Научная статья на тему 'Adhesion Proteins Profile and Localization in Ovarian Carcinoma Cell Lines'

Adhesion Proteins Profile and Localization in Ovarian Carcinoma Cell Lines Текст научной статьи по специальности «Биотехнологии в медицине»

CC BY
93
19
i Надоели баннеры? Вы всегда можете отключить рекламу.
Ключевые слова
cell–cell contacts / cell-extracellular matrix contacts / cancer cell phenotype / adherens junctions / gap junctions / integrin-based junctions

Аннотация научной статьи по биотехнологиям в медицине, автор научной работы — O.M. Kutova, A.D. Pospelov, L.V. Krylova, E.N. Gorshkova, I.V. Balalaeva

The three-dimensional structure of tumor tissue and particularly cell-cell and cell-extracellular matrix adhesion is an important factor that can determine the phenotype of tumor cells. In this work, we have investigated the abundance profile of actin-binding adhesion proteins in human ovarian adenocarcinoma cell lines SKOV-3 and SKOV3.ip. We have investigated levels of total and superficially localized adherens junctions proteins Eand N-cadherin, gap junction protein сonnexin-43 and cell-extracellular matrix contacting integrin beta-1. Our results indicate a complete absence of epithelial marker E-cadherin, a low level of mesenchymal N-cadherin and high levels of connexin-43 and integrin beta-1. Modest superficial localization of the represented proteins was observed, indicating their mislocalization. SKOV-3 cell line was characterized by higher levels of the total content of studied cell-cell contacts proteins and a lower level of superficially localized integrin beta-1, which is both considered to be associated with lower tumor aggressiveness. The revealed differences in the profile of adhesion proteins are in line with the accepted view on SKOV3.ip cell line having a more aggressive phenotype than that of SKOV-3. The revealed features of the total abundance of the adhesion proteins and their superficially localized pool made it possible to supplement the information on the nature of phenotypic differences between the studied cell lines.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «Adhesion Proteins Profile and Localization in Ovarian Carcinoma Cell Lines»

ADHESION PROTEINS PROFILE AND LOCALIZATION IN OVARIAN CARCINOMA CELL LINES

O.M. Kutova, A.D. Pospelov, L.V. Krylova, E.N. Gorshkova, I.V. Balalaeva*

Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, 603950,Russia.

* Corresponding author: irin-b@mail.ru

Abstract. The three-dimensional structure of tumor tissue and particularly cell-cell and cell-extracellular matrix adhesion is an important factor that can determine the phenotype of tumor cells. In this work, we have investigated the abundance profile of actin-binding adhesion proteins in human ovarian adenocarcinoma cell lines SKOV-3 and SKOV-3.ip. We have investigated levels of total and superficially localized adherens junctions proteins E- and N-cadherin, gap junction protein connexin-43 and cell-extracellular matrix contacting integrin beta-1. Our results indicate a complete absence of epithelial marker E-cadherin, a low level of mesenchymal N-cadherin and high levels of connexin-43 and integrin beta-1. Modest superficial localization of the represented proteins was observed, indicating their mislocaliza-tion. SKOV-3 cell line was characterized by higher levels of the total content of studied cell-cell contacts proteins and a lower level of superficially localized integrin beta-1, which is both considered to be associated with lower tumor aggressiveness. The revealed differences in the profile of adhesion proteins are in line with the accepted view on SKOV-3.ip cell line having a more aggressive phenotype than that of SKOV-3. The revealed features of the total abundance of the adhesion proteins and their superficially localized pool made it possible to supplement the information on the nature of phenotypic differences between the studied cell lines.

Keywords: cell-cell contacts, cell-extracellular matrix contacts, cancer cell phenotype, adherens junctions, gap junctions, integrin-based junctions.

List of Abbreviations

SMT - somatic mutation theory TOFT - tissue organization field theory ECM - extracellular matrix RF - relative fluorescence EMT - epithelial-to-mesenchymal transition WNT - Wingless-integrated GJIC - gap junction intercellular communication

EGF - epidermal growth factor Bax - Bcl2 associated X protein FAK - focal adhesion kinase

Introduction

Contribution of the three-dimensional structure of the tumor to its development and drug resistance is now a cutting edge in cancer research. Thus the prevailing 'somatic mutation theory' (SMT) of carcinogenesis which declares that cancer stems from one compromised cell is now being revised and complemented by elements of 'tissue organization field theory' (TOFT) of cancer, which in turn suggests that the development of the

tumor process lies in the disorder of the interaction between all of the participants of the tissue microenvironment (Soto, Sonnenschein., 2020; Neophytou et al, 2021). Disturbance in cellular communication has been widely described for tumors of various locations and stages of development and is being considered as a potential target for various treatment approaches (Brucher & Jamall., 2014; Song et al., 2019; Dominiak et al., 2020).

