Open access peer-reviewed chapter

Alterations in Calcium Signaling Pathways in Breast Cancer

Written By

Adrian Dumitru, Daniela Oana Toader, Sanda Maria Cretoiu, Dragos Cretoiu, Nicolae Suciu and Beatrice Mihaela Radu

Submitted: 14 February 2018 Reviewed: 08 August 2018 Published: 24 October 2018

DOI: 10.5772/intechopen.80811

From the Edited Volume

Calcium and Signal Transduction

Edited by John N. Buchholz and Erik J. Behringer

Chapter metrics overview

1,558 Chapter Downloads

View Full Metrics

Abstract

Breast cancer is the second most common cancer in women and the fifth cause contributing to death due to the cancer condition. It is essential to deeply understand the complex cellular mechanisms leading to this disease. There are multiple connections between calcium homeostasis alterations and breast cancer in the literature, but no consensus links the mechanism to the disease prognosis. Among the cells contributing to the breast cancer are the breast telocytes, which connect through gap junctions to other cells, including cancer cells and myoepithelial cells. Multiple proteins (i.e., voltage-gated calcium channels, transient receptor potential channels, STIM and Orai proteins, ether à go-go potassium channels, calcium-activated potassium channels, calcium-activated chloride channels, muscarinic acetylcholine receptors, etc.) coupled with calcium signaling pathways undergo functional and/or expression changes associated with breast cancer development and progression, and might represent promising pharmacological targets. Unraveling the mechanisms of altered calcium homeostasis in various breast cells due to the cancer condition might contribute to personalized therapeutic approaches.

Keywords

  • breast cancer
  • human breast stem cells
  • human breast epithelial cells
  • human breast myoepithelial cells
  • human breast adipocytes
  • human breast telocytes
  • calcium homeostasis alterations

1. Introduction

Breast cancer is the most common and the most frequently diagnosed cancer and is one of the most lethal malignant lesions in women worldwide, being the second leading cause of cancer death in women, after lung cancer [1].

1.1. Cell subtypes in human breast cancer and the alterations in calcium homeostasis

The mammary gland is an exocrine, compound tubuloalveolar gland [2]. Each mammary gland has 15–20 glandular lobes in its structure, each lobe being a separate gland with its excretory channel (galactophore channel) that opens at the level of the nipple via the galactophore pore [3]. The glandular lobe consists of glandular lobules delimited by fibrous connective tissue and fat. Lobules have a radiant arrangement, each opening through a lactiferous duct in the nipple, presenting a dilation called lactiferous sinus before opening. Lobules are composed of parenchyma and stroma, and made of loose connective tissue [4]. Each lobule consists of alveoli, lined by cuboidal epithelial cells, which secrete milk, and lactiferous ducts, lined by columnar epithelium, both epithelia surrounded by an outer layer of myoepithelial cells. The stromal cell population is composed of mesenchymal cells, including adipocytes, fibroblasts, and immune cells.

1.1.1. Human breast stem cells

The mammary gland structures are capable of self-renewal due to the presence of mammary stem cells (MSCs) and multipotent adult stem cells. Mammary stem cells lead to the growth of the mammary gland during puberty and are responsible for its further development during pregnancy. Besides their intrinsic self-renewal capacity, normal stem cells call for the adoption of other additional mechanisms to protect them from the microenvironmental pressure that may overdrive the stem cell pool [5]. MSCs are able to reconstitute a completely functional mammary gland upon orthotopic transplantation [6]. MSCs can differentiate into mature epithelial cells of either myoepithelial or luminal lineage through a series of lineage-restricted progenitor intermediate cells. Of major importance is also the long-term survival and expansion of MSCs [7] and the additional accumulation of genetic and/or epigenetic alterations in those cells that may increase the susceptibility of neoplastic transformation [8]. Van Keymeulen et al. demonstrated that the mammary gland contains different types of long-lived stem cells [9].

In breast cancer, both stem cells and progenitor cells are a potential candidate for tumorigenesis, and this could underpin the extraordinary phenotypic heterogeneity of malignant breast tumors. Also, different breast tumor subtypes might be linked to distinctive mammary stem cells and progenitor cells within the mammary epithelia as suggested by different studies [10]. Understanding how these cell subpopulations influence the normal epithelial differentiation hierarchy and the development of mammary tumors could have a significant impact on the proper taxonomy of these lesions.

Unlike the mouse counterparts, the detection of human breast stem cells may be difficult due to the lack of reliable surface makers. Also, the profile of MSCs is less predictable compared to mouse models [11].

To identify MSCs, several studies have employed multiple methods, including: 5-bromo-2-deoxy-uridine (BrdU) label-retention studies, nonadherent mammosphere cultures, cell-surface markers, such as Sca1 and CD49f, labeling MSCs with lipophilic fluorescent dye PKH26 during mammosphere growth and Hoechst dye efflux [12, 13]. These methods were very helpful for the further detection and characterization of signal transduction pathways such as the Notch, Wnt, and Hedgehog pathways that may be crucial for the self-renewal and fate determination of MSCs.

Identification of breast cancer stem cells is strictly dependent on cell-surface markers. Several markers have been proposed such as hyaluronan receptor (CD44), signal transducer CD24 (CD24), CD133 (Prominin-1), integrins CD29 (β1) and CD49f (α6), aldehyde dehydrogenase-1 (ALDH1), as tumor-initiating cells in breast cancer progression with high metastatic potential and in high-grade tumors resistant to therapeutic treatments [14, 15, 16, 17]. However, currently, there is no agreement regarding the phenotypic characterization of breast cancer stem cells. In addition to this discord, the great heterogeneity of breast tumors reflected by a myriad of histological subtypes with variable clinical presentations and diverse molecular signatures also contributes to this major shortcoming. The intrinsic molecular taxonomy describes five major subtypes of breast cancer (luminal-A, luminal-B, basal-like, HER2, and normal-like) which overlap with various clinicopathological classification systems and correlate with clinical behavior being vital for patient’s management. In addition, different breast cancer stem cells phenotypes have been described contributing to the proper characterization and nomenclature of breast malignant lesions. Several immunohistochemical markers have been characterized showing that the prevalence of stem cell-like markers varies according to tumor histological subtype [18].

In the light of these facts, some studies have proposed ALDH1 as an independent prognostic marker in breast cancer. Ginestier et al. showed a prevalence of 30% for ALDH1 positivity in a cohort comprising 577 breast tumors from two independent tumor sets. They also showed that ALDH1 expression correlates with a high histological grade, human epidermal growth factor receptor type 2 (HER2) overexpression, and absence of estrogen receptor and progesterone receptor expression [19]. A similar study [20] highlighted the worst prognosis of breast cancer patients with ALDH1 expression. There is no agreement in this matter, as other studies fail to find these correlations [21] even in more aggressive breast tumor subtypes such as inflammatory breast cancer.

With all these conflicting results, the reliability of ALDH1 expression as a clinical prognostic factor is doubtful, thus increasing the need for a standard protocol and more rigorous evaluation criteria, as well as consideration of the dissimilarity between whole-tissue staining versus tissue microarray staining [22].

Alterations in calcium homeostasis frequently occur in some pathological conditions such as malignant proliferation and could have a key role to play in the near future of some targeted therapeutic approaches. Some recent studies have shown that exposure of breast cancer cells to chemotherapy (i.e., carboplatin) induces Ca2+ release and leads to an enrichment of breast cancer stem cells [23]. Lu et al. have documented that chemotherapy induces the expression of glutathione S-transferase omega 1 (GSTO1), a factor which is dependent on hypoxia-inducible factor 1 (HIF-1) and HIF-2. In turn, low level of GSTO1 revokes carboplatin-induced breast cancer stem cell enrichment, decreasing tumor initiation and metastatic potential and delaying tumor recurrence after chemotherapy. The authors also found that GSTO1 interacts with the ryanodine receptor (RYR1) and increases calcium release from the endoplasmic reticulum. In this manner, high levels of cytosolic calcium activate proline-rich tyrosine kinase 2 (PYK2)/tyrosine-protein kinase (SRC)/signal transducers and activators of transcription factors 3 (STAT3) signaling pathways, leading to an increased expression of pluripotency factors and breast cancer stem cell enrichment. Concurrent HIF inhibition blocks chemotherapy-induced GSTO1 expression and breast cancer enrichment [23]. The authors have concluded that these combining effects may improve clinical outcome in breast cancer patients.

Not just chemotherapeutic agents are responsible for the release of free intracellular calcium. Petrou et al. investigated the effect of several ion channel modulators such as amiodarone, dofetilide, furosemide, minoxidil, loxapine, and nicorandil in prostate and breast cancer cell lines, PC3 and MCF7, respectively and found that in all investigated cases, calcium levels were increased by modulator concentrations comparable to those used clinically [24]. However, the way these modulators act on breast cancer stem cells remains unknown.

Calcium-and integrin-binding protein (CIB1) depletion impairs cell survival and tumor growth in triple-negative breast cancer by inducing genetic programs that reduce proliferation and survival and mediate differentiation and cancer stem cell function and epithelial to mesenchymal transition [25]. The authors also observed an almost complete cell death in MDA-468 cells after extended CIB1 depletion, suggesting that CIB1-depleted cells do not become stem cells, but rather gain some stem-like features as they are dying.

1.1.2. Human breast epithelial cells

The lactiferous ducts spread into the breast through a series of branches decreasing in caliber from the nipple to the terminal ductal-lobular units (TDLU) that are surrounded in specialized, hormonally responsive stroma. Extralobular ducts are lined by columnar epithelium that is supported by myoepithelial cells and a basement membrane composed of elastic fibers. The epithelium of the luminal duct can give rise to ductules, and fully formed lobules can originate directly from these anatomic structures in the nipple or at deeper levels of the mammary ductal system [26]. The lobules that consist of groups of alveolar glands encompassed by specialized vascularized stroma are connected by intralobular ductules that combine to form a single TDLU that drains into the extralobular ductal system.

