGA-microparticles as carriers of CBD in combination with paclitaxel, the therapeutic efficacy for OC was increased without any worsening of paclitaxel-related side effects (38). However, whether CBD can improve chemotherapy prognosis for patients with OC by targeting TRPV2 remains to be elucidated. Additionally, TRPV2 also been proposed to be a novel marker for type II EOC, especially for the plasmacytic subtypes and high-grade tumors (39). Further research is required to fully establish the role of TRPV2 in OC.

TRPV4 is a non-selective mechanosensitive transmembrane Ca2+-permeable cation channel (40). Increased expression of TRPV4 in OC has been associated with poorer overall survival, disease-specific survival, disease-free interval and progression-free interval (41). Furthermore, patients with OC who express higher levels of TRPV4 may be more resistant to the chemotherapeutic drugs cisplatin and oxaliplatin. Zhang et al (42) previously reported that screened high TRPV4 expression was associated with poor prognosis in patients with ovarian serous cystadenocarcinoma and also demonstrated by Cox regression analysis that TRPV4 was the most probable therapeutic target for ovarian serous cystadenocarcinoma. The role of TRPV4 in the dysregulation of cell migration and adhesion may be crucial for the poor prognosis in OC. This is because it has been known to physiologically regulate endothelial vasodilatation and shear stress sensing, cell migration and skin adhesion junctions (43).

TRPV6 is a highly selective Ca2+ channel with its own spontaneous activity that depends on intracellular and extracellular Ca2+ concentration (44,45). Its overexpression has been observed in several types of cancer, including prostate, breast and ovarian cancers, and is strongly associated with tumorigenesis, metastasis and prognosis (46,47). Clear cell carcinoma, endometrioid carcinoma, high-grade plasmacytoma, low-grade plasmacytoma and mucinous carcinoma all possess higher TRPV6 mRNA and protein expression levels compared with those in normal tissue, suggesting that targeting TRPV6 channels may inhibit the growth of tumor cells in OC xenograft models and that TRPV6 is a viable target for OC therapy (48). Lidocaine, an anesthetic at concentrations below clinical levels, has been shown to reduce TRPV6 expression in OC cells, to prevent cell invasion and migration (49). By contrast, lapatinib has been observed to suppress TRPV6 mRNA expression in breast and lung adenocarcinoma cells, but has not been studied in OC cells (50).

TRPM2 is a cation channel that allows the passage of Na+, K+ and Ca2+, which are in turn regulated by [Ca2+]i in a manner that is dependent on the CaM-binding IQ-like motif (51). TRPM2-antisense (AS) is a lncRNA that acts as an antisense RNA for TRPM2 (52). It has been discovered that TRPM2-AS expression is increased in OC tissues and cells, where it may serve a role in cell proliferation, colony formation, cell migration and invasion in vitro. By primarily activating syndecan 3 expression by sponging microRNA (miR-)138-5p, TRPM2-AS was documented to promote tumor growth by OC cells and increase resistance to cisplatin (53). As a novel treatment target for OC, focusing on TRPM2 may prove fruitful.

TRPM7 is a channel that allows the passage of Ca2+ and magnesium ions, where it has been found to be abnormally expressed in various types of cancer, including OC (54,55). It probably serves an important role in the carcinogenic process and is strongly associated with tumorigenesis, metastasis and prognosis in patients with OC (56,57). Previous studies have shown that downregulating TRPM7 activity with inhibitors of 5-lipoxygenase and [Ca2+]i chelators can inhibit OC epithelial-mesenchymal (EMT) transition and metastasis by inhibiting the Ca2+-related PI3k/AKT activation (58,59). Therefore, TRPM7 is considered as a potential therapeutic target for the intervention of OC.

The present section discussed the TRP channels that are most likely associated with OC, including TRPC1, TRPC3, TRPV1, TRPV2, TRPV4, TRPV6, TRPM2 and TRPM7. While these channels have been associated with various physiological processes, such as cell proliferation, apoptosis, migration and invasion, in addition to [Ca2+]i regulation, the precise mechanisms underlying their roles in OC development remain to be elucidated. Additionally, there are other TRP channels associated with cancer, but their links to OC have not been confirmed. Therefore, further research is required to fully understand the role and mechanism of TRP channels in the development and therapeutic intervention of OC. Fig. 1 and Table I summarize the TRP channels discussed and their possible associations with OC.

Table I

TRP channels in ovarian cancer.

Table I

TRP channels in ovarian cancer.

TRP channelsExpressionEffects(Refs.)
TRPC1DecreasedLower TRPC1 expression may be associated with drug resistance and a high histological tumor grade.(22)
TRPC3IncreasedHigher TRPC3 expression levels are correlated with early relapse, metastasis and worse prognosis.(23)
TRPV1IncreasedHigher TRPV1 expression are correlated with a poor overall survival.(33)
TRPV4IncreasedHigh expression of TRPV4 is associated with poor overall survival, disease specific survival, disease-free and progression free intervals, and increases drug resistance.(42,43)
TRPV6IncreasedThere is a strong relationship between tumorigenesis, metastasis, and prognosis.(50)
TRPM2IncreasedTRPM2-AS promotes cell proliferation, colony formation, Cell migration and cell invasion in vitro.(55)
TRPM7IncreasedPromotes pelvic metastasis of ovarian cancer cells, resulting in a poor prognosis.(59,60)

[i] TRP, transient receptor potential; TRPM2-AS, TRPM2 antisense RNA.

Voltage-gated Ca2+Channels (VGCCs)

VGCCs, including T-type Ca2+ channels (TTCC) and L-type Ca2+ channels (LTCC), play a significant role in regulating the physiological activities of cells (60). Studies highlight that the TTCC-mediated influx of Ca2+ regulates cell proliferation, which has been associated with different types of cancer, including OC (61,62). Blocking TTCC expression with NNC 550396, mibefradil or TTCC subunits (Cav3.1/3.2) downregulation impairs the proliferation of OC cells, increases G0/G1 phase distribution and slows down OC formation in nude mice (61). TTCC inhibitors, such as mibefradil and related 3,4-dihydroquinazoline derivatives, alter the normal progression of cells through the cell cycle, leading to a similar decline in OC cell proliferation (62). Survivin is an antiapoptotic protein encoded by the BIRC5 gene and is also downregulated during this process (63). Inhibiting TTCC in OC cells not only suppresses growth and increases apoptosis but also downregulates the expression of BIRC5 (63). Additionally, the effectiveness of platinum agents in treating OC can be improved by inhibiting Survivin (63). TTCC blocker and platinum treatment, in combination, promote apoptosis of OC cells and reduce ectopic metastasis of platinum-resistant tumors in mice, providing a model to investigate OC metastasis in humans (63,64). In the study by Fornaro et al (65), it was found that the expression of TTCC genes (three isoforms: CACNA-1G, CACNA-1H and CACNA-1I) were correlated with overall survival in patients with tumors, especially in gastric cancer. The correlation between the expression of all CACNA genes and overall survival when considering staging was also significant in OC, demonstrating that altered CACNA gene expression correlates with tumor prognosis and promising for further evaluation in OC.

In addition, when the LTCC is affected by adverse factors, such as serum gonadotropins and lysophosphatidic acid (LPA), resulting in its abnormal activation of Ca2+ inward flow, it is strongly associated with the proliferation and metastasis of OC cells (66,67). Nifedipine (a LTCC blocker) could inhibit LPA-induced OC cell migration and adhesion (67). OC stem cells are a major contributor to drug resistance in OC patients. LTCC blockers (manidipine, lacidipine, benidipine and lomepizine) and trimebutine maleate could inhibit the viability and proliferation of OC stem cells by downregulating the expression of the LTCC gene, thereby inducing apoptosis (68,69). CACNA1C, as an important type of LTCC ion transmembrane channel, plays regulatory roles in the development and progress of multiple tumors. Chang and Dong (70) revealed that CACNA1C could be a prognostic predictor of overall survival in OC and it was closely related to immunity. In conclusion, the current research indicates that TTCC and LTCC play an important role in OC cell proliferation, cell cycle progression and metastasis and is also linked to OC prognosis (Table II).

Table II

Alterations in Ca2+ signaling in ovarian cancers.

Table II

Alterations in Ca2+ signaling in ovarian cancers.

Calcium signalsExpressionEffects(Refs.)
VGCCsTTCCIncreasedPlays a reinforcing role in ovarian cancer cell proliferation, cell cycle progression, and apoptosis evasion.(63)
LTCCIncreasedIncreases ovarian cancer cell proliferation and metastasis.(68)
CRACOrai & STIMIncreasedStimulates tumor growth and metastasis and enhances tumor drug resistance.(75)
BKCaKCNMA1IncreasedIs associated with the malignancy and poor prognosis of the cancer.(83)
KCNMA1DecreasedEnhances the drug resistance of ovarian cancer.(84)
IP3RsIP3R1IncreasedPossesses pro-proliferative and anti-apoptotic effects.(104)
IP3R2IncreasedPromotes development and progression of ovarian cancer.(112)
RyRsRyR1IncreasedIs involved in the onset and development of ovarian cancer.(112)
RyR2DecreasedPromotes tumorigenesis and development.
Ca2+-ATPaseSERCAIncreasedInvolved in the progression of ovarian cancer.(121)
PMCAIncreasedDisrupts calcium homeostasis and contributes to the progression of drug-resistant cancer cells.(126)
MCUMICU1IncreasedMICU1 overexpression correlates with poor overall survival and resistance to chemotherapy.(132)

[i] VGCC, Voltage-gated Calcium Channel; TTCC, T-type Ca2+ channel; LTCC, L-type Ca2+ channel; CRAC, Ca2+ Release-Activated Ca2+ Channel; STIM, stromal interaction molecule; BKCa, large conductance Ca2+-activated K+; KCNMA1, Ca2+-activated potassium channel subunit α-1; IP3R, inositol 1,4,5-trisphosphate receptor; RyR, lysine receptor; SERCA, sarco/endoplasmic reticulum; PMCA, plasma membrane; MCU, mitochondrial Ca2+ uniporter; MICU1, mitochondrial Ca2+ uniporter regulator 1.

Ca2+ release-activated Ca2+ channel (CRAC)

The CRAC channel, composed of Ca2+ release-activated Ca2+ channel protein 1 (Orai1) and stromal interaction molecule (STIM), serves a vital role in regulating Ca2+ signaling and gene expression in cells by activating store-operated Ca2+-entry (71). Previous studies suggest that targeting this channel can be a potential therapeutic strategy for treating cancer. Khan et al (72) reported that blocking the CRAC channel, which is overexpressed in various cancer cells and tissues, may benefit patients with cancer. Hypoxia inducible factor (HIF)-1α has been found to promote tumor growth and metastasis by elevating expression the levels of Orai1 and STIM1 in OC cells after exposure to placental growth factor (73). In addition, Schmidt et al (74) found that Orai1 and STIM1 expression levels were slightly higher in drug-resistant OC cells, suggesting the potential involvement of CRAC channel in supporting the survival of OC cells. Other studies have demonstrated that upregulated Orai1 expression in OC cells could lead to increased cell proliferation and metastasis; meanwhile, silencing Orai1 expression was demonstrated to inhibit these aforementioned effects (75,76). Therefore, currently available evidence suggests that CRAC channel likely serves a role in the initiation and progression of OC, contributing to poorer patient outcomes (Table II). Although there is limited data on other subtypes of Orai/STIM and few clinical trials have used CRAC channel blockers, targeting this channel may be a promising approach for developing OC therapies.