Communication within the tissue is mediated by cell-cell contacts (adherens, tight, gap junctions and desmosomes) and cell-extracellular matrix (ECM) contacts (mainly integrin-based) comprised by complexes of integral proteins. Adherens junctions are comprised by calcium-dependent proteins cadherins which organize in belt-like adhesion plaques thus providing the organization of the epithelial sheet and participate in maintaining its polarization along with tight junctions and cell-ECM contacts (Harris & Tepass, 2010). Tight junctions are comprised by caludins and occludin

which form the most prohibitive junctions strictly regulating paracellular transport (Tsu-kita et al, 2001). Gap junctions constituted by connexins on contrary act as direct cell connectors as they form hexameric transmembrane channels (Nielsen et al., 2012). Desmosomes are formed by desmogleins and desmocollins which are calcium-independent non-classical cadherins (Garrod & Chidgey, 2008). Integrin-based contacts to ECM are dimers of various alpha- and beta-integrin subunits, where the specificity of adhesion is defined by the composition of a dimer (Takada et al., 2007). Concerning intracellular connections of integral adhesion proteins, they bind to various components of the cytoskeleton: desmosomes bind to intermediate filaments (Hatzfeld et al., 2017), while proteins of adherens and tight junctions bind to the actin cytoskeleton (Campbell et al., 2017; Vicente-Manzanares et al., 2009). Emerging data testifies that gap junction proteins con-nexins are also connected to actin cytoskeleton via adapter proteins which bind to specific sites on their cytoplasmatic C-tail (Herve et al., 2007; Ambrosi et al, 2016).

Actin cytoskeleton is recognized as one of the central components orchestrating cancer progression and metastasis (Galluzzi & Thomas, 2020 a,b), thus profile assessment of actin-connected adhesion proteins may be a relevant tool in cancer research. Integral proteins in general are the first-line contributors to adhesion and communication and in these terms their representation profile serves as a reflection of interactions in tumor microenvironment in a certain time point and becomes a potent characteristic which is appropriate to rely on both in fundamental cancer research and drug efficiency evaluation (Farahani et al, 2014; Kutova et al., 2020). In this study we focus on representative integral actin-binding proteins of adherens junctions, namely epithelial marker E-cadherin and mesenchymal marker N-cadherin (Loh et al., 2019); gap junctions, namely con-nexin-43 which is the most widely represented connexin in mammalian tissues (Bonacquisti & Nguyen, 2019); and integrin-based cell-ECM contacts, namely integrin beta-1, which is the most preferential partner in a-ß integrin dimer-

ization and thus may be considered as the basic member of the majority of integrin-based contacts (Hynes, 1992). A summary of the studied proteins and their common binding partners among adapter proteins are represented in Fig. 1.

It should be noted that to date a considerable amount of data was obtained indicating mislo-calization of adhesion proteins (Wang & Li, 2014; Alaga et al., 2017; Seraya-Bareket et al., 2020) which shifts cancer cells towards aggressiveness. Our work aims to investigate the expression profile and localization of actin binding integral adhesion proteins representing cell-cell and cell-matrix contacts in two lines of human ovarian adenocarcinoma cells with different aggressiveness.

Materials and Methods

Cell lines. Cells of human ovarian adenocarcinoma cell lines SKOV-3 and SKOV-3.ip were used. Cells were cultured in DMEM medium containing 2 mM glutamine (PanEco, Russia), 10% (v/v) fetal bovine serum (Hy-Clone, USA), 50pg/ml of Penicillin and 50 pg/ml of Streptomycin (PanEco, Russia) at 37 °C in 5% CO2. For passaging, cells were detached with Versene solution (PanEco, Russia).

Cell preparation protocol to assess localization of adhesion proteins. In order to perform flow cytometry analysis, monolayer culture was detached from the substrate by incubation with TrypLE solution (Thermo Fischer Scientific, USA) for 20 min at 37 °C in 5% CO2. Cells were fixed in 4% formaldehyde (Appli-chem, Germany) for 20 min at room temperature to exclude a possibility of protein profile changes during the analysis. In order to assess the simultaneous superficial and intracellular abundance of studied proteins fixed cells were permeabilized with 0,02% Triton X-100 (VWR Life Science, USA) for 20 min at room temperature. Cells intended for assessing the abundance of superficially located proteins of interest proceeded to staining directly after fixation. Cells were thoroughly washed with PBS to remove formaldehyde and Triton-X100 before staining.

Fig. 1. Summary of studied proteins: E-cadherin, N-cadherin (cell-cell adherens junctions); connexin-43 (cell-cell gap junctions); integrin beta-1 (cell-matrix junctions) with common adapter proteins

Cell staining andflow cytometry analysis. In order to minimize non-specific binding, cells were blocked in a solution of 3% milk (Appli-chem, Germany) in PBS for 1 h at room temperature and incubated with antibodies specific to the proteins of interest and antibodies of corresponding isotypic control according to manufacturer's instructions. The following specific antibodies were used: E-cadherin Monoclonal Antibody (67A4), FITC (Thermo Fischer Scientific, USA, Cat#A15757), N-cadherin Monoclonal Antibody (8C11), PE (Thermo Fischer Scientific, USA, Cat#12-3259-42), connexin-43 Monoclonal Antibody (CX-1B1) Alexa Fluor 488 (Thermo Fischer Scientific, USA, Cat#138388), integrin beta-1 Monoclonal Antibody (TS2/16), PE (Thermo Fischer Scientific, USA, Cat#12-0299-42). Excess antibodies were washed out from the specimens with 1% BSA (Sigma-Al-drich, USA) solution in PBS and analyzed by flow cytometry using a CytoFlex S (Beckman Coulter, USA). Fluorescence of FITC, Alexa-Fuor 488 and PE was excited with a 488-nm laser and the signal was detected with 525/40 (FITC, AlexaFluor488) and 585/42 (PE) band pass filters.