The normal microscopic appearance of the lobules is not constant because the structure and histological appearance of the lobule in the mature breast are subject to individual changes associated with the menstrual cycle, pregnancy, lactation, exogenous hormone administration, therapy, aging, and menopause. The inactive lobular glands are lined by a single layer of cuboidal epithelial cells supported by underlying, loosely connected myoepithelial cells. The intralobular stroma contains more capillaries and is less densely collagenized than the interlobular surrounding stroma. Immunoreactivity for hormone receptors (ER and PR) is also variably expressed in lobules in a checkerboard staining pattern: the intensity and frequency of staining varies considerably from patient to patient and in many individual slides from one area to another. ER PR are sporadically positive in normal luminal epithelial cells, but some areas are, however, completely negative for those receptors. ER, PR, and AR are almost always negative in myoepithelial cells.

The 3D cultures of human breast epithelial cells have been designed in order to mimic the normal and pathological tissue architecture [27].

The regulation of signaling pathways and homeostasis of free intracellular Ca2+ can entail many cellular and physiological consequences, which may lead to changes in Ca2+ levels during lactation [28]. The Ca2+ influx has a decisive part in determining the concentration of Ca2+ in breast epithelial cells. Breast glandular proliferation, differentiation, and lactation are regulated by several local and systemic hormones, of which estrogen is one of the most important hormones. The regulators of estrogen and its receptor are modulators of proliferation and differentiation of breast epithelial cells [29]. The effect of estrogen on epithelial breast cells is done mainly through genomicpathways, but nongenomic mechanisms are particularly dependent on Ca2+ signaling [30].

Some studies on the MCF-7 breast cell line concluded that breast epithelium proliferation is influenced by Ca2+ through activation of mitogen-activated protein kinase (MAPK) by 17β-estradiol [31]. It is known that several Ca2+-related proteins can cause changes in cellular functions, leading to many breast lesions, including cancer and hypercalcemia-related malignancy, which have a poorer prognosis and have often a more aggressive nature been associated with metastasis [32]. The way calcium is involved in the differentiation of breast epithelial cells is closely dependent on vitamin D3. By modulating Ca2+ metabolism, vitamin D3 plays a crucial role in the regulation of cell proliferation and differentiation [33].

Various epidemiological studies [34, 35] suggest that vitamin D3 deficiency might increase cancer incidence, but no spontaneous tumors have been reported in mice models lacking 1,25(OH)2D3 or deficient in its receptor until recently [36]. The authors observed, for the first time, diverse types of spontaneous tumors in vitamin-D3-deficient mice for more than 1 year of age. The authors concluded that the tumors developed due to increased oxidative stress, cellular senescence, and senescence-associated secretory phenotype molecules, such as hepatocyte growth factor, mediated via its receptor c-Met. As such, vitamin D3 prevents tumorigenesis by inhibiting oxidative stress and inducing tumor cellular senescence in mice, and the study provides direct evidence supporting the role of vitamin D deficiency in increasing cancer incidence.

Calcium levels play an important role in mitochondria-induced apoptosis and epithelial breast cell necrosis [37], and reduction of Ca2+ content in the endoplasmic reticulum lumen is associated with resistance to apoptosis [38]. The release of calcium from the endoplasmic reticulum can be triggered by different molecules, even natural ones like resveratrol, a product commonly found in grapes. Resveratrol triggers the release of calcium from the endoplasmic reticulum, which in turn activates the calpain protease that ultimately leads to degradation of the plasma membrane by calcium-dependent ATPase isoform 1 [39].

Human breast epithelial cells with stem-like phenotype have been also demonstrated to be sensitive to the pathophysiological changes in calcium metabolism. To date, Wang et al. showed that how antioxidant medium is superior in terms of prolonged growth for normal breast epithelial cells that expressed stem cell phenotypes. The characteristics of these mammary stem cells include the deficiency in gap junctional intercellular communication, expression of Oct-4, and the ability to differentiate into basal epithelial cells and to form organoid showing mammary ductal and terminal end bud-like structures [40]. Their study concluded that using this new method of growing breast cancer epithelial cell with stem cell phenotype in a medium with low calcium levels and antioxidants will be of real use in the future studies of mammary development, breast carcinogenesis, chemoprevention, and cancer therapy.

1.1.3. Human breast myoepithelial cells

Myoepithelial cells lie between the epithelial cell layer and the basal lamina, where they establish a network of slender processes covering the overlying epithelial cells. The branching cytoplasmic network of the myoepithelial cell processes can be seen especially in scanning electron micrographs [41]. Spindle-shaped ductal myoepithelial cells lie parallel to the long axis of the duct and form a continuous layer. Contraction of myoepithelial cells in lobules and around ducts contributes to the flow of milk during lactation. The location of the myoepithelial cells between luminal epithelial cells and the basal lamina is an ideal location for them favoring communication with both compartments.

The histologic appearance and immunoreactivity of myoepithelial cells are highly variable, especially in pathologic conditions, and depend on the degree to which the myoid or epithelial phenotype is accentuated in a particular situation. Myoepithelial cells usually display nuclear reactivity for p63, which is the most useful marker for detecting these cells in normal and lesional tissues. Epithelioid myoepithelial cells can have reduced p63 reactivity. Other useful myoepithelial markers are α-smooth muscle actin, calponin, CD10, CKS/6, myosin, p75, and S100 (Figure 1, left). The presence or absence of myoepithelial cells, at least as demonstrated by routinely used immunostains and is very valuable in discriminating against neoplastic in situ lesions (Figure 1, right) versus malignant, infiltrative ones.

Figure 1.

(Left) Immunostain for S100 highlighting the myoepithelial cells layer in a normal terminal ductal lobular unit (TDLU)—green arrow and some adjacent adipocytes—yellow arrow showing strong nuclear and cytoplasmic positivity (immunostain S100 with DAB chromogen, 100× magnification). (Right) Immunostain for S100 highlighting the intact myoepithelial cell layer (blue arrow) at the periphery of some foci of high-grade ductal carcinoma in situ (DCIS) with central comedo-type necrosis (red triangle). Also, note the adjacent normal TDLU (green arrow) and adipocytes (yellow arrow). Immunostain S100 with DAB chromogen, 40× objective.

While the luminal epithelial cell has received plenty of attention as the most functionally active milk-producing cells and as the most probable target cell for tumorigenesis, attention on myoepithelial cells has begun to grow with the acknowledgment that these cells play an active role in branching morphogenesis and tumor suppression.

The function of myoepithelial cells is strongly dependent on regulation of intracellular calcium. These cells contract in response to oxytocin secretion during lactation to generate the contractile force required for milk ejection. It is difficult to understand whether the alteration of calcium metabolism of myoepithelial cells plays a role, if any, in carcinogenesis. Even if it has been demonstrated that store-operated Ca2+ entry was mediated by a functional Orai3 in estrogen receptor-expressing (ER+) breast cancer cells [42], the tumorigenesis impact of these findings on myoepithelial cell remains largely unknown, because it is known that most breast carcinomas originate in the epithelial cells and the spectrum of myoepithelial proliferative breast lesions in scarce.

However, disruption in calcium metabolism may alter the functionality of myoepithelial cells. On this issue, some studies have shown the important role of Orai1 store-operated calcium channels in lactation [43]. Davis et al., using genetically modified mouse models, observed that the store-operated Ca2+ channel Orai1 delivers over 50% of the calcium ions present in the secreted milk. They also demonstrated the role of Orai1 as a principal regulator of oxytocin-mediated alveolar unit contractility, milk ejection, general myoepithelial function, and survival.

S100 protein is expressed in myoepithelial cells. The S100 gene family is a Ca2+-binding protein with low molecular weight [44]. The members of the S100 family have a myriad of cell functions such as cell proliferation, apoptosis, differentiation, cancer invasion, and metastasis. S100A2 is involved in breast tumorigenesis being downregulated in some cases, which led to the invasion of breast cancer cells [45]. The S100A4 expression is associated with tumor progression and metastatic potential [46]. In a similar manner, S100A7 is not only overexpressed in high-grade ductal breast carcinoma but also in in situ high-grade lesion (DCIS), and some studies suggested that the concomitant expression of S100A7, S100A8, and S100A9 in a class of breast cancers was associated with poor prognosis [47].

1.1.4. Human breast adipocytes

Apart from the epigenetic and genetic changes that occur within epithelial cells leading to breast proliferative lesions, it has shown that tumor initiation and progression also depend on the intricate intercellular dialog between tumor epithelial cells and the surrounding stromal cells [48]. Among the different cell types comprised in the breast stroma, the most abundant are those of adipose origin, mainly mature adipocytes, preadipocytes, and adipose-derived stromal/stem cells (ASCs). Besides the structural role that breast tissue has, it also has an important bioactive function [49, 50]. In pathologic condition, it is of great importance the interaction that is established between tumor cells and stromal adipocytes within the invasive front characteristic of breast cancers [51, 52]. Similar to breast myoepithelial cells, breast adipocytes also express s100 protein—Figure 1 [53].

The role of calcium metabolism in such events is poorly understood, but some studies have found that some proteins such as calpains [54] and calpastatin [55] may be altered in adipose-derived stromal cells being responsible for their enhanced invasion potential.

Vitamin D3 triggers apoptosis in breast cancer cells and adipocytes by inducing an apoptotic signal by increasing the concentration of intracellular Ca2+. This signal acts as an apoptotic initiator that bluntly recruits calcium-dependent apoptotic effectors such as calpain and caspase 12, in both breast cancer cells and adipocytes. Some studies suggested that inducing apoptosis with vitamin D3, particularly in the tumor-surrounding adipose tissue involved in tumor progression, can contribute to the antitumoral effects of this hormone and may be of real therapeutic interest to include calcium-dependent apoptotic proteases as molecular targets for new therapeutic and preventive agents in breast cancer and obese patients [56].