Large conductance Ca2+-activated K+ (BKCa) channel

The BKCa channel has been implicated in human cancer development, including OC, by contributing to cell cycle disruption, proliferation and migration (77,78). The BKCa channel opener NS1619 has been found to reduce proliferation while inducing apoptosis in OC cells by upregulating death-inducing proteins (such as P53, P21 and Bax) (79). The α-subunit of BKCa channel is encoded by the Ca2+-activated potassium channel subunit α-1 (KCNMA1) gene, which has been shown to serve a role in the formation of macromolecular signaling complexes through the action of local Ca2+ introductory channels (80). The BKCa channel subunit KCNMA1 contributes to macromolecular signaling complexes, whereby KCNMA1 amplification is associated with higher proliferation rates and higher degrees of malignancy in ovarian, endometrial and breast cancers (81,82). However, a study reported that knocking out KCNMA1 expression increases cisplatin resistance in OC cells (68). A recent study found that trimebutine maleate inhibits the viability of OC stem cells by targeting the BKCa channel and can prevent drug resistance and recurrence in OC (69). Further investigation into the precise mechanism of BKCa channel-regulated proliferation, apoptosis and resistance in OC cells is necessary to determine their potential as biomarkers or therapeutic targets for OC (Table II).

G-protein-coupled receptors (GPCRs)

GPCRs form a class of receptor proteins that can activate G proteins to elicit cascade reactions affecting a wide range of biological functions including cancer progression (83,84). GPCRs can exert different types of effects on OC (Table III). The Ca2+-sensing receptor (CaR) is a GPCR that mediates Ca2+ signaling and disrupts normal epidermal differentiation by sensing extracellular Ca2+ (85). The CaR rs17251221 G allele has reported protective effects, reducing the risk of OC development (86,87). By contrast, lysophosphatidylglycerol-induced proliferation and migration of human OC cells are mediated by pertussis toxin-sensitive GPCRs (88). Previous studies have highlighted the role of G protein-coupled estrogen receptor (GPER) in OC pathogenesis, where its high expression is associated with malignant OC, tumor cell invasion and poorer patient survival (89,90). GPER activation may be involved in OC initiation and progression, although GPER has also been reported as possessing anti-cancer properties (91,92). Notably, OC cells treated with GPER-specific agonist G1 exhibited increased levels of apoptosis and impeded cancer progression (93). GPCRs are primarily stimulated by most neurotransmitters and inflammation-related ligands, which can in turn promote OC proliferation, metastasis and invasion, as reviewed by Predescu et al (94). Despite the scarcity of animal models and clinically relevant data, GPCRs have emerged as promising therapeutic targets for OC (94). It is reported that 2-thioureidothiophene-3-carboxylates (TUTPs), a novel class of antagonists for the GPCR C-X-C chemokine receptor type 2, effectively inhibits C-X-C motif ligand 8-mediated cell migration while exhibiting a synergistic effect with doxorubicin on OC cells (95). These findings suggest that TUTPs hold promise as potential anticancer agent for OC treatment, highlighting the potential of GPCR-based approaches for OC therapy.

Table III

Alterations in Ca2+ exchange proteins in ovarian cancer.

Table III

Alterations in Ca2+ exchange proteins in ovarian cancer.

Calcium signalsExpressionEffects(Refs.)
GPCRsCaRDecreasedCauses changes in the physiology of tumor cells and acceleratestumor progression.(88)
GPERIncreasedInduces metastasis and invasion of tumor cells, with a poor prognosis.(91-94)
S100 family S100A1/2/4/5/6/7/10/11/13/14/16IncreasedCorrelated with lymph node metastasis, FIGO staging and tumor grade.(155,157,178)
S100B/S100PIncreasedUpregulation linked to tumor growth, survival, prognosis and resistance to cancer drugs.(174,176)
TROPTROP2IncreasedEnhances the ability of ovarian cancer cells to proliferate, invade and migrate.(177)
CaMKKCaMKK2IncreasedEnhances the ability of ovarian cancer cells to proliferate, invade and migrate.(181)

[i] GPCR, G-protein-coupled receptor; CaR, Ca2+-sensing receptor; GPER, G protein-coupled estrogen receptor; FIGO, International Federation of Gynecology and Obstetrics; TROP, tumor-associated calcium signal transducer; CaMKK, Ca2+/calmodulin-dependent protein kinase.

3. Intracellular Ca2+ channels and transporters

Inositol 1,4,5-triphosphate receptor (IP3R) and ryanodine receptors (RyR) channels

IP3Rs are a family of Ca2+-releasing channels located in the ER membrane (96). There are three main isoforms of IP3Rs: IP3R type 1 (IP3R1), InsP3R type 2 (IP3R2) and IP3R type 3 (IP3R3). They serve a crucial role in the regulation of Ca2+ release from the ER and sarcoplasmic reticulum and are expressed to varying degrees in different mammalian tissues (97-99). When activated, IP3Rs regulate the release of Ca2+ from the ER to either the mitochondria and/or cytoplasm, where they serve an essential role in regulating cellular metabolism and survival (100). All three isoforms of IP3R can be detected in ovarian tissue sections from normal experimental animals and OC A2780 cells and some studies suggest that IP3R is emerging as a key locus for the regulation of pro- and anti-apoptotic factors (101,102). Hypoxia has been shown to control the intensity of Ca2+ signaling in cancer cells through IP3Rs (103). In addition, Lencesova et al (104) previously demonstrated that hydrogen sulfide causes ER stress and apoptosis under hypoxic conditions, suggesting that IP3Rs are closely associated with apoptosis in OC cells. Additionally, stable TAT-fused IP3R1-derived peptides can increase cisplatin-induced Ca2+ flux from the ER into the cytosol and mitochondria, sensitizing OC cells to cisplatin by targeting the BH4 structural domain of Bcl-2 (Table II) (105,106).

Compared with IP3R1, IP3R3 has been shown to exert both pro-proliferative and anti-apoptotic effects on cancer cells. Elevated IP3R3 expression levels have been observed to enhance the migratory and invasive properties of cancer cells by increasing mitochondrial metabolism and driving anabolic pathways (107,108). By contrast, a recent study found that OC cells become more resistant to chemotherapy-induced apoptosis after IP3R3-mediated Ca2+ flux to mitochondria was blocked (109). Therefore, further research is necessary to elucidate the function of IP3R3 in OC cells. By comparison, IP3R2 has received less attention. Nonetheless, increased expression of the IP3R2 gene was observed in iron-treated epithelial OC cells and cisplatin-resistant cells of the same cell line, indicating its probable role in the development and progression of OC (110).

RyRs are also members of the Ca2+-releasing channels family located on the ER membrane, of which three known isoforms (RyR1, RyR2 and RyR3) exist. RyRs are widely expressed and mediate Ca2+ release from intracellular membrane compartments, leading to transient and reversible alterations in cytoplasmic and ER Ca2+ levels (111). RyRs have been found to be useful in determining the severity of malignant diseases and their prognosis, as they may serve a role in the onset and progression of prostate, breast and head and neck cancers (112-114). In OC cells, RyRs can interfere with the estrogen receptor α (ERα-)/PLCγ-/IP3R pathway by altering the activity of ERα biomodulators (115). Furthermore, RyRs have been reported to regulate the activation of the unfolded protein response in OC cells and in turn their sensitivity to paclitaxel and adriamycin (116). A previous study found that both epithelial OC cells (MDAH-2774) and cisplatin-resistant OC cells of the same cell line (MDAH-2774/DDP) showed increased RyR1 gene expression after iron treatment, while only EOC cells (MDAH-2774) showed decreased RyR2 mRNA levels (110). This suggests that RyRs can affect the progression of OC by regulating Ca2+ levels in OC cells through multiple mechanisms (Table II).

IP3Rs and RyRs are considered to serve central roles in [Ca2+]i movement and have an important and complex role in the development of types of cancer. However, the role and mechanism of IP3Rs and RyRs in OC remain poorly understood. Further studies are therefore necessary to gain an in-depth understanding into the occurrence, development and prognosis of OC. Information from these studies are likely to provide novel insights into possible therapeutic approaches for OC by targeting the ER-related Ca2+-releasing channels.

Ca2+-ATPases

Ca2+-ATPases or Ca2+ pumps regulate Ca2+ homeostasis and are essential for reproduction. However, their dysregulation can interfere with the production of sex hormones and disrupt normal ovarian physiology (117,118). To date, three major families of Ca2+-ATPases have been identified: Those located on the sarco/endoplasmic reticulum (SERCA); on the plasma membrane (PMCA); and on the secretory pathway (SPCA) (118). In OC tissues, aberrant SERCA expression has been observed, the inhibition of which has been revealed to increases cytoplasmic Ca2+ concentration, resulting in OC cell apoptosis. This suggests that SERCA serves a role in OC progression (119,120). Seo et al (120) found that curcumin inhibits SERCA activity to disrupt [Ca2+]i homeostasis, which promotes apoptosis in OC cells. Transmembrane and coiled-coil domains 1 (TMCO1), which is essential for ovarian follicle development and female fertility in granulosa cells, is regulated by SERCA (121). The disruption of TMCO1 was demonstrated to cause Ca2+ overload in the ER and increased ROS levels in granulosa cells, which ultimately caused follicular dysgenesis. These phenomena have been associated with various ovarian-associated pathological conditions, such as OC (121,122).

Ovarian granulosa cells rely on PMCA to regulate their [Ca2+]i concentrations in response to basic fibroblast growth factor (bFGF), which suggests that bFGF regulates PMCA as part of an anti-apoptotic mechanism in ovarian granulosa cells (123,124). Iron treatment was found to upregulate the mRNA expression of both PMCA1 and PMCA3 in cisplatin-resistant epithelial OC cells, suggesting that PMCA is an independent pathway of drug resistance in OC cells (110). Additionally, the mRNA expression profiles of Ca2+ homeostasis-associated genes (SERCA1/2/3, PMCA1/2/3/4) were decreased in a cisplatin-resistant cell line compared with those in their parental cell lines (Table II) (125). Currently, there is a lack of research focusing on the role of Ca2+-ATPases in OC, which should be explored as a starting point for future investigations into expanding the understanding of Ca2+-ATPases and their potential as therapeutic targets.

4. Mitochondrial Ca2+ transport

Mitochondria serve as a central hub of [Ca2+]i regulation and mediate Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCU) channel, which regulates [Ca2+]i. MCU transports Ca2+ within the mitochondrial lumen, using the negative charge of the inner mitochondrial membrane to sustain the Ca2+ levels (126). The MCU complex consists of endosomal channel-dependent MCU proteins and their regulators, including the MCU-dominant negative b subunit (MCUb) and MCU-related regulatory proteins, such as the mitochondrial Ca2+ uptake (MICU) family (MICU1, MICU2 and MICU3), MCU regulator 1 (MCUR1) and essential MCU regulator (EMRE) (127). The activity of the MCU channel is regulated by MICU1 and MICU2, which form 95 kDa dimers through disulfide bonds. At higher Ca2+ levels, Ca2+-dependent MICU1 activation and MICU2 inhibition ensure a rapid mitochondrial response to Ca2+ signals generated in the cytoplasm (128,129). In OC, it has been discovered that mitochondrial Ca2+ uptake by the gatekeeper mitochondrial calcium uptake 1 (MICU1/CBARA1) drives aerobic glycolysis. MICU1 is overexpressed in a panel of OC cell lines (CP20 and OV90) and its overexpression is associated with decreased overall survival (130). In vitro silencing of MICU1 increases oxygen consumption, decreases lactate production and inhibits clonal growth, migration and invasion of OC cells, while in vivo silencing inhibits tumor growth and enhances cisplatin efficacy and overall survival (Table II) (130).

It is known that the electron transport chain (ETC) drives physiological mitochondrial Ca2+ uptake. However, ETC overload and partial ETC inhibition can cause ROS production, leading to oxidative damage to the mitochondrial membrane. This in turn results in cell death and ROS-dependent tumor cell metastasis and invasion (131-133). Several studies have shown that modifying the Ca2+ concentration in mitochondria can be a potential treatment method for OC (134-136). Gentisyl alcohol, which has antibacterial, antifungal, antiviral and anticancer properties, is observed to inhibit cell proliferation while inducing apoptosis in human OC cells through DNA fragmentation (134). In addition, β-Sitosterol (135), Campesterol (136), Stigmasterol (137), Osthole (138), Fucosterol (139), Laminarin (140), Chrysophanol (141), Chrysin (142) and Epothilone B (143) have all been shown to increase ROS production by dose-dependently elevating Ca2+ concentrations in the cytoplasm and mitochondria of OC cells, leading to oxidative stress through the endogenous pathway and initiate apoptotic signaling (Table IV). Mitochondrial Ca2+ overload activates the unfolded protein response and the ER/mitochondrial axis, which then disrupt [Ca2+]i homeostasis, initiate apoptosis and inhibit cell proliferation (144). Treatment of cells with β-Sitosterol or Campesterol impairs mitochondrial membrane function, leading to the loss of membrane potential and disruption of Ca2+ homeostasis (135,136). Furthermore, laminarin suppresses the expression of the ER mitochondrial coupling protein glucose-regulated protein 75 (GRP75) in OC cells (140), where the lack of GRP75 expression has been associated with Ca2+ overload (145).

Table IV

Summary of drugs/compounds targeting Ca2+ signaling for ovarian cancer treatment.

Table IV

Summary of drugs/compounds targeting Ca2+ signaling for ovarian cancer treatment.

Calcium signals Drugs/CompoundsMechanism(Refs.)
TRPTRPC3PLAAInhibitor(27)
TRPV1DWP05195, Ursolic acidAntagonist(34,35)
TRPM7MK886, BAPTA-AMInhibitor(61)
VGCCsTTCCMibedil, 3,4-dihydroquinazoline derivativesBlocker(64)
LTCCNifedipineBlocker(69)
TTCC&LTCCCCBsBlocker(70,71)
BKCa-NS1619Activator(81)
-Trimebutine MaleateInhibitor(71)
GPCRs-TUTPAntagonist(97)
MCU-Gentisyl Alcohol, β-Sitosterol, Campesterol, Stigmasterol, Osthole, Fucosterol, Laminarin, Chrysophanol, Chrysin, Epothilone BInhibitor(136-145)
-ABT-737, GRP75Activator(150,151)
Others-Saikosaponin DCalcium mobilizer(182)

[i] TRP, transient receptor potential; PLAA, phospholipase A2 activating protein; MK886, 5-Lipoxygenase inhibitor; BAPTA-AM, Intracellular calcium chelator; VGCC, Voltage-gated Calcium Channel; TTCC, T-type Ca2+ channel; LTCC, L-type Ca2+ channel; CCBs, calcium channel blockers; BKCa, large conductance Ca2+-activated K+; NS1619, the BKCa channel opener; GPCR, G-protein-coupled receptor; TUTP, 2-thioureidothiophene-3-carboxylate; MCU, mitochondrial Ca2+ uniporter; ABT-737, a small-molecule Bcl-2 inhibitor; GRP75, glucose-regulated protein 75.

The high mortality rate of OC is largely attributed to its resistance to currently available chemotherapeutic drugs (7). Cisplatin is commonly used for the treatment of malignant OC, but acquired resistance limits its application. The inability to upregulate [Ca2+]i in OC cells results in cisplatin resistance by reducing oxidative stress (146). Bcl-2, a key regulator of survival and apoptosis, is known to block cisplatin-induced apoptosis by regulating Ca2+ signaling in various cancer cell lines. Bcl-2 overexpression inhibits ER mitochondrial Ca2+ signaling and increases cisplatin resistance in OC cells (147). ABT-737, a small-molecule Bcl-2 inhibitor, has been shown to increase free Ca2+ levels in the mitochondria in combination with cisplatin treatment of cisplatin-resistant OC cells, thereby enhancing mitochondria-mediated cell apoptosis (148). Increased mitochondrial Ca2+ may induce apoptosis in cisplatin-resistant OC cells, where the enrichment of GRP75 in the mitochondria-associated ER membranes may be responsible for this effect (149). In paclitaxel-resistant OC cells, lncRNA-RNA component of mitochondrial RNA processing endoribonuclease (RMRP) has been shown to increase MICU1 expression through miR-580-3p aggregation. By contrast, targeting lncRNA-RMRP was found to inhibit the miR-580-3p/MICU1 axis to increase paclitaxel sensitivity (150). Overall, mitochondrial Ca2+ alterations probably serve a significant role in the treatment of OC. Further in-depth studies into MCU channels can aid in understanding their roles in the occurrence, development and prognosis of OC. These are expected to facilitate the development of novel therapeutic targets and search for new therapeutic methods.

5. S100 family and other Ca2+ signaling pathways

S100 family

There are 21 members in the S100 family known to date, all of which are found in human tissues and are acidic Ca2+-binding proteins. These proteins are highly homologous both in terms of sequence and structure, can switch roles within a given biological process and are involved in a wide variety of cellular events, such as proliferation, apoptosis, migration, inflammation and differentiation (151). The proteins that make up the S100 family can serve as both Ca2+ sensors on the inside of cells and as extracellular factors promoting proliferation from the outside. Therefore, aberrant expression of S100 proteins has been proposed to be another factor in tumor development and progression (152,153). In a previous review, Bresnick et al (151) discussed the importance of S100 family members in diagnosing and treating cancer, how S100 signaling can affects the growth of tumors and how S100 inhibitors were found to treat cancer. With the progression of the disease, multi-drug resistance to tumor therapy remains to be a problem. Hua et al (154) found that the dysregulation of different S100 proteins can contribute to the development of tumor drug resistance, which worsens the prognosis of patients with cancer. A summary was also provided of how S100 family members can affect tumor resistance to therapy, pointing out that inhibition of S100 proteins can mediate the response of tumors to therapy. Accumulating evidence suggests multiple members of the S100 family are involved in OC development and progression (Fig. 2 and Table III) (153).

Compared with fallopian tube and normal ovarian epithelial tissues, S100A1 expression tends to be significantly higher in OC tissues, which is also associated with lymph node metastasis, International Federation of Gynecology and Obstetrics (FIGO) staging and tumor grade (155). S100A2 has also been hypothesized to be a tumor suppressor that aids in the stabilization and response to the transcription of mutant p53, thereby controlling cell proliferation (156). Higher expression levels of S100A2 have been shown to predict superior overall survival in patients with OC expressing wild-type TP53, but had no prognostic value in patients with mutant p53 OC. This suggests that the interaction between S100A2 and TP53 may mediate the tumor suppressive effects of S100A2 (153). The function of S100A3 in OC remains to be elucidated. Kikuchi et al (157) found that S100A4 is highly expressed in the nucleus in OC tissues; OC patients with stronger nuclear S100A4 expression showed a significantly shorter survival time compared those without. Subsequent treatment with the recombinant S100A4 resulted in the translocation of S100A4 into the nucleus, the enhancement of which enhanced OC cell invasiveness. These findings suggest that the nuclear expression of S100A4 is involved in the aggressive behavior of OC. Furthermore, nuclear expression of S100A4 in combination with the nuclear HIF-1α protein under hypoxic conditions has been demonstrated to induce hypoxia response element-free methylation of the S100A4 gene and promote OC aggressiveness (158). In addition, miR-296 is an important upstream regulator of S100A4 and aberrant regulation of the miR-296/S100A4 axis has been reported to promote the EMT process and hasten OC progression (159). It was first proposed by Link et al (160) that high levels of circulating metastasis-associated in colon cancer 1 and S100A4 transcripts could predict the prognosis of patients with OC, because they were associated with advanced FIGO staging. Another previous study has shown that the insulin-like growth factor 1 receptor 6-/integrin-/S100A4 molecular network can regulate the organ-specific metastasis of chemoresistant epithelial OC cells. Genetic and pharmacological inhibition of S100A4 was found to significantly reduce distant metastasis and completely eliminated lung invasion by advanced chemoresistant epithelial OC cells (161). S100A5 is a novel member of the S100 protein family that can interact with Ca2+, Zn2+ and Cu2+ (162). High S100A5 expression was previously reported to predict overall survival in all patients with EOC (153).

S100A6 expression has been documented in cancer xenografts and OC tissues. Wei et al (163) found that serum S100A6 concentrations are higher in patients with advanced OC compared with those with early OC. This suggests that S100A6 concentrations are associated with experimental tumor load and clinical disease stage, making S100A6 a useful biomarker for detecting and/or monitoring OC (163). In addition, in a previous study by Bai et al (153), a positive association between S100A6 mRNA expression levels and overall survival was identified in stage II patients but a negative association was found with stage IV patients. This suggests that S100A6 may serve different roles in patients with early and advanced OC. It is necessary to be able to independently reproduce these results followed by a deeper investigation into the associated underlying mechanism. S100A7 has been shown to promote tumor cell proliferation, migration, invasion and tumor metastasis. Metastasis and chemoresistance in OC cells have been shown to be controlled by S100A7 through the MAPK signaling pathway. miR-330-5p can target the 3'-untranslated region of S100A7, thereby reducing the activity of the protein and then the proliferation of OC cells (164). S100A10 is found in the plasma membrane, where it associates with Annexin A2 to form a heterotetramer (165). S100A10 expression in OC tissues has been associated with decreased overall survival and progression-free survival (165). In addition, a high S100A10 expression was found to be an independent predictor of OC prognosis, increasing the risk of progression and mortality from OC (166). Supporting this, OC cells were rendered more sensitive to carboplatin when the expression of S100A10 was downregulated (167).