1.1.5. Human breast telocytes

Recently, several papers were describing the presence of a new cellular type—the telocytes (TCs)—in the stroma of the mammary gland [57, 58, 59]. TCs are characterized by a small cellular body and extremely long telopodes with alternative regions of dilations called podoms and veil-like cytoplasmic extensions called podomeres [60, 61, 62]. Although there are numerous attempts to differentiate these cells from other cellular types such as fibroblasts, endothelial cells, mesenchymal stem cells, immune cells, a specific immunohistochemical marker was still not found [63]. The most specific markers, which are nowadays used for their identification, are CD34 and PDGFR alpha or beta [64, 65]. Genomic and proteomic approaches were also used to determine their uniqueness and have shown that telocytes are distinct from the other types listed above [66, 67, 68, 69, 70].

Mou et al. investigated the immunohistochemical characteristics and potential functions of TCs in reconstituted breast cancer tissue and found that they express c-kit/CD117, CD34, and vimentin. A very interesting observation is that TCs communicate with breast cancer cells as well as with other stromal cells [58, 71, 72]. Together with other stromal cells, TCs inhibited the breast cancer cell apoptosis and facilitated their proliferation and the formation of typical nest structure assembly in breast cancer, in vitro [58].

Rusu et al. described in an immunohistochemical study some CD34+/CD10±/c-kit-/vimentin-cells found in the inter- and intralobular stroma, which they considered to be TCs and suggested a stem cell-like features based on the expressed markers and changing phenotype [59]. Although a lot of studies are needed to talk about a certain function of TCs, a possible contribution to the mechanisms of carcinogenesis is not negligible, by the modification of the tumor microenvironment. As our team previously showed, TCs are not pacemakers but modulate the activity of the surrounding cells using calcium signaling [73]. Our results showed that uterine TCs express T-type calcium channels that might play a role in the generation of endogenous bioelectric signals responsible for the regulation of the surrounding cell behavior [74, 75].

Advertisement

2. Calcium signaling alterations in breast cancer

The most important proteins coupled with calcium signaling pathways and that have been described as key players in breast cancer cells are summarized in Figure 2.

Figure 2.

Proteins associated with calcium signaling pathways that have been described to undergo alterations in breast cancer cells.

2.1. Resting potential and calcium oscillations in breast cancer

The resting potential of human breast adenocarcinoma cells (estrogen receptor-positive MCF7 and triple-negative MDA-MB-231) is more positive (with approximately 27–30 mV) than normal human mammary epithelial cells [76]. Additionally, breast cancer cells react distinctly with respect to normal mammary epithelial cells in response to the changes in the extracellular ions (e.g., K+ or Ca2+) [76]. The differences in resting potential between breast cancer cells and normal cells may be useful in the development of anticancer-targeted therapies based on charged liposomes, which are considered among the promising liposome-based therapeutical approaches [77].

MDA-MB-231 breast cancer cells with high-metastatic potential also exhibit spontaneous Ca2+ oscillations in comparison with MCF7 breast cancer cells with a low-metastatic potential [78]. Interestingly, these spontaneous oscillations were absent in breast cancer cells with a low-metastatic potential, even in increased extracellular K+ concentration conditions that determined the augmentation of their basal Ca2+ level [78]. This feature of presenting spontaneous calcium oscillations in metastatic cells might be further exploited in understanding the cellular mechanisms standing behind and in finding adequate therapies.

2.2. Voltage-gated calcium channels in breast cancer

Patch-clamp studies on human mammary epithelial cells (HMEC) indicated the absence of voltage-gated calcium currents [79].

The contribution of T-type calcium channels to breast cancer was investigated in several studies. To date, the expression of the α1H subunit, but not the α1C or α1G subunits, of the voltage-gated calcium channels was demonstrated in MCF-7 breast cancer cells [80]. Among T-type calcium channels, Cav3.1, but not Cav3.2, was demonstrated to play an important role in the inhibition of proliferation and apoptosis in MCF-7 human breast cancer cells [81]. Patch-clamp recordings demonstrated the presence of T-type voltage-gated calcium currents (ICaT) in MCF7 breast cancer cells [79].

A meta-analysis of public microarray datasets in clinical cancer tissue samples identified the upregulation of several VGCCs transcripts (e.g., CACNA1C, CACNA1D, CACNA1B, CACNA1G, and CACNA1I) in breast cancer and their involvement in the development and cancer progression [82]. Oppositely, another meta-analysis indicated the downregulation of the same VGCCs transcripts in breast cancer [83].

2.3. Transient receptor potential channels in breast cancer

Transient receptor potential (TRP) channels have been documented to play an important role in the development and progression of cancer. TRPC1, TRPC6, TRPM7, TRPM8, and TRPV6 channels were described to be upregulated in human breast ductal adenocarcinoma in comparison with the adjacent nontumoral tissue, and correlations with the proliferative parameters or the invasiveness cell capacity have been evidenced [84]. Moreover, several studies documented the role of TRP channels in different breast cancer cell lines. In detail, TRPM7 was demonstrated by silencing experiments to contribute to the migration and invasiveness of MDA-MB-435 breast cancer cells by signaling through the MAPK, but not through Akt, pathway [85]. TRPM8 was detected in BT-474 and MDA-MB-231, but not in MCF7, breast carcinoma cells, while menthol-evoked Ca2+-oscillations were recorded in all three breast cancer cell lines [86]. TRPV6 had a high level of expression in breast adenocarcinoma tissue [87]. TRPV1 is functionally expressed in SUM149PT breast cancer cells, a model for a very aggressive form of breast cancer (i.e., triple-negative breast cancer) [88], and in MCF7 breast cancer cells [89, 90].

2.4. STIM and Orai proteins in breast cancer

In nonexcitable cells, such is the case of breast cells, both store-operated calcium entry (SOCE) and store-independent calcium pathways have been described, which involve the activation of stromal interaction molecules (STIM) and Orai proteins.

STIM/Orai proteins play an important role in the breast physiology. To date, in lactation, Orai1 is upregulated, while Stim1, but not Stim2, is downregulated [91]. Moreover, calcium-influx through Orai1 was described to be necessary for mammary epithelial cells for concentrating milk with Ca2+ and for milk expulsion through alveolar unit contraction [91].

In cancer metastasis, STIM/Orai proteins have been described to be involved by two major mechanisms: (i) upregulation or increased functional activation and (ii) molecular switching [92]. In estrogen-receptor-positive breast cancer cells (e.g., MCF7, BT474, ZR751, T47D, and HCC1500 cancerous cell lines) but not in estrogen-receptor-negative breast cancer cells (e.g., MCF10A and 184A1 normal breast epithelial cells, and MDA-MB231, BT20, and HCC1937 cancerous cell lines), Orai 3 mediates the STIM1/2 and Orai3 pathway and the Ca2+-selective Ca2+ release-activated Ca2+ current (I(CRAC)), while Orai1 does not mediate the STIM1/Orai1 pathway [42]. Further on, estrogen-receptor-α knockdown downregulates Orai3 without any effect on Orai1, decreases Orai3-mediated SOCE, and diminishes the I(CRAC) current in estrogen-receptor-positive MCF7 cells [93].

The expression of Orai3 was identified to be higher in MCF-7 breast cancer cells versus normal MCF-10A mammary epithelial cells, while its silencing inhibits the MCF-7 cell proliferation, arrests the cell cycle in the G1 phase, downregulates cyclin-dependent kinases 4/2, cyclins E and D1, and determines the accumulation of p21(Waf1/Cip1) (a cyclin-dependent kinase inhibitor) and p53 (a tumor-suppressing protein) [94].

In STIM1 siRNA- or Orai1 siRNA-treated MDA-MB-231 human breast tumor cells, a reduction in the migration process was identified [95]. Additionally, overexpression of STIM1 and Orai1 in MCF-10A epithelial cells increased their invasiveness [95]. Experiments on immunodeficient NOD/SCID mice injected with MDA-MB-231 human breast tumor cells were stably transfected with STIM1 siRNA, or Orai1 siRNA, but not with control siRNA, which demonstrated the inhibition of metastasis [95].

Interestingly, a signaling pathway independent of endoplasmic reticulum calcium stores or STIM1 and STIM2 protein activation was identified in MCF-7 breast cancer cells (the secretory pathway calcium ATPase 2 (SPCA2)/Orai1 signaling) [96], where SPCA2 is located in the Golgi apparatus.

2.5. Ether à go-go (hEag1) K+ channels in breast cancer

Several pieces of evidence indicate the role played by Ether à go-go (hEag1) K+ channels in breast cancer cells invasiveness. To date, blocking or silencing hEag1 channels in MDA-MB-231 breast cancer cells induces membrane depolarization and subsequent diminishment of Ca2+ influx through Orai1, which affects cell migration and proliferation [97].

2.6. Calcium-activated potassium channels in breast cancer

Prolactin increases the current density of the human Ca2+-activated K+ channels (hIKCa1) in MCF-7 breast cancer cells, involved in the cell proliferation in a dose-dependent manner and activating the Janus kinase (JAK2)-coupled cytokine receptor pathway [98].

Large conductance Ca2+-activated K+ channels (BKCa) were described to be expressed in several breast cancer cell lines (e.g., UACC893, SK-BR-3, and MDA-MB-231) [99]. Intermediate-conductance, Ca2+-activated K+ channels (hIK1) are also functionally expressed in MCF7 breast cancer cells, and their current density and basal cytosolic Ca2+ concentration being augmented in cells synchronized at the end of the G1 or S phase with respect to the cells in the early G1 phase [100]. Caveolin-1 was demonstrated to colocalize with BKCa in MCF-7 breast cancer cells, and silencing caveolin-1 induces increased activation and upregulation of BKCa [101].

2.7. Calcium-activated chloride channels in breast cancer

Calcium-activated chloride channel anoctamin 1 (ANO1) was demonstrated to be highly expressed in breast cancer cell lines and primary tumors and was considered to be a predictive factor for the disease degree and poor prognosis [102]. ANO1 activation was demonstrated to be done via the EGF receptor and calmodulin-dependent protein kinase II signaling, and its expression was associated with tumor cell survival [102].