High expression of S100A11 in the serum of patients with OC and increased proliferation, migration and invasion of OC cells are attributed to the lncRNA SNHG8, which regulates OC progression by targeting miR-1270 and S100A11 (168,169). Patients with grade II, stage I+II and p53 mutant OC had a longer overall survival if S100A13 levels were elevated (153). Serum S100A14 levels was found to be consistently higher in patients with OC, where a link was also found between elevated S100A14 and resistance to platinum-based chemotherapy (170). Higher levels of S100A16, a member of the S100 family isolated from astrocytomas (171), have also been associated with worse prognosis in patients with OC, particularly those with grade II, III and stage III EOC (153). S100B protein is overexpressed in OC tissues compared with that in normal ovaries and is in turn associated with advanced tumor stage, decreased differentiation and shorter overall survival (172). In addition, S100B has been documented to mediate chemotherapy resistance in OC cells through p53 (173) and controls the stemness of OC stem cell-like cells (172). Although high S100P expression is associated with a worse prognosis in OC patients in terms of overall survival and progression free survival, S100P has been shown to increase chemosensitivity of OC cells to carboplatin and paclitaxel in vitro (174,175). Overall, a comprehensive understanding of the function of S100 family members is clinically instructive for the diagnosis and prognosis of OC patients. According to results from a previous survey, S100 protein mRNA expression is strongly associated with overall survival in patients with OC, with high levels of S100 family members S100A10, S100A11, S100A16, S100B and S100P predicting worse overall survival, while S100A1, S100A2, S100A5, S100A6 and S100A13 were associated with longer overall survival, depending in part on OC subtype and clinicopathological features (153,176).

Several promising approaches are currently proposed and make use of current knowledge to assess S100 proteins as potential therapeutic targets of cancer therapy, as evidenced by the aforementioned studies. However, additional research is required to firmly establish S100 proteins as reliable biomarkers for OC therapy and to further characterize their roles in OC pathophysiology. Although these initial findings show promise, the true extent of the function of S100 proteins in OC remains unknown, which requires unravelling it can be fully exploited in the clinic.

Other Ca2+ signaling pathways

Ca2+ signaling is also associated with other molecules such as tumor-associated calcium signal transducer 2 (TROP2), calcium/calmodulin-dependent protein kinase (CaMKK) and saikosaponin-D (SSD) in OC (Table III) (177-179). TROP2 is a newly identified marker that plays a vital role in the proliferation and invasion of various tumors by transducing [Ca2+]i signaling (177). Wu et al (177) found that suppressing TROP2 expression in OC cells significantly slows cell proliferation, invasion and migration. CaMKK-β-mediated AMPK activity is required for regulating autophagy induction in OC spheroids and supporting cell viability, at least in part (180). Elevated [Ca2+]i activates the expression of CaMKK2, which mediates epidermal growth factor signaling through Akt signaling and is used by cancer cells as a signal for growth and survival (178). Chen et al (181) reported that CaMKK2 promotes the progression of ovarian carcinoma through the PI3K/PDK1/Akt Axis. SSD, a major bioactive component of Radix Bupleuri, exhibits anti-inflammatory, anti-tumor, anti-oxidant and anti-viral effects (179). Tsuyoshi et al (182) found that SSD may be a new adjuvant for the treatment of chemoresistant OC because it acts as a Ca2+ mobilizer and sensitizes OC cells to cisplatin by promoting mitochondrial division and G2/M blockade through multiple signaling pathways. These findings suggest that TROP2, CaMKK2 and SSD are critical molecules in OC cell proliferation and point to a promising direction for future research.

6. Conclusion and outlook

Healthy and malignant cells both rely on cytosolic Ca2+ signaling for the regulation of their intracellular cellular processes. However, alterations in Ca2+ fluxes can overlap with crucial stages of the life cycle of types of cancer. Changes in the expression of Ca2+ channels, pumps and exchange proteins in OC tissues all suggest that Ca2+ plays an important role in regulating OC cell proliferation, migration and invasion (Fig. 3). In addition, drug-resistant OC cells have lower levels of Ca2+ and key genes involved in Ca2+ homeostasis, supporting the hypothesis that alterations in Ca2+ regulation contribute to tumorigenesis, metastasis, prognosis and drug resistance. The currently proposed inhibitors or activators targeting Ca2+ channels for the treatment of OC are summarized in Table IV.

Although recent advancements in Ca2+ channel research have provided promising insights, further investigations are required to establish effective combinatorial methods of targeted medications and chemotherapy to boost the survival rate of patients with OC. Understanding the biological processes that govern the modulation of Ca2+ signaling pathways in OC cells is urgently required. Ca2+ signaling appears to be a valid target for anticancer therapy in patients with OC and is supported by a substantial body of preclinical and clinical evidence. However, gaps in knowledge remain in our understanding of the role of Ca2+ signaling in types of cancer which is required to uncover the mechanisms of Ca2+ signaling in OC pathogenesis and facilitate the creation of novel therapeutic strategies. In conclusion, alterations in Ca2+ fluxes can influence malignant transformation, tumor progression and response to therapy in patients with OC by affecting a complex network of OC cell-intrinsic and extrinsic functions. Further research is necessary to disentangle its molecular and functional complexity.

Availability of data and materials

Data sharing is not applicable to this article, as no data sets were generated or analyzed during the current study.

Ethics approval and consent to participate

Not applicable.

Authors' contributions

QG and FD wrote the original draft of the manuscript. QG, YZ, JL, MF, CZ and BJ reviewed and edited the manuscript. QG, FD, YZ, HD and TX supervised the present study. QG, JQ, JC, FD, JL, MF and JQ performed project administration: Data authentication is not applicable. All authors have read and agreed to the published version of the manuscript.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported partly by the National Nature and Science Foundation of China (grant nos. 82271724, 81873841, 81741024 and 81401244), Ministry of Science and Technology (grant no. 2019YFA0802600), Suzhou City Wei Sheng Ren Cai program (grant no. GSWS2019029), General Programs of Jiangsu Commission of Health (grant no. M2021087) and Nature and Science Foundation of Jiangsu (grant no. BK20221243).

References

1 

Matz M, Coleman MP, Sant M, Chirlaque MD, Visser O, Gore M and Allemani C; the CONCORD Working Group: The histology of ovarian cancer: Worldwide distribution and implications for international survival comparisons (CONCORD-2). Gynecol Oncol. 144:405–413. 2017. View Article : Google Scholar

2 

Lisio MA, Fu L, Goyeneche A, Gao ZH and Telleria C: High-grade serous ovarian cancer: Basic sciences, clinical and therapeutic standpoints. Int J Mol Sci. 20:9522019. View Article : Google Scholar : PubMed/NCBI

3 

Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J and Karlan BY: Ovarian cancer. Nat Rev Dis Primers. 2:160612016. View Article : Google Scholar : PubMed/NCBI

4 

Mallen A, Soong TR, Townsend MK, Wenham RM, Crum CP and Tworoger SS: Surgical prevention strategies in ovarian cancer. Gynecol Oncol. 151:166–175. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Menon U, Karpinskyj C and Gentry-Maharaj A: Ovarian cancer prevention and screening. Obstet Gynecol. 131:909–927. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, Gaudet MM, Jemal A and Siegel RL: Ovarian cancer statistics, 2018. CA Cancer J Clin. 68:284–296. 2018. View Article : Google Scholar : PubMed/NCBI

8 

O'Malley DM: New therapies for ovarian cancer. J Natl Compr Canc Netw. 17:619–621. 2019.PubMed/NCBI

9 

Zhang M, Cheng S, Jin Y, Zhao Y and Wang Y: Roles of CA125 in diagnosis, prediction and oncogenesis of ovarian cancer. Biochim Biophys Acta Rev Cancer. 1875:1885032021. View Article : Google Scholar

10 

Berridge MJ, Lipp P and Bootman MD: The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol. 1:11–21. 2000. View Article : Google Scholar

11 

Altamura C, Greco MR, Carratù MR, Cardone RA and Desaphy JF: Emerging roles for ion channels in ovarian cancer: Pathomechanisms and pharmacological treatment. Cancers (Basel). 13. pp. 6682021, View Article : Google Scholar

12 

Caravia L, Staicu CE, Radu BM, Condrat CE, Crețoiu D, Bacalbașa N, Suciu N, Crețoiu SM and Voinea SC: Altered organelle calcium transport in ovarian physiology and cancer. Cancers (Basel). 12:22322020. View Article : Google Scholar : PubMed/NCBI

13 

Monteith GR, McAndrew D, Faddy HM and Roberts-Thomson SJ: Calcium and cancer: Targeting Ca2+ transport. Nat Rev Cancer. 7:519–530. 2007. View Article : Google Scholar : PubMed/NCBI

14 

McConkey DJ and Orrenius S: The role of calcium in the regulation of apoptosis. Biochem Biophys Res Commun. 239:357–366. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Prevarskaya N, Skryma R and Shuba Y: Calcium in tumour metastasis: New roles for known actors. Nat Rev Cancer. 11:609–618. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Pulliam TL, Goli P, Awad D, Lin C, Wilkenfeld SR and Frigo DE: Regulation and role of CAMKK2 in prostate cancer. Nat Rev Urol. 19:367–380. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Venkatachalam K, Luo J and Montell C: Evolutionarily conserved, multitasking TRP channels: Lessons from worms and flies. Handb Exp Pharmacol. 223:937–962. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Chen JP, Wang J, Luan Y, Wang CX, Li WH, Zhang JB, Sha D, Shen R, Cui YG, Zhang Z, et al: TRPM7 promotes the metastatic process in human nasopharyngeal carcinoma. Cancer Lett. 356:483–490. 2015. View Article : Google Scholar

19 

Chen X, Sooch G, Demaree IS, White FA and Obukhov AG: Transient receptor potential canonical (TRPC) channels: Then and now. Cells. 9:19832020. View Article : Google Scholar : PubMed/NCBI

20 

He B, Liu F, Ruan J, Li A, Chen J, Li R, Shen J, Zheng D and Luo R: Silencing TRPC1 expression inhibits invasion of CNE2 nasopharyngeal tumor cells. Oncol Rep. 27:1548–1554. 2012.PubMed/NCBI

21 

Ong HL and Ambudkar IS: The dynamic complexity of the TRPC1 channelosome. Channels (Austin). 5:424–431. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Liu X, Zou J, Su J, Lu Y, Zhang J, Li L and Yin F: Downregulation of transient receptor potential cation channel, subfamily C, member 1 contributes to drug resistance and high histological grade in ovarian cancer. Int J Oncol. 48:243–252. 2016. View Article : Google Scholar

23 

Yang SL, Cao Q, Zhou KC, Feng YJ and Wang YZ: Transient receptor potential channel C3 contributes to the progression of human ovarian cancer. Oncogene. 28:1320–1328. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Tao X, Zhao N, Jin H, Zhang Z, Liu Y, Wu J, Bast RC Jr, Yu Y and Feng Y: FSH enhances the proliferation of ovarian cancer cells by activating transient receptor potential channel C3. Endocr Relat Cancer. 20:415–429. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Li S, Jiang K, Li J, Hao X, Chu W, Luo C, Zhu Y, Xie R and Chen B: Estrogen enhances the proliferation and migration of ovarian cancer cells by activating transient receptor potential channel C3. J Ovarian Res. 13:202020. View Article : Google Scholar : PubMed/NCBI

26 

Liu EL, Zhou YX, Li J, Zhang DH and Liang F: Long-chain non-coding RNA SNHG3 promotes the growth of ovarian cancer cells by targeting miR-339-5p/TRPC3 axis. Onco Targets Ther. 13:10959–10971. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Shen Z, Gu L, Liu Y, Wang L, Zhu J, Tang S, Wei X, Wang J, Zhang S, Wang X, et al: PLAA suppresses ovarian cancer metastasis via METTL3-mediated m6A modification of TRPC3 mRNA. Oncogene. 41:4145–4158. 2022. View Article : Google Scholar : PubMed/NCBI

28 

Farfariello V, Gordienko DV, Mesilmany L, Touil Y, Germain E, Fliniaux I, Desruelles E, Gkika D, Roudbaraki M, Shapovalov G, et al: TRPC3 shapes the ER-mitochondria Ca2+ transfer characterizing tumour-promoting senescence. Nat Commun. 13:9562022. View Article : Google Scholar

29 

Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD and Julius D: The capsaicin receptor: A heat-activated ion channel in the pain pathway. Nature. 389:816–824. 1997. View Article : Google Scholar : PubMed/NCBI

30 

Gunthorpe MJ, Benham CD, Randall A and Davis JB: The diversity in the vanilloid (TRPV) receptor family of ion channels. Trends Pharmacol Sci. 23:183–191. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Wang Z, Dong J, Tian W, Qiao S and Wang H: Role of TRPV1 ion channel in cervical squamous cell carcinoma genesis. Front Mol Biosci. 9:9802622022. View Article : Google Scholar : PubMed/NCBI

32 

Lucido CT, Wynja E, Madeo M, Williamson CS, Schwartz LE, Imblum BA, Drapkin R and Vermeer PD: Innervation of cervical carcinoma is mediated by cancer-derived exosomes. Gynecol Oncol. 154:228–235. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Han GH, Chay DB, Nam S, Cho H, Chung JY and Kim JH: Prognostic significance of transient receptor potential vanilloid type 1 (TRPV1) and phosphatase and tension homolog (PTEN) in epithelial ovarian cancer. Cancer Genomics Proteomics. 17:309–319. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Wang YY, Lee KT, Lim MC and Choi JH: TRPV1 antagonist DWP05195 induces ER stress-dependent apoptosis through the ROS-p38-CHOP pathway in human ovarian cancer cells. Cancers (Basel). 12:17022020. View Article : Google Scholar : PubMed/NCBI

35 

Di Y, Xu T, Tian Y, Ma T, Qu D, Wang Y, Lin Y, Bao D, Yu L, Liu S and Wang A: Ursolic acid protects against cisplatin-induced ototoxicity by inhibiting oxidative stress and TRPV1-mediated Ca2+-signaling. Int J Mol Med. 46:806–816. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Santoni G, Amantini C, Maggi F, Marinelli O, Santoni M, Nabissi M and Morelli MB: The TRPV2 cation channels: From urothelial cancer invasiveness to glioblastoma multiforme interactome signature. Lab Invest. 100:186–198. 2020. View Article : Google Scholar

37 

Liberati S, Morelli MB, Amantini C, Farfariello V, Santoni M, Conti A, Nabissi M, Cascinu S and Santoni G: Loss of TRPV2 homeostatic control of cell proliferation drives tumor progression. Cells. 3:112–128. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Fraguas-Sánchez AI, Fernández-Carballido A, Delie F, Cohen M, Martin-Sabroso C, Mezzanzanica D, Figini M, Satta A and Torres-Suárez AI: Enhancing ovarian cancer conventional chemotherapy through the combination with cannabidiol loaded microparticles. Eur J Pharm Biopharm. 154:246–258. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Griffiths C, Aikins J, Warshal D and Ostrovsky O: Can cannabidiol affect the efficacy of chemotherapy and epigenetic treatments in cancer? Biomolecules. 11:7662021. View Article : Google Scholar : PubMed/NCBI

40 

Dutta B, Arya RK, Goswami R, Alharbi MO, Sharma S and Rahaman SO: Role of macrophage TRPV4 in inflammation. Lab Invest. 100:178–185. 2020. View Article : Google Scholar :

41 

Wang K, Feng X, Zheng L, Chai Z, Yu J, You X, Li X and Cheng X: TRPV4 is a prognostic biomarker that correlates with the immunosuppressive microenvironment and chemoresistance of anti-cancer drugs. Front Mol Biosci. 8:6905002021. View Article : Google Scholar : PubMed/NCBI

42 

Zhang C, Xu C, Ma C, Zhang Q, Bu S, Zhang DL, Yu L and Wang H: TRPs in ovarian serous cystadenocarcinoma: The expression patterns, prognostic roles, and potential therapeutic targets. Front Mol Biosci. 9:9154092022. View Article : Google Scholar : PubMed/NCBI

43 

Yu S, Huang S, Ding Y, Wang W, Wang A and Lu Y: Transient receptor potential ion-channel subfamily V member 4: A potential target for cancer treatment. Cell Death Dis. 10:4972019. View Article : Google Scholar : PubMed/NCBI

44 

Bödding M and Flockerzi V: Ca2+ dependence of the Ca2+-selective TRPV6 channel. J Biol Chem. 279:36546–36552. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Gees M, Colsoul B and Nilius B: The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol. 2:a0039622010. View Article : Google Scholar : PubMed/NCBI

46 

Lehen'kyi V, Flourakis M, Skryma R and Prevarskaya N: TRPV6 channel controls prostate cancer cell proliferation via Ca(2+)/NFAT-dependent pathways. Oncogene. 26:7380–7385. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Xu X, Li N, Wang Y, Yu J and Mi J: Calcium channel TRPV6 promotes breast cancer metastasis by NFATC2IP. Cancer Lett. 519:150–160. 2021. View Article : Google Scholar : PubMed/NCBI

48 

Xue H, Wang Y, MacCormack TJ, Lutes T, Rice C, Davey M, Dugourd D, Ilenchuk TT and Stewart JM: Inhibition of transient receptor potential vanilloid 6 channel, elevated in human ovarian cancers, reduces tumour growth in a xenograft model. J Cancer. 9:3196–3207. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Jiang Y, Gou H, Zhu J, Tian S and Yu L: Lidocaine inhibits the invasion and migration of TRPV6-expressing cancer cells by TRPV6 downregulation. Oncol Lett. 12:1164–1170. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Wang X, Li G, Zhang Y, Li L, Qiu L, Qian Z, Zhou S, Wang X, Li Q and Zhang H: Pan-cancer analysis reveals genomic and clinical characteristics of TRPV channel-related genes. Front Oncol. 12:8131002022. View Article : Google Scholar : PubMed/NCBI

51 

Tong Q, Zhang W, Conrad K, Mostoller K, Cheung JY, Peterson BZ and Miller BA: Regulation of the transient receptor potential channel TRPM2 by the Ca2+ sensor calmodulin. J Biol Chem. 281:9076–9085. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Orfanelli U, Wenke AK, Doglioni C, Russo V, Bosserhoff AK and Lavorgna G: Identification of novel sense and antisense transcription at the TRPM2 locus in cancer. Cell Res. 18:1128–1140. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Ding Y, Tan X, Abasi A, Dai Y, Wu R, Zhang T, Li K, Yan M and Huang X: LncRNA TRPM2-AS promotes ovarian cancer progression and cisplatin resistance by sponging miR-138-5pto release SDC3 mRNA. Aging (Albany NY). 13:6832–6848. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Dai W, Bai Y, Hebda L, Zhong X, Liu J, Kao J and Duan C: Calcium deficiency-induced and TRP channel-regulated IGF1R-PI3K-Akt signaling regulates abnormal epithelial cell proliferation. Cell Death Differ. 21:568–581. 2014. View Article : Google Scholar :

55 

Abed E, Martineau C and Moreau R: Role of melastatin transient receptor potential 7 channels in the osteoblastic differentiation of murine MC3T3 cells. Calcif Tissue Int. 88:246–253. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Yee NS, Kazi AA and Yee RK: Cellular and developmental biology of TRPM7 channel-kinase: Implicated roles in cancer. Cells. 3:751–777. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Wang J, Xiao L, Luo CH, Zhou H, Hu J, Tang YX, Fang KN and Zhang Y: Overexpression of TRPM7 is associated with poor prognosis in human ovarian carcinoma. Asian Pac J Cancer Prev. 15:3955–3958. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Wang J, Liao QJ, Zhang Y, Zhou H, Luo CH, Tang J, Wang Y, Tang Y, Zhao M, Zhao XH, et al: TRPM7 is required for ovarian cancer cell growth, migration and invasion. Biochem Biophys Res Commun. 454:547–553. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Liu L, Wu N, Wang Y, Zhang X, Xia B, Tang J, Cai J, Zhao Z, Liao Q and Wang J: TRPM7 promotes the epithelial-mesenchymal transition in ovarian cancer through the calcium-related PI3K/AKT oncogenic signaling. J Exp Clin Cancer Res. 38:1062019. View Article : Google Scholar

60 

Catterall WA: Voltage-gated calcium channels. Cold Spring Harb Perspect Biol. 3:a0039472011. View Article : Google Scholar : PubMed/NCBI

61 

Li W, Zhang SL, Wang N, Zhang BB and Li M: Blockade of T-type Ca(2+) channels inhibits human ovarian cancer cell proliferation. Cancer Invest. 29:339–346. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Jang SJ, Choi HW, Choi DL, Cho S, Rim HK, Choi HE, Kim KS, Huang M, Rhim H, Lee KT and Lee JY: In vitro cytotoxicity on human ovarian cancer cells by T-type calcium channel blockers. Bioorg Med Chem Lett. 23:6656–6662. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Dziegielewska B, Casarez EV, Yang WZ, Gray LS, Dziegielewski J and Slack-Davis JK: T-type Ca2+ channel inhibition sensitizes ovarian cancer to carboplatin. Mol Cancer Ther. 15:460–470. 2016. View Article : Google Scholar : PubMed/NCBI

64 

Mir R, Stanzani E, Martinez-Soler F, Villanueva A, Vidal A, Condom E, Ponce J, Gil J, Tortosa A and Giménez-Bonafé P: YM155 sensitizes ovarian cancer cells to cisplatin inducing apoptosis and tumor regression. Gynecol Oncol. 132:211–220. 2014. View Article : Google Scholar

65 

Fornaro L, Vivaldi C, Lin D, Xue H, Falcone A, Wang Y, Crea F and Bootman MD: Prognostic relevance of a T-type calcium channels gene signature in solid tumours: A correlation ready for clinical validation. PLoS One. 12:e01828182017. View Article : Google Scholar : PubMed/NCBI

66 

Mertens-Walker I, Bolitho C, Baxter RC and Marsh DJ: Gonadotropin-induced ovarian cancer cell migration and proliferation require extracellular signal-regulated kinase 1/2 activation regulated by calcium and protein kinase C{delta}. Endocr Relat Cancer. 17:335–349. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Kim EK, Ha JM, Kim YW, Jin SY, Ha HK and Bae SS: Inhibitory role of polyunsaturated fatty acids on lysophosphatidic acid-induced cancer cell migration and adhesion. FEBS Lett. 588:2971–2977. 2014. View Article : Google Scholar : PubMed/NCBI

68 

Lee H, Kim JW, Kim DK, Choi DK, Lee S, Yu JH, Kwon OB, Lee J, Lee DS, Kim JH and Min SH: Calcium channels as novel therapeutic targets for ovarian cancer stem cells. Int J Mol Sci. 21:23272020. View Article : Google Scholar : PubMed/NCBI