An extensive clinical study including 431 patients with invasive ductal breast carcinoma and 46 patients with fibroadenoma analyzed the expression of anoctamin 1 (Ano1, TMEM16A), one of the members in the Ano family [103]. The study identified a correlation between Ano1 overexpression and the good prognosis in patients with lower clinical stage (stage I or II) of the breast cancer or in patients with triple-negative breast cancers [103].

Integrating these results, it is still premature to evaluate if Ano1 is or is not a predictive factor in good/poor prognosis in breast cancer patients, and if it is a suitable pharmacological target. It would be very useful to have more insights into the cellular mechanisms related to Ano1 activation. In our opinion, a possible scenario would be that the opening of calcium-activated chloride channels induces chloride efflux, membrane depolarization followed by the calcium influx through VGCCs. The high level of expression of calcium-activated chloride channels in breast cancer cells might be correlated with the tendency of these cells to be more depolarized with respect to the normal surrounding cells. However, this scenario would explain only the clinical data showing the association between Ano1 overexpression and the poor prognosis in patients with breast cancer.

2.8. Muscarinic acetylcholine receptors in breast cancer

Muscarinic receptors have been described to be expressed in several non-neuronal cell types, including endothelial cells, smooth muscle cells, or bladder and gastrointestinal tract [104, 105, 106, 107]. In multiple malignancies, including breast, prostate, lung, ovary, pancreas, prostate, skin, stomach, uterus and colon cancer, muscarinic receptors have been demonstrated to contribute to cell proliferation and cancer progression [108, 109, 110].

In particular, muscarinic receptors have also been described in normal murine mammary (NMuMG) cells, normal human-breast-derived MCF-10A cells, and homogenates of surgical samples derived from normal human mammary tissue by Western blot and radioligand binding assays [111, 112, 113]. The expression of muscarinic acetylcholine receptors, M3 and M4, was detected in MCF-7 breast cancer cells [114]. Additionally, the expression of muscarinic acetylcholine receptors was evidenced in LM2, LM3, and LMM3 cell lines derived from spontaneous mammary adenocarcinomas developed in Balb/C mice [115, 116, 117]. These studies demonstrated the involvement of muscarinic receptors in the tumor cells proliferation, progression, and angiogenesis [115, 116, 117], by a mechanism involving the stimulation of the nitric oxide synthase activity [114].

Muscarinic receptors, M1, M3 and M5, are coupled with Gq/11 proteins that activate phospholipase C (PLC) and determine the release of calcium from intracellular reservoirs. In MCF7, human breast cancer cells evidenced the activation of MAP by muscarinic receptors [118]. Considering that acetylcholine exerts neurocrine, paracrine, and autocrine regulation of cancer cell proliferation, migration, etc., we might consider muscarinic receptors among the master-players in breast cancer.

Advertisement

3. Calcium signaling pathways as pharmacological targets in breast cancer

3.1. Calcium oscillations as pharmacological targets in breast cancer

Tetrodotoxin (TTX), a blocker of voltage-gated sodium channels (VGSCs), was demonstrated to diminish the number of oscillating MDA-MB-231 breast cancer cells and to reduce the amplitude and the frequency of the Ca2+ oscillations (i.e., spontaneous calcium transients) in the same cells [78]. While TTX had no effect on the basal calcium level in nonoscillating MDA-MB-231 breast cells, when applied in high extracellular K+ conditions augmented the intracellular calcium concentration in both oscillating and nonoscillating MDA-MB-231 cells [78].

Previous studies have demonstrated that the blockade of VGSCs inhibits the invasion of endocrine-resistant breast cancer cells [119]. It is very interesting the correlation between the blockade of VGSCs and the reduction of spontaneous calcium oscillations. Therefore, considering that only metastatic cells are characterized by spontaneous calcium transients in comparison with low-metastatic or normal cells, a possible therapeutic strategy would be to diminish calcium transients by applying targeted pharmacological agents against VGSCs. An interesting approach would be to encapsulate voltage-gated sodium channels antagonists in charged liposomes and to target only highly metastatic breast cancer cells.

3.2. Voltage-gated calcium channels as pharmacological targets in breast cancer

1 mM Mn2+ and 0.1 mM Ni2+ ions blocked the fast activation and inactivation of the T-type calcium currents in MCF7 breast cancer cells [79].

Low doses (10–20 μM) of verapamil, an antagonist of voltage-gated calcium channels, blocked the growth of triple-negative MDA-MB-231 breast cancer cells, while high doses (100 μM) reduced by 90% the triple-negative MDA-MB-231 and MCF7 breast cancer cells [76].

Cav3.2 channels were demonstrated to play an important role in the mechanisms involved in chemoresistance. To date, trastuzumab resistance was demonstrated to be correlated with high mRNA Cav3.2 levels in SKBR3 breast cancer cells [120]. Patients with estrogen receptor-positive breast cancer that had a poor clinical outcome presented a significant Cav3.2 upregulation [120]. Moreover, patients with HER2-positive breast cancer presented a positive correlation between the Cav3.2 expression and patient survival upon chemotherapy [120].

3.3. TRP channels as pharmacological targets in breast cancer

TRP channels have been demonstrated to be actively involved in the development and progression of breast cancer. Therefore, finding efficient strategies for blocking-/silencing-specific TRPs in breast cancer cells might represent a good strategy to diminish the breast cancer progression.

Among the active compounds that act on TRP channels, polyunsaturated fatty acids (PUFA) have been evidenced to act on TRPC3, to reduce MCF-7 breast cancer cell proliferation and migration and to inhibit the TRPC-mediated calcium entry [80]. These compounds are particularly interesting as they are common in the diet, and previous studies have already shown their positive effects in cardiovascular diseases, including atherosclerosis, arrhythmias, etc. [121, 122]. The increased tumor vascularization is among the typical alterations and some PUFA have been demonstrated to exert effects against angiogenesis, inflammation, and cancer [123]. In this context, the action of PUFA on TRP channels is explaining their effects against breast tumors, either by diminishing angiogenesis in tumors or by reducing the proliferation/migration of tumor cells.

The Rho-associated kinases, ROCK1 and ROCK2, are considered as important therapeutic targets in breast cancer, being already demonstrated their critical role in cancer cell migration and invasion [124]. Identifying efficient Rho-kinase inhibitors is of particular interest in several subtypes of cancer, including breast cancer. Some Rho-kinase inhibitors have been identified to inhibit TRP channels. To date, fasudil, a Rho-kinase inhibitor, upregulates the expression of TRPC1 and TRPV2 in breast cancer cell lines (e.g., ZR-75-1, MCF7, and MDA-MB-231) [125]. Additionally, another Rho-kinase inhibitor, Y-27632, was demonstrated to downregulate TRPM6 and to upregulate TRPC7 in the same breast cancer cells [125].

TRPV1 seems to be one of the most promising TRP channels as a pharmacological target in breast cancer. To date, capsaicin inhibits cancer growth in SUM149PT breast cancer cells, by triggering apoptotic/necrotic mechanisms, while capsazepine diminishes these effects [88]. MRS1477, a positive allosteric modulator of TRPV1, when co-applied with capsaicin, diminished the fraction of MCF7 breast cancer apoptotic cells but was ineffective against tumor growth in MCF7 tumor-bearing immunodeficient mice [89]. Another study demonstrated that doxorubicin and melatonin exert synergistic effects against apoptosis and mitochondrial oxidative stress in MCF-7 breast cancer cells by activating TRPV1 [126]. The chemotherapy agent, 5-fluorouracil, also exerts its apoptotic effect in MCF7 breast cancer cells via TRPV1 channels [127]. The anticancer effects of cisplatin mediated by TRPV1 have to be potentiated by co-application of selenium or alpha-lipoic acid on MCF-7 breast cancer cells [128, 129]. Interestingly, selenium was shown to diminish the electromagnetic radiation (900 MHz) effects in MDA-MB-231 breast cancer cell line mediated by TRPV1 activation [130].

Although, tamoxifen is commonly known as a selective estrogen receptor modulator and used in estrogen receptor-positive breast cancer cells, it was also shown to be effective in estrogen-receptors negative tumors. Recent data indicate that it exerts antiproliferative effects in MCF7 breast cancer cells by an estrogen receptor-independent pathway that involves TRPV6 channels [87].

N-(3-aminopropyl)-2-([(3-methylphenyl)methyl]oxy)(20)-N-(2-thienylmethyl)benzamide (AMTB), an inhibitor of TRPM8, was also described to diminish the proliferation and migration of MDA-MB-231 and SK-BR-3 breast cancer cells via a TRPM8-independent mechanism involving voltage-gated sodium channels [131].

3.4. STIM and Orai as pharmacological targets in breast cancer

Blocking Ca2+ influx with EGTA, Ni2+, or SKF96365 in STIM1 siRNA-, or Orai1 siRNA-treated MDA-MB-231 human breast tumor cells decreased the number of invasive tumor cells [95]. BALB/c mice with 4 T1 tumor cells implanted in the mammary glands were injected with the store-operated channel blocker SKF96365 (10 mg/kg, daily, 20 days treatment), and the lung metastasis was diminished up to 20% [95]. Moreover, intraperitoneal administration of SKF96365 (10 mg/kg, daily, 4 weeks treatment) in NOD/SCID mice with MDA-MB-231 human breast tumor cells determined a significant reduction of lung metastasis after 1 week of treatment, and no metastasis recurrence was observed in 2 weeks after drug withdrawal [95].

3.5. hEAG1 channels as pharmacological targets in breast cancer

Astemizole, an antihistaminic drug, was shown to block hEAG1 channels in MDA-MB-231 breast cancer cells [97]. The authors have demonstrated that blocking hEag1 with astemizole or silencing induces the breast cancer depolarization and consequently reduces the calcium influx and the cell migration without any influence on the cell proliferation [97]. These data are clinically valuable as hEAG1 are overexpressed in invasive ductal carcinoma breast cancer or metastatic lymph nodes [97, 132] and their co-expression with HIF-1α is correlated with tumor size, lymph node status, and tumor stage [132], and the possibility of pharmacologically blocking these channels might represent a promising therapy. Moreover, astemizole may be used to pharmacologically discriminate hEAG from the related hERG potassium channels in MCF-7 breast cancer cells [133].