69 

Lee H, Kwon OB, Lee JE, Jeon YH, Lee DS, Min SH and Kim JW: Repositioning trimebutine maleate as a cancer treatment targeting ovarian cancer stem cells. Cells. 10:9182021. View Article : Google Scholar : PubMed/NCBI

70 

Chang X and Dong Y: CACNA1C is a prognostic predictor for patients with ovarian cancer. J Ovarian Res. 14:882021. View Article : Google Scholar : PubMed/NCBI

71 

Niemeyer BA: Changing calcium: CRAC channel (STIM and Orai) expression, splicing, and posttranslational modifiers. Am J Physiol Cell Physiol. 310:C701–C709. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Khan HY, Mazahir I, Reddy S, Fazili F and Azmi A: Roles of CRAC channel in cancer: Implications for therapeutic development. Expert Rev Precis Med Drug Dev. 5:371–382. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Abdelazeem KNM, Droppova B, Sukkar B, Al-Maghout T, Pelzl L, Zacharopoulou N, Ali Hassan NH, Abdel-Fattah KI, Stournaras C and Lang F: Upregulation of Orai1 and STIM1 expression as well as store-operated Ca2+ entry in ovary carcinoma cells by placental growth factor. Biochem Biophys Res Commun. 512:467–472. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Schmidt S, Liu G, Liu G, Yang W, Honisch S, Pantelakos S, Stournaras C, Hönig A and Lang F: Enhanced Orai1 and STIM1 expression as well as store operated Ca2+ entry in therapy resistant ovary carcinoma cells. Oncotarget. 5:4799–4810. 2014. View Article : Google Scholar : PubMed/NCBI

75 

Zahid M, Beseler CL, Hall JB, LeVan T, Cavalieri EL and Rogan EG: Unbalanced estrogen metabolism in ovarian cancer. Int J Cancer. 134:2414–2423. 2014. View Article : Google Scholar :

76 

Lv X, Miao C, Liu M, Wang X, Wang L and Wang D: 17β-Estradiol via Orai1 activates calcium mobilization to induce cell proliferation in epithelial ovarian cancer. J Biochem Mol Toxicol. 34:e226032020. View Article : Google Scholar

77 

Ouadid-Ahidouch H, Roudbaraki M, Delcourt P, Ahidouch A, Joury N and Prevarskaya N: Functional and molecular identification of intermediate-conductance Ca(2+)-activated K(+) channels in breast cancer cells: Association with cell cycle progression. Am J Physiol Cell Physiol. 287:C125–C134. 2004. View Article : Google Scholar : PubMed/NCBI

78 

Kunzelmann K: Ion channels and cancer. J Membr Biol. 205:159–173. 2005. View Article : Google Scholar : PubMed/NCBI

79 

Han X, Xi L, Wang H, Huang X, Ma X, Han Z, Wu P, Ma X, Lu Y, Wang G, et al: The potassium ion channel opener NS1619 inhibits proliferation and induces apoptosis in A2780 ovarian cancer cells. Biochem Biophys Res Commun. 375:205–209. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Berkefeld H, Sailer CA, Bildl W, Rohde V, Thumfart JO, Eble S, Klugbauer N, Reisinger E, Bischofberger J, Oliver D, et al: BKCa-Cav channel complexes mediate rapid and localized Ca2+-activated K+ signaling. Science. 314:615–620. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Oeggerli M, Tian Y, Ruiz C, Wijker B, Sauter G, Obermann E, Güth U, Zlobec I, Sausbier M, Kunzelmann K and Bubendorf L: Role of KCNMA1 in breast cancer. PLoS One. 7:e416642012. View Article : Google Scholar : PubMed/NCBI

82 

Samuel P, Pink RC, Caley DP, Currie JM, Brooks SA and Carter DR: Over-expression of miR-31 or loss of KCNMA1 leads to increased cisplatin resistance in ovarian cancer cells. Tumour Biol. 37:2565–2573. 2016. View Article : Google Scholar

83 

Lundstrom K: Structural genomics of GPCRs. Trends Biotechnol. 23:103–108. 2005. View Article : Google Scholar : PubMed/NCBI

84 

Oldham WM and Hamm HE: Heterotrimeric G protein activation by G-protein-coupled receptors. Nat Rev Mol Cell Biol. 9:60–71. 2008. View Article : Google Scholar

85 

Tu CL, Oda Y, Komuves L and Bikle DD: The role of the calcium-sensing receptor in epidermal differentiation. Cell Calcium. 35:265–273. 2004. View Article : Google Scholar : PubMed/NCBI

86 

Rodland KD: The role of the calcium-sensing receptor in cancer. Cell Calcium. 35:291–295. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Yan S, Yuan C, Yang Q, Li X, Yang N, Liu X, Dong R, Zhang X, Yuan Z, Zhang N and Kong B: A genetic polymorphism (rs17251221) in the calcium-sensing receptor is associated with ovarian cancer susceptibility. Oncol Rep. 34:2151–2155. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Park KS, Kim MK, Im DS and Bae YS: Effect of lysophosphatidylglycerol on several signaling molecules in OVCAR-3 human ovarian cancer cells: Involvement of pertussis toxin-sensitive G-protein coupled receptor. Biochem Pharmacol. 73:675–681. 2007. View Article : Google Scholar

89 

Smith HO, Arias-Pulido H, Kuo DY, Howard T, Qualls CR, Lee SJ, Verschraegen CF, Hathaway HJ, Joste NE and Prossnitz ER: GPR30 predicts poor survival for ovarian cancer. Gynecol Oncol. 114:465–471. 2009. View Article : Google Scholar : PubMed/NCBI

90 

Yan Y, Liu H, Wen H, Jiang X, Cao X, Zhang G and Liu G: The novel estrogen receptor GPER regulates the migration and invasion of ovarian cancer cells. Mol Cell Biochem. 378:1–7. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Heublein S, Mayr D, Friese K, Jarrin-Franco MC, Lenhard M, Mayerhofer A and Jeschke U: The G-protein-coupled estrogen receptor (GPER/GPR30) in ovarian granulosa cell tumors. Int J Mol Sci. 15:15161–15172. 2014. View Article : Google Scholar : PubMed/NCBI

92 

Yan Y, Jiang X, Zhao Y, Wen H and Liu G: Role of GPER on proliferation, migration and invasion in ligand-independent manner in human ovarian cancer cell line SKOV3. Cell Biochem Funct. 33:552–559. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Ignatov T, Modl S, Thulig M, Weißenborn C, Treeck O, Ortmann O, Zenclussen A, Costa SD, Kalinski T and Ignatov A: GPER-1 acts as a tumor suppressor in ovarian cancer. J Ovarian Res. 6:512013. View Article : Google Scholar : PubMed/NCBI

94 

Predescu DV, Crețoiu SM, Crețoiu D, Pavelescu LA, Suciu N, Radu BM and Voinea SC: G protein-coupled receptors (GPCRs)-mediated calcium signaling in ovarian cancer: Focus on GPCRs activated by neurotransmitters and inflammation-associated molecules. Int J Mol Sci. 20:55682019. View Article : Google Scholar : PubMed/NCBI

95 

Xue D, Chen W and Neamati N: Discovery, structure-activity relationship study and biological evaluation of 2-thioureidothiophene-3-carboxylates as a novel class of C-X-C chemokine receptor 2 (CXCR2) antagonists. Eur J Med Chem. 204:1123872020. View Article : Google Scholar : PubMed/NCBI

96 

Seo MD, Velamakanni S, Ishiyama N, Stathopulos PB, Rossi AM, Khan SA, Dale P, Li C, Ames JB, Ikura M and Taylor CW: Structural and functional conservation of key domains in InsP3 and ryanodine receptors. Nature. 483:108–112. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Vermassen E, Parys JB and Mauger JP: Subcellular distribution of the inositol 1,4,5-trisphosphate receptors: Functional relevance and molecular determinants. Biol Cell. 96:3–17. 2004. View Article : Google Scholar : PubMed/NCBI

98 

Giannini G, Clementi E, Ceci R, Marziali G and Sorrentino V: Expression of a ryanodine receptor-Ca2+ channel that is regulated by TGF-beta. Science. 257:91–94. 1992. View Article : Google Scholar : PubMed/NCBI

99 

Santulli G, Nakashima R, Yuan Q and Marks AR: Intracellular calcium release channels: An update. J Physiol. 595:3041–3051. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Ando H, Hirose M and Mikoshiba K: Aberrant IP3 receptor activities revealed by comprehensive analysis of pathological mutations causing spinocerebellar ataxia 29. Proc Natl Acad Sci USA. 115:12259–12264. 2018. View Article : Google Scholar

101 

Díaz-Muñoz M, de la Rosa Santander P, Juárez-Espinosa AB, Arellano RO and Morales-Tlalpan V: Granulosa cells express three inositol 1,4,5-trisphosphate receptor isoforms: Cytoplasmic and nuclear Ca2+ mobilization. Reprod Biol Endocrinol. 6:602008. View Article : Google Scholar : PubMed/NCBI

102 

Hanson CJ, Bootman MD and Roderick HL: Cell signalling: IP3 receptors channel calcium into cell death. Curr Biol. 14:R933–R935. 2004. View Article : Google Scholar : PubMed/NCBI

103 

Lahiri S, Roy A, Li J, Mokashi A and Baby SM: Ca2+ responses to hypoxia are mediated by IP3-R on Ca2+ store depletion. Adv Exp Med Biol. 536:25–32. 2003. View Article : Google Scholar : PubMed/NCBI

104 

Lencesova L, Vlcek M, Krizanova O and Hudecova S: Hypoxic conditions increases H2S-induced ER stress in A2870 cells. Mol Cell Biochem. 414:67–76. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Yu Y, Xie Q, Liu W, Guo Y, Xu N, Xu L, Liu S, Li S, Xu Y and Sun L: Increased intracellular Ca2+ decreases cisplatin resistance by regulating iNOS expression in human ovarian cancer cells. Biomed Pharmacother. 86:8–15. 2017. View Article : Google Scholar

106 

Xie Q, Xu Y, Gao W, Zhang Y, Su J, Liu Y, Guo Y, Dou M, Hu K and Sun L: TAT-fused IP3R-derived peptide enhances cisplatin sensitivity of ovarian cancer cells by increasing ER Ca2+ release. Int J Mol Med. 41:809–817. 2018.

107 

Rezuchova I, Hudecova S, Soltysova A, Matuskova M, Durinikova E, Chovancova B, Zuzcak M, Cihova M, Burikova M, Penesova A, et al: Type 3 inositol 1,4,5-trisphosphate receptor has antiapoptotic and proliferative role in cancer cells. Cell Death Dis. 10:1862019. View Article : Google Scholar : PubMed/NCBI

108 

Sneyers F, Rosa N and Bultynck G: Type 3 IP3 receptors driving oncogenesis. Cell Calcium. 86:1021412020. View Article : Google Scholar

109 

Xue Y, Morris JL, Yang K, Fu Z, Zhu X, Johnson F, Meehan B, Witkowski L, Yasmeen A, Golenar T, et al: SMARCA4/2 loss inhibits chemotherapy-induced apoptosis by restricting IP3R3-mediated Ca2+ flux to mitochondria. Nat Commun. 12:54042021. View Article : Google Scholar

110 

Kucukkaya B, Erdag D, Akbas F and Yalcintepe L: The effect of iron on the expression levels of calcium related gene in cisplatin resistant epithelial ovarian cancer cells. Explor Target Antitumor Ther. 2:309–322. 2021.PubMed/NCBI

111 

Meissner G: The structural basis of ryanodine receptor ion channel function. J Gen Physiol. 149:1065–1089. 2017. View Article : Google Scholar : PubMed/NCBI

112 

Mariot P, Prevarskaya N, Roudbaraki MM, Le Bourhis X, Van Coppenolle F, Vanoverberghe K and Skryma R: Evidence of functional ryanodine receptor involved in apoptosis of prostate cancer (LNCaP) cells. Prostate. 43:205–214. 2000. View Article : Google Scholar : PubMed/NCBI

113 

Zhang L, Liu Y, Song F, Zheng H, Hu L, Lu H, Liu P, Hao X, Zhang W and Chen K: Functional SNP in the microRNA-367 binding site in the 3'UTR of the calcium channel ryanodine receptor gene 3 (RYR3) affects breast cancer risk and calcification. Proc Natl Acad Sci USA. 108:13653–13658. 2011. View Article : Google Scholar : PubMed/NCBI

114 

Schmitt K, Molfenter B, Laureano NK, Tawk B, Bieg M, Hostench XP, Weichenhan D, Ullrich ND, Shang V, Richter D, et al: Somatic mutations and promotor methylation of the ryanodine receptor 2 is a common event in the pathogenesis of head and neck cancer. Int J Cancer. 145:3299–3310. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Andruska ND, Zheng X, Yang X, Mao C, Cherian MM, Mahapatra L, Helferich WG and Shapiro DJ: Estrogen receptor α inhibitor activates the unfolded protein response, blocks protein synthesis, and induces tumor regression. Proc Natl Acad Sci USA. 112:4737–4742. 2015. View Article : Google Scholar

116 

Zheng X, Andruska N, Lambrecht MJ, He S, Parissenti A, Hergenrother PJ, Nelson ER and Shapiro DJ: Targeting multidrug-resistant ovarian cancer through estrogen receptor α dependent ATP depletion caused by hyperactivation of the unfolded protein response. Oncotarget. 9:14741–14753. 2018. View Article : Google Scholar

117 

Williams CJ and Erickson GF: Morphology and Physiology of the Ovary. Endotext. Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, et al: South Dartmouth, MA: MDText.com, Inc.; 2000

118 

Cui C, Merritt R, Fu L and Pan Z: Targeting calcium signaling in cancer therapy. Acta Pharm Sin B. 7:3–17. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Bowen NJ, Walker LD, Matyunina LV, Logani S, Totten KA, Benigno BB and McDonald JF: Gene expression profiling supports the hypothesis that human ovarian surface epithelia are multipotent and capable of serving as ovarian cancer initiating cells. BMC Med Genomics. 2:712009. View Article : Google Scholar : PubMed/NCBI

120 

Seo JA, Kim B, Dhanasekaran DN, Tsang BK and Song YS: Curcumin induces apoptosis by inhibiting sarco/endoplasmic reticulum Ca2+ ATPase activity in ovarian cancer cells. Cancer Lett. 371:30–37. 2016. View Article : Google Scholar

121 

Sun Z, Zhang H, Wang X, Wang QC, Zhang C, Wang JQ, Wang YH, An CQ, Yang KY, Wang Y, et al: TMCO1 is essential for ovarian follicle development by regulating ER Ca2+ store of granulosa cells. Cell Death Differ. 25:1686–1701. 2018. View Article : Google Scholar : PubMed/NCBI

122 

Huang N, Yu Y and Qiao J: Dual role for the unfolded protein response in the ovary: Adaption and apoptosis. Protein Cell. 8:14–24. 2017. View Article : Google Scholar :

123 

Peluso JJ: Basic fibroblast growth factor (bFGF) regulation of the plasma membrane calcium ATPase (PMCA) as part of an anti-apoptotic mechanism of action. Biochem Pharmacol. 66:1363–1369. 2003. View Article : Google Scholar : PubMed/NCBI

124 

Solár P and Sytkowski AJ: Differentially expressed genes associated with cisplatin resistance in human ovarian adenocarcinoma cell line A2780. Cancer Lett. 309:11–18. 2011. View Article : Google Scholar : PubMed/NCBI

125 

Kucukkaya B, Basoglu H, Erdag D, Akbas F, Susgun S and Yalcintepe L: Calcium homeostasis in cisplatin resistant epithelial ovarian cancer. Gen Physiol Biophys. 38:353–363. 2019. View Article : Google Scholar : PubMed/NCBI

126 

Baughman JM, Perocchi F, Girgis HS, Plovanich M, Belcher-Timme CA, Sancak Y, Bao XR, Strittmatter L, Goldberger O, Bogorad RL, et al: Integrative genomics identifies MCU as an essential component of the mitochondrial calcium uniporter. Nature. 476:341–345. 2011. View Article : Google Scholar : PubMed/NCBI

127 

Marchi S and Pinton P: The mitochondrial calcium uniporter complex: Molecular components, structure and physiopathological implications. J Physiol. 592:829–839. 2014. View Article : Google Scholar :

128 

Patron M, Checchetto V, Raffaello A, Teardo E, Vecellio Reane D, Mantoan M, Granatiero V, Szabò I, De Stefani D and Rizzuto R: MICU1 and MICU2 finely tune the mitochondrial Ca2+ uniporter by exerting opposite effects on MCU activity. Mol Cell. 53:726–737. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Denton RM: Regulation of mitochondrial dehydrogenases by calcium ions. Biochim Biophys Acta. 1787:1309–1316. 2009. View Article : Google Scholar : PubMed/NCBI

130 

Chakraborty PK, Mustafi SB, Xiong X, Dwivedi SKD, Nesin V, Saha S, Zhang M, Dhanasekaran D, Jayaraman M, Mannel R, et al: MICU1 drives glycolysis and chemoresistance in ovarian cancer. Nat Commun. 8:146342017. View Article : Google Scholar : PubMed/NCBI

131 

Hempel N and Trebak M: Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell Calcium. 63:70–96. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Porporato PE, Payen VL, Pérez-Escuredo J, De Saedeleer CJ, Danhier P, Copetti T, Dhup S, Tardy M, Vazeille T, Bouzin C, et al: A mitochondrial switch promotes tumor metastasis. Cell Rep. 8:754–766. 2014. View Article : Google Scholar : PubMed/NCBI

133 

Moloney JN and Cotter TG: ROS signalling in the biology of cancer. Semin Cell Dev Biol. 80:50–64. 2018. View Article : Google Scholar

134 

Ham J, Lim W, Kim K, Heo YM, Ryu SM, Lee D, Kim JJ and Song G: Gentisyl alcohol inhibits proliferation and induces apoptosis via mitochondrial dysfunction and regulation of MAPK and PI3K/AKT pathways in epithelial ovarian cancer cells. Mar Drugs. 17:3312019. View Article : Google Scholar : PubMed/NCBI

135 

Bae H, Park S, Ham J, Song J, Hong T, Choi JH, Song G and Lim W: ER-mitochondria calcium flux by β-sitosterol promotes cell death in ovarian cancer. Antioxidants (Basel). 10:15832021. View Article : Google Scholar

136 

Bae H, Park S, Yang C, Song G and Lim W: Disruption of endoplasmic reticulum and ROS production in human ovarian cancer by campesterol. Antioxidants (Basel). 10:3792021. View Article : Google Scholar : PubMed/NCBI

137 

Bae H, Song G and Lim W: Stigmasterol causes ovarian cancer cell apoptosis by inducing endoplasmic reticulum and mitochondrial dysfunction. Pharmaceutics. 12:4882020. View Article : Google Scholar : PubMed/NCBI

138 

Bae H, Lee JY, Song J, Song G and Lim W: Osthole interacts with an ER-mitochondria axis and facilitates tumor suppression in ovarian cancer. J Cell Physiol. 236:1025–1042. 2021. View Article : Google Scholar

139 

Bae H, Lee JY, Song G and Lim W: Fucosterol suppresses the progression of human ovarian cancer by inducing mitochondrial dysfunction and endoplasmic reticulum stress. Mar Drugs. 18:2612020. View Article : Google Scholar : PubMed/NCBI

140 

Bae H, Song G, Lee JY, Hong T, Chang MJ and Lim W: Laminarin-derived from brown algae suppresses the growth of ovarian cancer cells via mitochondrial dysfunction and ER stress. Mar Drugs. 18:1522020. View Article : Google Scholar : PubMed/NCBI

141 

Lim W, An Y, Yang C, Bazer FW and Song G: Chrysophanol induces cell death and inhibits invasiveness via mitochondrial calcium overload in ovarian cancer cells. J Cell Biochem. 119:10216–10227. 2018. View Article : Google Scholar : PubMed/NCBI

142 

Lim W, Ryu S, Bazer FW, Kim SM and Song G: Chrysin attenuates progression of ovarian cancer cells by regulating signaling cascades and mitochondrial dysfunction. J Cell Physiol. 233:3129–3140. 2018. View Article : Google Scholar

143 

Rogalska A, Szula E, Gajek A, Marczak A and Jóźwiak Z: Activation of apoptotic pathway in normal, cancer ovarian cells by epothilone B. Environ Toxicol Pharmacol. 36:600–610. 2013. View Article : Google Scholar : PubMed/NCBI

144 

Giorgi C, Baldassari F, Bononi A, Bonora M, De Marchi E, Marchi S, Missiroli S, Patergnani S, Rimessi A, Suski JM, et al: Mitochondrial Ca(2+) and apoptosis. Cell Calcium. 52:36–43. 2012. View Article : Google Scholar : PubMed/NCBI

145 

Honrath B, Metz I, Bendridi N, Rieusset J, Culmsee C and Dolga AM: Glucose-regulated protein 75 determines ER-mitochondrial coupling and sensitivity to oxidative stress in neuronal cells. Cell Death Discov. 3:170762017. View Article : Google Scholar

146 

Ma L, Wang H, Wang C, Su J, Xie Q, Xu L, Yu Y, Liu S, Li S, Xu Y and Li Z: Failure of elevating calcium induces oxidative stress tolerance and imparts cisplatin resistance in ovarian cancer cells. Aging Dis. 7:254–266. 2016. View Article : Google Scholar : PubMed/NCBI

147 

Xu L, Xie Q, Qi L, Wang C, Xu N, Liu W, Yu Y, Li S and Xu Y: Bcl-2 overexpression reduces cisplatin cytotoxicity by decreasing ER-mitochondrial Ca2+ signaling in SKOV3 cells. Oncol Rep. 39:985–992. 2018.