Insulin-like growth factor-1 (IGF1) is known to ubiquitary stimulate the growth of various cells in the human body, and also to strongly inhibit the programmed cell death [134]. IGF1 was shown to activate hEAG1 channels in breast cancer cells via an Akt-dependent signaling pathway [135]. Corroborating the antiapoptotic activity of IGF1 with its activatory effect exerted on hEAG1, we can conclude that hEAG1 plays an important role in breast cancer proliferation and its blocking is of particular interest in finding an efficient anticancer therapy.

3.6. Calcium-activated potassium channels as pharmacological targets in breast cancer

The hIKCa1 blockers, TRAM-34, and clotrimazole, or siRNA-hIKCa1 inhibit the prolactin-induced proliferation of the MCF7 breast cancer cells [98].

Iberiotoxin inhibits large conductance of Ca2+-activated K+ channels (BKCa) in three types of breast cancer cell lines (e.g., UACC893, SK-BR-3, and MDA-MB-231), eliciting cellular depolarization, attenuating the anchorage-independent growth [99]. Oppositely, HER-2/neu-overexpressing SK-BR-3 cells were insensitive to iberiotoxin [99].

3.7. Calcium-activated chloride channels as pharmacological targets in breast cancer

CaCCinh-A01, an inhibitor of calcium-activated chloride channels ANO1, diminishes breast cancer cell viability and colony formation [102]. Recent clinical studies showed that ANO1 is upregulated in breast cancer in comparison with fibroadenoma [103]. Moreover, patients with progesterone receptor-positive or HER2-negative breast cancer, or breast cancer patients treated with tamoxifen, have an upregulation of ANO1, which can be considered as a predictive factor for longer overall survival [103].

3.8. Muscarinic acetylcholine receptors as pharmacological targets in breast cancer

IgG purified from the serum of breast cancer patients mimics the effect of carbachol by activating muscarinic acetylcholine receptors in MCF-7 breast cancer cells [136]. Moreover, these autoantibodies purified from the serum of breast cancer patients regulate the MCF7 breast cancer cell migration and the MMP-9 activity, and these effects are reduced by atropine, 4-DAMP (M3 receptor antagonist), and tropicamide (M4 receptor antagonist) [137].

Carbachol, an agonist of muscarinic acetylcholine receptors, acts on M1 and M3 receptors in the MCF7 breast tumor cells and potentiates tumor progression, by activating nitric oxide synthase via phospholipase C and protein kinase C signaling pathways [114]. Carbachol also elicits the mobilization of intracellular-free Ca2+ and induces the phosphorylation of MAPK/ERK in MCF-7 human breast cancer cells, while pretreatment with wortmannin or LY294002 (selective inhibitors of phosphoinositide 3-kinase), with genistein (nonselective inhibitor of tyrosine kinases) or with PP2 (specific Src tyrosine kinase inhibitor), diminished the carbachol-induced MAPK/ERK phosphorylation [118].

Moreover, carbachol upregulates the vascular endothelial growth factor-A in MCF7 tumor cells and determines angiogenesis, while atropine reverts its effects [136]. Carbachol treatment (20 hours) increased the tumor cell death and its administration in subthreshold concentrations in conjunction with paclitaxel potentiates cell death [138, 139], while atropine reverts these combined effects [138]. Interestingly, the combined treatment with carbachol (low doses) and paclitaxel induced the death of breast tumor MCF-7 cells, via the increased activity of nitric oxide synthase 1 and 3, and the reduced activity of arginase II, but the drug combination was ineffective against the nontumorigenic epithelial MCF-10A cell line, due to the absence of muscarinic acetylcholine receptors [140].

Although several preclinical studies indicated the pharmacological potential of M3 antagonists in inhibiting tumor growth (e.g., melanoma, pancreatic, breast, ovarian, prostate, and brain cancers), no clinical trials have been done [109].

Advertisement

4. Conclusion

In conclusion, calcium signaling alterations occur in multiple cellular components, human breast stem cells, human breast epithelial cells, human breast myoepithelial cells, human breast adipocytes, human breast telocytes, etc., including those which contribute to the development and progression of breast cancer. Moreover, several molecular actors (e.g., voltage-gated calcium channels, TRP channels, STIM/Orai proteins, hEag1 K+ channels, calcium-activated potassium channels, calcium-activated chloride channels, muscarinic acetylcholine receptors, etc.) are playing an important role in calcium-altered homeostasis associated with breast cancer that might be considered as potential pharmacological targets. Considering the interplay between the above-described calcium signaling pathways, the most efficient strategy against breast cancer would simultaneously target several molecular players.

Advertisement

Acknowledgments

This work was supported by a grant of the Romanian Ministry of Research and Innovation, CCCDI—UEFISCDI, project number PN-III-P1—1.2-PCCDI-2017-0833/68/2018, within PNCDI III.

References

  1. 1. Bosetti C, Bertuccio P, Chatenoud L, Negri E, La Vecchia C, Levi F. Trends in mortality from urologic cancers in Europe, 1970-2008. European Urology. 2011;60(1):1-15
  2. 2. Ramsay DT, Kent JC, Hartmann RA, Hartmann PE. Anatomy of the lactating human breast redefined with ultrasound imaging. Journal of Anatomy. 2005;206(6):525-534
  3. 3. Tobon H, Salazar H. Ultrastructure of the human mammary gland. II. Postpartum lactogenesis. The Journal of Clinical Endocrinology and Metabolism. 1975;40(5):834-844
  4. 4. Vorherr H. The Breast: Morphology, Physiology and Lactation. London, UK: Academic Press; 1974
  5. 5. Celia-Terrassa T, Liu DD, Choudhury A, Hang X, Wei Y, Zamalloa J, et al. Normal and cancerous mammary stem cells evade interferon-induced constraint through the miR-199a-LCOR axis. Nature Cell Biology. 2017;19(6):711-723
  6. 6. Chen X, Liu Q, Song E. Mammary stem cells: Angels or demons in mammary gland? Signal Transduction and Targeted Therapy. 2017;2:16038
  7. 7. Dontu G, Al-Hajj M, Abdallah WM, Clarke MF, Wicha MS. Stem cells in normal breast development and breast cancer. Cell Proliferation. 2003;36(Suppl 1):59-72
  8. 8. Rangel MC, Bertolette D, Castro NP, Klauzinska M, Cuttitta F, Salomon DS. Developmental signaling pathways regulating mammary stem cells and contributing to the etiology of triple-negative breast cancer. Breast Cancer Research and Treatment. 2016;156(2):211-226
  9. 9. Van Keymeulen A, Rocha AS, Ousset M, Beck B, Bouvencourt G, Rock J, et al. Distinct stem cells contribute to mammary gland development and maintenance. Nature. 2011;479(7372):189-193
  10. 10. Bhagirath D, Zhao X, West WW, Qiu F, Band H, Band V. Cell type of origin as well as genetic alterations contribute to breast cancer phenotypes. Oncotarget. 2015;6(11):9018-9030
  11. 11. Wang D, Cai C, Dong X, Yu QC, Zhang XO, Yang L, et al. Identification of multipotent mammary stem cells by protein C receptor expression. Nature. 2015;517(7532):81-84
  12. 12. Woodward WA, Chen MS, Behbod F, Rosen JM. On mammary stem cells. Journal of Cell Science. 2005;118(Pt 16):3585-3594
  13. 13. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, et al. Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell. 2010;140(1):62-73
  14. 14. Li X, Lewis MT, Huang J, Gutierrez C, Osborne CK, Wu MF, et al. Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy. Journal of the National Cancer Institute. 2008;100(9):672-679
  15. 15. Tanei T, Morimoto K, Shimazu K, Kim SJ, Tanji Y, Taguchi T, et al. Association of breast cancer stem cells identified by aldehyde dehydrogenase 1 expression with resistance to sequential Paclitaxel and epirubicin-based chemotherapy for breast cancers. Clinical Cancer Research. 2009;15(12):4234-4241
  16. 16. Liu Q, Li JG, Zheng XY, Jin F, Dong HT. Expression of CD133, PAX2, ESA, and GPR30 in invasive ductal breast carcinomas. Chinese Medical Journal. 2009;122(22):2763-2769
  17. 17. Vassilopoulos A, Wang RH, Petrovas C, Ambrozak D, Koup R, Deng CX. Identification and characterization of cancer initiating cells from BRCA1 related mammary tumors using markers for normal mammary stem cells. International Journal of Biological Sciences. 2008;4(3):133-142
  18. 18. Park SY, Lee HE, Li H, Shipitsin M, Gelman R, Polyak K. Heterogeneity for stem cell-related markers according to tumor subtype and histologic stage in breast cancer. Clinical Cancer Research. 2010;16(3):876-887
  19. 19. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1(5):555-567
  20. 20. Morimoto K, Kim SJ, Tanei T, Shimazu K, Tanji Y, Taguchi T, et al. Stem cell marker aldehyde dehydrogenase 1-positive breast cancers are characterized by negative estrogen receptor, positive human epidermal growth factor receptor type 2, and high Ki67 expression. Cancer Science. 2009;100(6):1062-1068
  21. 21. Gong Y, Wang J, Huo L, Wei W, Ueno NT, Woodward WA. Aldehyde dehydrogenase 1 expression in inflammatory breast cancer as measured by immunohistochemical staining. Clinical Breast Cancer. 2014;14(3):e81-e88
  22. 22. Wolfe AR, Woodward WA. Breast cancer stem cell correlates as predicative factors for radiation therapy. Seminars in Radiation Oncology. 2015;25(4):251-259
  23. 23. Lu H, Chen I, Shimoda LA, Park Y, Zhang C, Tran L, et al. Chemotherapy-induced Ca(2+) release stimulates breast cancer stem cell enrichment. Cell Reports. 2017;18(8):1946-1957
  24. 24. Petrou T, Olsen HL, Thrasivoulou C, Masters JR, Ashmore JF, Ahmed A. Intracellular calcium mobilization in response to ion channel regulators via a calcium-induced calcium release mechanism. The Journal of Pharmacology and Experimental Therapeutics. 2017;360(2):378-387
  25. 25. Black JL, Harrell JC, Leisner TM, Fellmeth MJ, George SD, Reinhold D, et al. CIB1 depletion impairs cell survival and tumor growth in triple-negative breast cancer. Breast Cancer Research and Treatment. 2015;152(2):337-346
  26. 26. Rosen PP, Tench W. Lobules in the nipple. Frequency and significance for breast cancer treatment. Pathology Annual. 1985;20(Pt 2):317-322
  27. 27. Vidi PA, Bissell MJ, Lelievre SA. Three-dimensional culture of human breast epithelial cells: The how and the why. Methods in Molecular Biology. 2013;945:193-219
  28. 28. Hoenderop JG, Nilius B, Bindels RJ. Calcium absorption across epithelia. Physiological Reviews. 2005;85(1):373-422
  29. 29. Barnes DM, Millis RR, Gillett CE, Ryder K, Skilton D, Fentiman IS, et al. The interaction of oestrogen receptor status and pathological features with adjuvant treatment in relation to survival in patients with operable breast cancer: A retrospective study of 2660 patients. Endocrine-Related Cancer. 2004;11(1):85-96
  30. 30. Nadal A, Ropero AB, Laribi O, Maillet M, Fuentes E, Soria B. Nongenomic actions of estrogens and xenoestrogens by binding at a plasma membrane receptor unrelated to estrogen receptor alpha and estrogen receptor beta. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(21):11603-11608
  31. 31. Improta-Brears T, Whorton AR, Codazzi F, York JD, Meyer T, McDonnell DP. Estrogen-induced activation of mitogen-activated protein kinase requires mobilization of intracellular calcium. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(8):4686-4691
  32. 32. de Wit S, Cleton FJ. Hypercalcemia in patients with breast cancer: A survival study. Journal of Cancer Research and Clinical Oncology. 1994;120(10):610-614
  33. 33. Bikle DD, Oda Y, Xie Z. Calcium and 1,25(OH)2D: Interacting drivers of epidermal differentiation. The Journal of Steroid Biochemistry and Molecular Biology. 2004;89-90(1-5):355-360
  34. 34. Bentle MS, Reinicke KE, Bey EA, Spitz DR, Boothman DA. Calcium-dependent modulation of poly(ADP-ribose) polymerase-1 alters cellular metabolism and DNA repair. The Journal of Biological Chemistry. 2006;281(44):33684-33696
  35. 35. Monteith GR, McAndrew D, Faddy HM, Roberts-Thomson SJ. Calcium and cancer: Targeting Ca2+ transport. Nature Reviews. Cancer. 2007;7(7):519-530
  36. 36. Chen L, Yang R, Qiao W, Yuan X, Wang S, Goltzman D, et al. 1,25-Dihydroxy vitamin D prevents tumorigenesis by inhibiting oxidative stress and inducing tumor cellular senescence in mice. International Journal of Cancer. 2018
  37. 37. Schwab BL, Guerini D, Didszun C, Bano D, Ferrando-May E, Fava E, et al. Cleavage of plasma membrane calcium pumps by caspases: A link between apoptosis and necrosis. Cell Death and Differentiation. 2002;9(8):818-831
  38. 38. Pinton P, Rizzuto R. Bcl-2 and Ca2+ homeostasis in the endoplasmic reticulum. Cell Death and Differentiation. 2006;13(8):1409-1418
  39. 39. Sareen D, Darjatmoko SR, Albert DM, Polans AS. Mitochondria, calcium, and calpain are key mediators of resveratrol-induced apoptosis in breast cancer. Molecular Pharmacology. 2007;72(6):1466-1475
  40. 40. Wang KH, Kao AP, Chang CC, Lin TC, Kuo TC. Lifespan extension and sustained expression of stem cell phenotype of human breast epithelial stem cells in a medium with antioxidants. Stem Cells International. 2016;2016:4591310
  41. 41. Nagato T, Yoshida H, Yoshida A, Uehara Y. A scanning electron microscope study of myoepithelial cells in exocrine glands. Cell and Tissue Research. 1980;209(1):1-10
  42. 42. Motiani RK, Abdullaev IF, Trebak M. A novel native store-operated calcium channel encoded by Orai3: Selective requirement of Orai3 versus Orai1 in estrogen receptor-positive versus estrogen receptor-negative breast cancer cells. The Journal of Biological Chemistry. 2010;285(25):19173-19183
  43. 43. Davis FM, Janoshazi A, Janardhan KS, Steinckwich N, D'Agostin DM, Petranka JG, et al. Essential role of Orai1 store-operated calcium channels in lactation. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(18):5827-5832
  44. 44. Salama I, Malone PS, Mihaimeed F, Jones JL. A review of the S100 proteins in cancer. European Journal of Surgical Oncology. 2008;34(4):357-364
  45. 45. Vogl T, Leukert N, Barczyk K, Strupat K, Roth J. Biophysical characterization of S100A8 and S100A9 in the absence and presence of bivalent cations. Biochimica et Biophysica Acta. 2006;1763(11):1298-1306
  46. 46. Davies BR, Barraclough R, Davies MP, Rudland PS. Production of the metastatic phenotype by DNA transfection in a rat mammary model. Cell Biology International. 1993;17(9):871-879
  47. 47. Carlsson H, Petersson S, Enerback C. Cluster analysis of S100 gene expression and genes correlating to psoriasin (S100A7) expression at different stages of breast cancer development. International Journal of Oncology. 2005;27(6):1473-1481
  48. 48. Mueller MM, Fusenig NE. Friends or foes—Bipolar effects of the tumour stroma in cancer. Nature Reviews. Cancer. 2004;4(11):839-849
  49. 49. Wu Y, Kim JY, Zhou S, Smas CM. Differential screening identifies transcripts with depot-dependent expression in white adipose tissues. BMC Genomics. 2008;9:397
  50. 50. Park J, Euhus DM, Scherer PE. Paracrine and endocrine effects of adipose tissue on cancer development and progression. Endocrine Reviews. 2011;32(4):550-570