148 

Xie Q, Su J, Jiao B, Shen L, Ma L, Qu X, Yu C, Jiang X, Xu Y and Sun L: ABT737 reverses cisplatin resistance by regulating ER-mitochondria Ca2+ signal transduction in human ovarian cancer cells. Int J Oncol. 49:2507–2519. 2016. View Article : Google Scholar : PubMed/NCBI

149 

Li J, Qi F, Su H, Zhang C, Zhang Q, Chen Y, Chen P, Su L, Chen Y, Yang Y, et al: GRP75-faciliated mitochondria-associated ER membrane (MAM) integrity controls cisplatin-resistance in ovarian cancer patients. Int J Biol Sci. 18:2914–2931. 2022. View Article : Google Scholar : PubMed/NCBI

150 

Li L, Zeng S, Guo L, Huang P, Xi J, Feng J, Li Q, Li Y, Xiao X, Yan R and Zhang J: Long noncoding RNA RMRP contributes to paclitaxel sensitivity of ovarian cancer by regulating miR-580-3p/MICU1 signaling. J Oncol. 2022:83019412022.PubMed/NCBI

151 

Bresnick AR, Weber DJ and Zimmer DB: S100 proteins in cancer. Nat Rev Cancer. 15:96–109. 2015. View Article : Google Scholar : PubMed/NCBI

152 

Zimmer DB, Eubanks JO, Ramakrishnan D and Criscitiello MF: Evolution of the S100 family of calcium sensor proteins. Cell Calcium. 53:170–179. 2013. View Article : Google Scholar

153 

Bai Y, Li LD, Li J and Lu X: Prognostic values of S100 family members in ovarian cancer patients. BMC Cancer. 18:12562018. View Article : Google Scholar : PubMed/NCBI

154 

Hua X, Zhang H, Jia J, Chen S, Sun Y and Zhu X: Roles of S100 family members in drug resistance in tumors: Status and prospects. Biomed Pharmacother. 127:1101562020. View Article : Google Scholar : PubMed/NCBI

155 

Tian T, Li X, Hua Z, Ma J, Liu Z, Chen H and Cui Z: S100A1 promotes cell proliferation and migration and is associated with lymph node metastasis in ovarian cancer. Discov Med. 23:235–245. 2017.PubMed/NCBI

156 

Buckley NE, D'Costa Z, Kaminska M and Mullan PB: S100A2 is a BRCA1/p63 coregulated tumour suppressor gene with roles in the regulation of mutant p53 stability. Cell Death Dis. 5:e10702014. View Article : Google Scholar : PubMed/NCBI

157 

Kikuchi N, Horiuchi A, Osada R, Imai T, Wang C, Chen X and Konishi I: Nuclear expression of S100A4 is associated with aggressive behavior of epithelial ovarian carcinoma: An important autocrine/paracrine factor in tumor progression. Cancer Sci. 97:1061–1069. 2006. View Article : Google Scholar : PubMed/NCBI

158 

Horiuchi A, Hayashi T, Kikuchi N, Hayashi A, Fuseya C, Shiozawa T and Konishi I: Hypoxia upregulates ovarian cancer invasiveness via the binding of HIF-1α to a hypoxia-induced, methylation-free hypoxia response element of S100A4 gene. Int J Cancer. 131:1755–1767. 2012. View Article : Google Scholar : PubMed/NCBI

159 

Yan W, Chen J, Chen Z and Chen H: Deregulated miR-296/S100A4 axis promotes tumor invasion by inducing epithelial-mesenchymal transition in human ovarian cancer. Am J Cancer Res. 6:260–269. 2016.PubMed/NCBI

160 

Link T, Kuhlmann JD, Kobelt D, Herrmann P, Vassileva YD, Kramer M, Frank K, Göckenjan M, Wimberger P and Stein U: Clinical relevance of circulating MACC1 and S100A4 transcripts for ovarian cancer. Mol Oncol. 13:1268–1279. 2019. View Article : Google Scholar : PubMed/NCBI

161 

Deo AN, Thorat R, Dhadve AC, De A, Rekhi B and Ray P: IGF1R-α6 integrin-S100A4 network governs the organ-specific metastasis of chemoresistant epithelial ovarian cancer cells. Biochim Biophys Acta Mol Basis Dis. 1868:1662822022. View Article : Google Scholar

162 

Schäfer BW, Fritschy JM, Murmann P, Troxler H, Durussel I, Heizmann CW and Cox JA: Brain S100A5 is a novel calcium-, zinc-, and copper ion-binding protein of the EF-hand superfamily. J Biol Chem. 275:30623–30630. 2000. View Article : Google Scholar : PubMed/NCBI

163 

Wei BR, Hoover SB, Ross MM, Zhou W, Meani F, Edwards JB, Spehalski EI, Risinger JI, Alvord WG, Quiñones OA, et al: Serum S100A6 concentration predicts peritoneal tumor burden in mice with epithelial ovarian cancer and is associated with advanced stage in patients. PLoS One. 4:e76702009. View Article : Google Scholar : PubMed/NCBI

164 

Lin M, Xia B, Qin L, Chen H and Lou G: S100A7 regulates ovarian cancer cell metastasis and chemoresistance through MAPK signaling and is targeted by miR-330-5p. DNA Cell Biol. 37:491–500. 2018. View Article : Google Scholar : PubMed/NCBI

165 

Nymoen DA, Hetland Falkenthal TE, Holth A, Ow GS, Ivshina AV, Tropé CG, Kuznetsov VA, Staff AC and Davidson B: Expression and clinical role of chemoresponse-associated genes in ovarian serous carcinoma. Gynecol Oncol. 139:30–39. 2015. View Article : Google Scholar : PubMed/NCBI

166 

Lokman NA, Pyragius CE, Ruszkiewicz A, Oehler MK and Ricciardelli C: Annexin A2 and S100A10 are independent predictors of serous ovarian cancer outcome. Transl Res. 171:83–95.e1-e2. 2016. View Article : Google Scholar : PubMed/NCBI

167 

Wang L, Yan W, Li X, Liu Z, Tian T, Chen T, Zou L and Cui Z: S100A10 silencing suppresses proliferation, migration and invasion of ovarian cancer cells and enhances sensitivity to carboplatin. J Ovarian Res. 12:1132019. View Article : Google Scholar : PubMed/NCBI

168 

Xuan L, Sun Z, Wang J and Gao S: lncRNA SNHG8 promotes ovarian cancer progression through serving as sponge for miR-1270 to regulate S100A11 expression. J Gene Med. e33152021.

169 

Li W, Cui Z, Kong Y, Liu X and Wang X: Serum levels of S100A11 and MMP-9 in patients with epithelial ovarian cancer and their clinical significance. Biomed Res Int. 2021:73412472021.PubMed/NCBI

170 

Qian J, Ding F, Luo A, Liu Z and Cui Z: Overexpression of S100A14 in human serous ovarian carcinoma. Oncol Lett. 11:1113–1119. 2016. View Article : Google Scholar : PubMed/NCBI

171 

Sturchler E, Cox JA, Durussel I, Weibel M and Heizmann CW: S100A16, a novel calcium-binding protein of the EF-hand superfamily. J Biol Chem. 281:38905–38917. 2006. View Article : Google Scholar : PubMed/NCBI

172 

Yang T, Cheng J, Yang Y, Qi W, Zhao Y, Long H, Xie R and Zhu B: S100B mediates stemness of ovarian cancer stem-like cells through inhibiting p53. Stem Cells. 35:325–336. 2017. View Article : Google Scholar

173 

Yang T, Cheng J, You J, Yan B, Liu H and Li F: S100B promotes chemoresistance in ovarian cancer stem cells by regulating p53. Oncol Rep. 40:1574–1582. 2018.PubMed/NCBI

174 

Wang X, Tian T, Li X, Zhao M, Lou Y, Qian J, Liu Z, Chen H and Cui Z: High expression of S100P is associated with unfavorable prognosis and tumor progression in patients with epithelial ovarian cancer. Am J Cancer Res. 5:2409–2421. 2015.PubMed/NCBI

175 

Wang Q, He Z, Gao J, Hu S, Huang M, Liu M, Zheng J and Tang H: S100P sensitizes ovarian cancer cells to carboplatin and paclitaxel in vitro. Cancer Lett. 272:277–284. 2008. View Article : Google Scholar : PubMed/NCBI

176 

Ma N, Zhu L, Yang L, Cui Y and Zhan Y: Prognostic values of S100 family mRNA expression in ovarian cancer. Cancer Biomark. 25:67–78. 2019. View Article : Google Scholar : PubMed/NCBI

177 

Wu B, Yu C, Zhou B, Huang T, Gao L, Liu T and Yang X: Overexpression of TROP2 promotes proliferation and invasion of ovarian cancer cells. Exp Ther Med. 14:1947–1952. 2017. View Article : Google Scholar : PubMed/NCBI

178 

Dai S, Venturini E, Yadav S, Lin X, Clapp D, Steckiewicz M, Gocher-Demske AM, Hardie DG and Edelman AM: Calcium/calmodulin-dependent protein kinase kinase 2 mediates pleiotropic effects of epidermal growth factor in cancer cells. Biochim Biophys Acta Mol Cell Res. 1869:1192522022. View Article : Google Scholar : PubMed/NCBI

179 

Chen LL, Xia LY, Zhang JP, Wang Y, Chen JY, Guo C and Xu WH: Saikosaponin D alleviates cancer cachexia by directly inhibiting STAT3. Phytother Res. 37:809–819. 2023. View Article : Google Scholar

180 

Laski J, Singha B, Wang X, Valdés YR, Collins O and Shepherd TG: Activated CAMKKβ-AMPK signaling promotes autophagy in a spheroid model of ovarian tumour metastasis. J Ovarian Res. 13:582020. View Article : Google Scholar

181 

Chen Z, Sun X, Xia Z, Wang J, Guo N and Zhang Y: CaMKK2 promotes the progression of ovarian carcinoma through the PI3K/PDK1/Akt axis. Comput Math Methods Med. 2022:71879402022.PubMed/NCBI

182 

Tsuyoshi H, Wong VKW, Han Y, Orisaka M, Yoshida Y and Tsang BK: Saikosaponin-d, a calcium mobilizing agent, sensitizes chemoresistant ovarian cancer cells to cisplatin-induced apoptosis by facilitating mitochondrial fission and G2/M arrest. Oncotarget. 8:99825–99840. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2023
Volume 63 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Deng F, Fu M, Zhao C, Lei J, Xu T, Ji B, Ding H, Zhang Y, Chen J, Qiu J, Qiu J, et al: Calcium signals and potential therapy targets in ovarian cancer (Review). Int J Oncol 63: 125, 2023
APA
Deng, F., Fu, M., Zhao, C., Lei, J., Xu, T., Ji, B. ... Gao, Q. (2023). Calcium signals and potential therapy targets in ovarian cancer (Review). International Journal of Oncology, 63, 125. https://doi.org/10.3892/ijo.2023.5573
MLA
Deng, F., Fu, M., Zhao, C., Lei, J., Xu, T., Ji, B., Ding, H., Zhang, Y., Chen, J., Qiu, J., Gao, Q."Calcium signals and potential therapy targets in ovarian cancer (Review)". International Journal of Oncology 63.5 (2023): 125.
Chicago
Deng, F., Fu, M., Zhao, C., Lei, J., Xu, T., Ji, B., Ding, H., Zhang, Y., Chen, J., Qiu, J., Gao, Q."Calcium signals and potential therapy targets in ovarian cancer (Review)". International Journal of Oncology 63, no. 5 (2023): 125. https://doi.org/10.3892/ijo.2023.5573