  51. 51. Iyengar P, Espina V, Williams TW, Lin Y, Berry D, Jelicks LA, et al. Adipocyte-derived collagen VI affects early mammary tumor progression in vivo, demonstrating a critical interaction in the tumor/stroma microenvironment. The Journal of Clinical Investigation. 2005;115(5):1163-1176
  52. 52. Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Research. 2011;71(7):2455-2465
  53. 53. Donato R, Cannon BR, Sorci G, Riuzzi F, Hsu K, Weber DJ, et al. Functions of S100 proteins. Current Molecular Medicine. 2013;13(1):24-57
  54. 54. Hanna RA, Campbell RL, Davies PL. Calcium-bound structure of calpain and its mechanism of inhibition by calpastatin. Nature. 2008;456(7220):409-412
  55. 55. Strong AL, Semon JA, Strong TA, Santoke TT, Zhang S, McFerrin HE, et al. Obesity-associated dysregulation of calpastatin and MMP-15 in adipose-derived stromal cells results in their enhanced invasion. Stem Cells. 2012;30(12):2774-2783
  56. 56. Sergeev IN. Regulation of apoptosis in adipocytes and breast cancer cells by 1,25-dihydroxyvitamin D3: A link between obesity and breast cancer. Hormone Molecular Biology and Clinical Investigation. 2013;14(3):99-106
  57. 57. Popescu LM, Faussone-Pellegrini MS. TELOCYTES—A case of serendipity: The winding way from interstitial cells of Cajal (ICC), via interstitial Cajal-like cells (ICLC) to TELOCYTES. Journal of Cellular and Molecular Medicine. 2010;14(4):729-740
  58. 58. Mou Y, Wang Y, Li J, Lu S, Duan C, Du Z, et al. Immunohistochemical characterization and functional identification of mammary gland telocytes in the self-assembly of reconstituted breast cancer tissue in vitro. Journal of Cellular and Molecular Medicine. 2013;17(1):65-75
  59. 59. Petre N, Rusu MC, Pop F, Jianu AM. Telocytes of the mammary gland stroma. Folia Morphologica. 2016;75(2):224-231
  60. 60. Cretoiu D, Xu J, Xiao J, Cretoiu SM. Telocytes and their extracellular vesicles-evidence and hypotheses. International Journal of Molecular Sciences. 2016;17(8)
  61. 61. Cretoiu D, Cretoiu SM. Telocytes in the reproductive organs: Current understanding and future challenges. Seminars in Cell & Developmental Biology. 2016;55:40-49
  62. 62. Cretoiu D, Radu BM, Banciu A, Banciu DD, Cretoiu SM. Telocytes heterogeneity: From cellular morphology to functional evidence. Seminars in Cell & Developmental Biology. 2017;64:26-39
  63. 63. Cretoiu SM. Immunohistochemistry of telocytes in the uterus and fallopian tubes. Advances in Experimental Medicine and Biology. 2016;913:335-357
  64. 64. Cretoiu SM, Popescu LM. Telocytes revisited. Biomolecular Concepts. 2014;5(5):353-369
  65. 65. Campeanu RA, Radu BM, Cretoiu SM, Banciu DD, Banciu A, Cretoiu D, et al. Near-infrared low-level laser stimulation of telocytes from human myometrium. Lasers in Medical Science. 2014;29(6):1867-1874
  66. 66. Zheng M, Sun X, Zhang M, Qian M, Zheng Y, Li M, et al. Variations of chromosomes 2 and 3 gene expression profiles among pulmonary telocytes, pneumocytes, airway cells, mesenchymal stem cells and lymphocytes. Journal of Cellular and Molecular Medicine. 2014;18(10):2044-2060
  67. 67. Zheng Y, Cretoiu D, Yan G, Cretoiu SM, Popescu LM, Fang H, et al. Protein profiling of human lung telocytes and microvascular endothelial cells using iTRAQ quantitative proteomics. Journal of Cellular and Molecular Medicine. 2014;18(6):1035-1059
  68. 68. Zheng Y, Cretoiu D, Yan G, Cretoiu SM, Popescu LM, Wang X. Comparative proteomic analysis of human lung telocytes with fibroblasts. Journal of Cellular and Molecular Medicine. 2014;18(4):568-589
  69. 69. Sun X, Zheng M, Zhang M, Qian M, Zheng Y, Li M, et al. Differences in the expression of chromosome 1 genes between lung telocytes and other cells: Mesenchymal stem cells, fibroblasts, alveolar type II cells, airway epithelial cells and lymphocytes. Journal of Cellular and Molecular Medicine. 2014;18(5):801-810
  70. 70. Song D, Cretoiu D, Zheng M, Qian M, Zhang M, Cretoiu SM, et al. Comparison of chromosome 4 gene expression profile between lung telocytes and other local cell types. Journal of Cellular and Molecular Medicine. 2016;20(1):71-80
  71. 71. Popescu LM, Gherghiceanu M, Cretoiu D, Radu E. The connective connection: Interstitial cells of Cajal (ICC) and ICC-like cells establish synapses with immunoreactive cells. Electron microscope study in situ. Journal of Cellular and Molecular Medicine. 2005;9(3):714-730
  72. 72. Cretoiu D, Cretoiu SM, Simionescu AA, Popescu LM. Telocytes, a distinct type of cell among the stromal cells present in the lamina propria of jejunum. Histology and Histopathology. 2012;27(8):1067-1078
  73. 73. Radu BM, Banciu A, Banciu DD, Radu M, Cretoiu D, Cretoiu SM. Calcium signaling in interstitial cells: Focus on telocytes. International Journal of Molecular Sciences. 2017;18(2):397. DOI: 10.3390/ijms18020397
  74. 74. Cretoiu SM, Cretoiu D, Marin A, Radu BM, Popescu LM. Telocytes: Ultrastructural, immunohistochemical and electrophysiological characteristics in human myometrium. Reproduction. 2013;145(4):357-370
  75. 75. Cretoiu SM, Radu BM, Banciu A, Banciu DD, Cretoiu D, Ceafalan LC, et al. Isolated human uterine telocytes: Immunocytochemistry and electrophysiology of T-type calcium channels. Histochemistry and Cell Biology. 2015;143(1):83-94
  76. 76. Berzingi S, Newman M, Yu HG. Altering bioelectricity on inhibition of human breast cancer cells. Cancer Cell International. 2016;16:72
  77. 77. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Frontiers in Pharmacology. 2015;6:286
  78. 78. Rizaner N, Onkal R, Fraser SP, Pristera A, Okuse K, Djamgoz MB. Intracellular calcium oscillations in strongly metastatic human breast and prostate cancer cells: Control by voltage-gated sodium channel activity. European Biophysics Journal. 2016;45(7):735-748
  79. 79. Yu HG, McLaughlin S, Newman M, Brundage K, Ammer A, Martin K, et al. Altering calcium influx for selective destruction of breast tumor. BMC Cancer. 2017;17(1):169
  80. 80. Zhang H, Zhou L, Shi W, Song N, Yu K, Gu Y. A mechanism underlying the effects of polyunsaturated fatty acids on breast cancer. International Journal of Molecular Medicine. 2012;30(3):487-494
  81. 81. Ohkubo T, Yamazaki J. T-type voltage-activated calcium channel Cav3.1, but not Cav3.2, is involved in the inhibition of proliferation and apoptosis in MCF-7 human breast cancer cells. International Journal of Oncology. 2012;41(1):267-275
  82. 82. Wang CY, Lai MD, Phan NN, Sun Z, Lin YC. Meta-analysis of public microarray datasets reveals voltage-gated calcium gene signatures in clinical cancer patients. PLoS One. 2015;10(7):e0125766
  83. 83. Phan NN, Wang CY, Chen CF, Sun Z, Lai MD, Lin YC. Voltage-gated calcium channels: Novel targets for cancer therapy. Oncology Letters. 2017;14(2):2059-2074
  84. 84. Dhennin-Duthille I, Gautier M, Faouzi M, Guilbert A, Brevet M, Vaudry D, et al. High expression of transient receptor potential channels in human breast cancer epithelial cells and tissues: Correlation with pathological parameters. Cellular Physiology and Biochemistry. 2011;28(5):813-822
  85. 85. Meng X, Cai C, Wu J, Cai S, Ye C, Chen H, et al. TRPM7 mediates breast cancer cell migration and invasion through the MAPK pathway. Cancer Letters. 2013;333(1):96-102
  86. 86. Naziroglu M, Blum W, Josvay K, Cig B, Henzi T, Olah Z, et al. Menthol evokes Ca(2+) signals and induces oxidative stress independently of the presence of TRPM8 (menthol) receptor in cancer cells. Redox Biology. 2018;14:439-449
  87. 87. Bolanz KA, Kovacs GG, Landowski CP, Hediger MA. Tamoxifen inhibits TRPV6 activity via estrogen receptor-independent pathways in TRPV6-expressing MCF-7 breast cancer cells. Molecular Cancer Research. 2009;7(12):2000-2010
  88. 88. Weber LV, Al-Refae K, Wolk G, Bonatz G, Altmuller J, Becker C, et al. Expression and functionality of TRPV1 in breast cancer cells. Breast Cancer. 2016;8:243-252
  89. 89. Naziroglu M, Cig B, Blum W, Vizler C, Buhala A, Marton A, et al. Targeting breast cancer cells by MRS1477, a positive allosteric modulator of TRPV1 channels. PLoS One. 2017;12(6):e0179950
  90. 90. Cig B, Naziroglu M. Investigation of the effects of distance from sources on apoptosis, oxidative stress and cytosolic calcium accumulation via TRPV1 channels induced by mobile phones and Wi-Fi in breast cancer cells. Biochimica et Biophysica Acta. 2015;1848(10 Pt B):2756-2765
  91. 91. McAndrew D, Grice DM, Peters AA, Davis FM, Stewart T, Rice M, et al. ORAI1-mediated calcium influx in lactation and in breast cancer. Molecular Cancer Therapeutics. 2011;10(3):448-460
  92. 92. Vashisht A, Trebak M, Motiani RK. STIM and Orai proteins as novel targets for cancer therapy. A review in the theme: Cell and molecular processes in cancer metastasis. American Journal of Physiology: Cell Physiology. 2015;309(7):C457-C469
  93. 93. Motiani RK, Zhang X, Harmon KE, Keller RS, Matrougui K, Bennett JA, et al. Orai3 is an estrogen receptor alpha-regulated Ca(2)(+) channel that promotes tumorigenesis. The FASEB Journal. 2013;27(1):63-75
  94. 94. Faouzi M, Hague F, Potier M, Ahidouch A, Sevestre H, Ouadid-Ahidouch H. Down-regulation of Orai3 arrests cell-cycle progression and induces apoptosis in breast cancer cells but not in normal breast epithelial cells. Journal of Cellular Physiology. 2011;226(2):542-551
  95. 95. Yang S, Zhang JJ, Huang XY. Orai1 and STIM1 are critical for breast tumor cell migration and metastasis. Cancer Cell. 2009;15(2):124-134
  96. 96. Feng M, Grice DM, Faddy HM, Nguyen N, Leitch S, Wang Y, et al. Store-independent activation of Orai1 by SPCA2 in mammary tumors. Cell. 2010;143(1):84-98
  97. 97. Hammadi M, Chopin V, Matifat F, Dhennin-Duthille I, Chasseraud M, Sevestre H, et al. Human ether a-gogo K(+) channel 1 (hEag1) regulates MDA-MB-231 breast cancer cell migration through Orai1-dependent calcium entry. Journal of Cellular Physiology. 2012;227(12):3837-3846
  98. 98. Faouzi M, Chopin V, Ahidouch A, Ouadid-Ahidouch H. Intermediate Ca2+-sensitive K+ channels are necessary for prolactin-induced proliferation in breast cancer cells. The Journal of Membrane Biology. 2010;234(1):47-56
  99. 99. Schickling BM, England SK, Aykin-Burns N, Norian LA, Leslie KK, Frieden-Korovkina VP. BKCa channel inhibitor modulates the tumorigenic ability of hormone-independent breast cancer cells via the Wnt pathway. Oncology Reports. 2015;33(2):533-538
  100. 100. Ouadid-Ahidouch H, Roudbaraki M, Delcourt P, Ahidouch A, Joury N, Prevarskaya N. Functional and molecular identification of intermediate-conductance Ca(2+)-activated K(+) channels in breast cancer cells: Association with cell cycle progression. American Journal of Physiology. Cell Physiology. 2004;287(1):C125-C134
  101. 101. Du C, Chen L, Zhang H, Wang Z, Liu W, Xie X, et al. Caveolin-1 limits the contribution of BKCa channel to MCF-7 breast cancer cell proliferation and invasion. International Journal of Molecular Sciences. 2014;15(11):20706-20722
  102. 102. Britschgi A, Bill A, Brinkhaus H, Rothwell C, Clay I, Duss S, et al. Calcium-activated chloride channel ANO1 promotes breast cancer progression by activating EGFR and CAMK signaling. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(11):E1026-E1034
  103. 103. Wu H, Guan S, Sun M, Yu Z, Zhao L, He M, et al. Ano1/TMEM16A overexpression is associated with good prognosis in PR-positive or HER2-negative breast cancer patients following tamoxifen treatment. PLoS One. 2015;10(5):e0126128
  104. 104. Eglen RM, Reddy H, Watson N, Challiss RA. Muscarinic acetylcholine receptor subtypes in smooth muscle. Trends in Pharmacological Sciences. 1994;15(4):114-119
  105. 105. Uchiyama T, Chess-Williams R. Muscarinic receptor subtypes of the bladder and gastrointestinal tract. Journal of Smooth Muscle Research = Nihon Heikatsukin Gakkai kikanshi. 2004;40(6):237-247
  106. 106. Eglen RM. Overview of muscarinic receptor subtypes. Handbook of Experimental Pharmacology. 2012;208:3-28
  107. 107. Radu BM, Osculati AMM, Suku E, Banciu A, Tsenov G, Merigo F, et al. All muscarinic acetylcholine receptors (M1–M5) are expressed in murine brain microvascular endothelium. Scientific Reports. 2017;7(1):5083
  108. 108. Shah N, Khurana S, Cheng K, Raufman JP. Muscarinic receptors and ligands in cancer. American Journal of Physiology. Cell Physiology. 2009;296(2):C221-C232
  109. 109. Spindel ER. Muscarinic receptor agonists and antagonists: Effects on cancer. Handbook of Experimental Pharmacology. 2012;208:451-468
  110. 110. Kruse AC, Kobilka BK, Gautam D, Sexton PM, Christopoulos A, Wess J. Muscarinic acetylcholine receptors: Novel opportunities for drug development. Nature Reviews Drug Discovery. 2014;13(7):549-560
  111. 111. Espanol AJ, Sales ME. Different muscarinc receptors are involved in the proliferation of murine mammary adenocarcinoma cell lines. International Journal of Molecular Medicine. 2004;13(2):311-317
  112. 112. Fiszman G, Cattaneo V, de la Torre E, Espanol A, Colombo L, Sacerdote de Lustig E, et al. Muscarinic receptors autoantibodies purified from mammary adenocarcinoma-bearing mice sera stimulate tumor progression. International Immunopharmacology. 2006;6(8):1323-1330
  113. 113. Negroni MP, Fiszman GL, Azar ME, Morgado CC, Espanol AJ, Pelegrina LT, et al. Immunoglobulin G from breast cancer patients in stage I stimulates muscarinic acetylcholine receptors in MCF7 cells and induces proliferation. Participation of nitric oxide synthase-derived nitric oxide. Journal of Clinical Immunology. 2010;30(3):474-484
  114. 114. Fiszman GL, Middonno MC, de la Torre E, Farina M, Espanol AJ, Sales ME. Activation of muscarinic cholinergic receptors induces MCF-7 cells proliferation and angiogenesis by stimulating nitric oxide synthase activity. Cancer Biology & Therapy. 2007;6(7):1106-1113
  115. 115. Espanol A, Eijan AM, Mazzoni E, Davel L, Jasnis MA, Sacerdote De Lustig E, et al. Nitric oxide synthase, arginase and cyclooxygenase are involved in muscarinic receptor activation in different murine mammary adenocarcinoma cell lines. International Journal of Molecular Medicine. 2002;9(6):651-657
  116. 116. Rimmaudo LE, de la Torre E, Sacerdote de Lustig E, Sales ME. Muscarinic receptors are involved in LMM3 tumor cells proliferation and angiogenesis. Biochemical and Biophysical Research Communications. 2005;334(4):1359-1364
  117. 117. Espanol AJ, de la Torre E, Fiszman GL, Sales ME. Role of non-neuronal cholinergic system in breast cancer progression. Life Sciences. 2007;80(24-25):2281-2285
  118. 118. Jimenez E, Montiel M. Activation of MAP kinase by muscarinic cholinergic receptors induces cell proliferation and protein synthesis in human breast cancer cells. Journal of Cellular Physiology. 2005;204(2):678-686
  119. 119. Mohammed FH, Khajah MA, Yang M, Brackenbury WJ, Luqmani YA. Blockade of voltage-gated sodium channels inhibits invasion of endocrine-resistant breast cancer cells. International Journal of Oncology. 2016;48(1):73-83
  120. 120. Pera E, Kaemmerer E, Milevskiy MJG, Yapa K, O'Donnell JS, Brown MA, et al. The voltage gated Ca(2+)-channel Cav3.2 and therapeutic responses in breast cancer. Cancer Cell International. 2016;16:24
  121. 121. Ander BP, Dupasquier CM, Prociuk MA, Pierce GN. Polyunsaturated fatty acids and their effects on cardiovascular disease. Experimental and Clinical Cardiology. 2003;8(4):164-172
  122. 122. Szostak-Wegierek D, Klosiewicz-Latoszek L, Szostak WB, Cybulska B. The role of dietary fats for preventing cardiovascular disease. A review. Roczniki Panstwowego Zakladu Higieny. 2013;64(4):263-269
  123. 123. Wang W, Zhu J, Lyu F, Panigrahy D, Ferrara KW. Hammock B, et al. omega-3 polyunsaturated fatty acids-derived lipid metabolites on angiogenesis, inflammation and cancer. Prostaglandins & Other Lipid Mediators. 2014;113-115:13-20
  124. 124. Patel RA, Forinash KD, Pireddu R, Sun Y, Sun N, Martin MP, et al. RKI-1447 is a potent inhibitor of the Rho-associated ROCK kinases with anti-invasive and antitumor activities in breast cancer. Cancer Research. 2012;72(19):5025-5034
  125. 125. Gogebakan B, Bayraktar R, Suner A, Balakan O, Ulasli M, Izmirli M, et al. Do fasudil and Y-27632 affect the level of transient receptor potential (TRP) gene expressions in breast cancer cell lines? Tumour Biology. 2014;35(8):8033-8041
  126. 126. Kosar PA, Naziroglu M, Ovey IS, Cig B. Synergic effects of doxorubicin and melatonin on apoptosis and mitochondrial oxidative stress in MCF-7 breast cancer cells: Involvement of TRPV1 channels. The Journal of Membrane Biology. 2016;249(1-2):129-140
  127. 127. Deveci HA, Naziroglu M, Nur G. 5-Fluorouracil-induced mitochondrial oxidative cytotoxicity and apoptosis are increased in MCF-7 human breast cancer cells by TRPV1 channel activation but not Hypericum perforatum treatment. Molecular and Cellular Biochemistry. 2018;439(1-2):189-198
  128. 128. Nur G, Naziroglu M, Deveci HA. Synergic prooxidant, apoptotic and TRPV1 channel activator effects of alpha-lipoic acid and cisplatin in MCF-7 breast cancer cells. Journal of Receptor and Signal Transduction Research. 2017;37(6):569-577
  129. 129. Sakalli Cetin E, Naziroglu M, Cig B, Ovey IS, Aslan Kosar P. Selenium potentiates the anticancer effect of cisplatin against oxidative stress and calcium ion signaling-induced intracellular toxicity in MCF-7 breast cancer cells: Involvement of the TRPV1 channel. Journal of Receptor and Signal Transduction Research. 2017;37(1):84-93
  130. 130. Kahya MC, Naziroglu M, Cig B. Selenium reduces mobile phone (900 MHz)-induced oxidative stress, mitochondrial function, and apoptosis in breast cancer cells. Biological Trace Element Research. 2014;160(2):285-293
  131. 131. Yapa K, Deuis J, Peters AA, Kenny PA, Roberts-Thomson SJ, Vetter I, et al. Assessment of the TRPM8 inhibitor AMTB in breast cancer cells and its identification as an inhibitor of voltage gated sodium channels. Life Sciences. 2018;198:128-135
  132. 132. Lai Q, Wang T, Guo Q, Zhang Y, Wang Y, Yuan L, et al. Positive correlation between the expression of hEag1 and HIF-1alpha in breast cancers: An observational study. BMJ Open. 2014;4(5):e005049
  133. 133. Roy J, Vantol B, Cowley EA, Blay J, Linsdell P. Pharmacological separation of hEAG and hERG K+ channel function in the human mammary carcinoma cell line MCF-7. Oncology Reports. 2008;19(6):1511-1516
  134. 134. Peruzzi F, Prisco M, Dews M, Salomoni P, Grassilli E, Romano G, et al. Multiple signaling pathways of the insulin-like growth factor 1 receptor in protection from apoptosis. Molecular and Cellular Biology. 1999;19(10):7203-7215
  135. 135. Borowiec AS, Hague F, Harir N, Guenin S, Guerineau F, Gouilleux F, et al. IGF-1 activates hEAG K(+) channels through an Akt-dependent signaling pathway in breast cancer cells: Role in cell proliferation. Journal of Cellular Physiology. 2007;212(3):690-701
  136. 136. Lombardi MG, Negroni MP, Pelegrina LT, Castro ME, Fiszman GL, Azar ME, et al. Autoantibodies against muscarinic receptors in breast cancer: Their role in tumor angiogenesis. PLoS One. 2013;8(2):e57572
  137. 137. Pelegrina LT, Lombardi MG, Fiszman GL, Azar ME, Morgado CC, Sales ME. Immunoglobulin g from breast cancer patients regulates MCF-7 cells migration and MMP-9 activity by stimulating muscarinic acetylcholine receptors. Journal of Clinical Immunology. 2013;33(2):427-435
  138. 138. Espanol AJ, Jacob G, Dmytrenko G, Sales ME. Muscarinic activation enhances the anti-proliferative effect of paclitaxel in murine breast tumor cells. Anti-Cancer Agents in Medicinal Chemistry. 2013;13(8):1273-1279
  139. 139. Sales ME. Muscarinic receptors as targets for metronomic therapy in breast cancer. Current Pharmaceutical Design. 2016;22(14):2170-2177
  140. 140. Espanol AJ, Salem A, Rojo D, Sales ME. Participation of non-neuronal muscarinic receptors in the effect of carbachol with paclitaxel on human breast adenocarcinoma cells. Roles of nitric oxide synthase and arginase. International Immunopharmacology. 2015;29(1):87-92

Written By

Adrian Dumitru, Daniela Oana Toader, Sanda Maria Cretoiu, Dragos Cretoiu, Nicolae Suciu and Beatrice Mihaela Radu

Submitted: 14 February 2018 Reviewed: 08 August 2018 Published: 24 October 2018