Results

Two ovarian carcinoma cell lines SKOV-3 and SKOV-3.ip were used in this study. SKOV-3 cell line was obtained from the malignant ascites of a 64-year-old Caucasian female (Hung et al., 1992) and SKOV-3.ip cell line was obtained from the malignant ascites of tumor bearing nu/nu mice which were intraperi-toneally inoculated with SKOV-3 cells. SKOV-3.ip cells were indicated to possess higher DNA synthesis rates, accelerated proliferation, increased colony-formation in soft agar, formation of larger subcutaneous tumors and reduced survival of nu/nu mice bearing in-traperitoneally inoculated tumors (Yu et al., 1993; Dar et al., 2017).

To evaluate the localization of adhesion proteins in monolayer cultures in ovarian carcinoma cell lines we have conducted flow cytom-etry analysis of adhesion proteins participating in formation of cell-cell contacts, namely E-cadherin and N-cadherin (adherens junctions), connexin-43 (gap junctions) and cell-ECM contacts namely integrin beta-1. The abundance level of each protein of interest is represented by relative fluorescence (RF) value calculated

Fig. 2. The distributions of SKOV-3 and SKOV-3. ip cells according to presence of total (A, upper plot) and superficially located (B, lower plot) E-cadherin. Cells were stained with E-cadherin-specific antibodies and analyzed by flow cytometry

Fig. 3. The distributions of SKOV-3 and SKOV-3. ip cells according to presence of total (A, upper plot) and superficially located (B, lower plot) N-cadherin. Cells were stained with N-cadherin-specific antibodies and analyzed by flow cytometry

as a ratio of mean fluorescence intensity of cells stained with antibodies specific to proteins of interest to mean fluorescence intensity of cells stained with antibodies of corresponding iso-typic control. RF value equal to 1 indicates that no analyzed protein is present in the cells.

E cadherin. The obtained data indicate that the studied cells do not possess E-cadherin at all; relative fluorescence values were equal or close to 1 (Figs 2, 6, 7).

N-cadherin was represented at very low level in both cell lines. Along with that, population heterogeneity was detected: a considerable part of the cells demonstrated the complete absence of N-cadherin (Fig. 3). Only a modest part of total N-cadherin was superficially located (Fig. 6). Of note, the total abundance of N-cadherin in N-cadherin-positive cells was significantly higher in SKOV-3 cells in comparison to SKOV-3. ip cells (Fig. 7,B).

Fig. 4. The distributions of SKOV-3 and SKOV-3. ip cells according to presence of total (A, upper plot) and superficially located (B, lower plot) connexin-43. Cells were stained with connexin-43 -specific antibodies and analyzed by flow cytometry

Fig. 5. The distributions of SKOV-3 and SKOV-3. ip cells according to presence of total (A, upper plot) and superficially located (B, lower plot) integrin beta-1. Cells were stained with integrin beta-1-spe-cific antibodies and analyzed by flow cytometry

Connexin-43 total representation was at a relatively high level both in SKOV-3 and SKOV-3.ip cell lines, at the same time similarly to N-cadherin, only a modest level of connexin-43 was represented at the cell surface (not more than 20% of total amount) (Figs 4, 6) and also similarly to N-cadherin, connexin-43 was significantly more abundant in SKOV-3 cell line compared to SKOV-3.ip (Fig. 7, B).

Integrin beta-1 was the most abundant of the studied proteins both in SKOV-3 and SKOV-3.ip cell lines and again only a part of

the total integrin beta-1 content was superficially localized. Also the studied cell lines showed heterogeneity as a minor part of the population represented loss of integrin beta-1 (Figs 5, 6). Statistically significant differences between the studied cell lines were detected, moreover, the peculiar phenomenon was revealed for integrin beta-1: SKOV-3 cell line possessed higher total integrin beta-1 level (Fig. 2,A) while superficially located integrin beta-1 prevailed in SKOV-3.ip cell line (Fig. 2,B).

Fig. 6. The abundance of total and superficially located adhesion proteins in SKOV-3 (A) and SKOV-3.ip (B) cell lines. Expression levels of E-cadherin, N-cadherin, connexin-43 and integrin beta-1 denoted as relative fluorescence values calculated as a ratio of mean fluorescence intensity of cells stained with specific antibodies to mean fluorescence intensity of cells stained with antibodies of corresponding isotypic control. Data are presented as mean ± SD (n = 3). "*" indicates significant difference in RF between total and superficially located protein (Unpaired t-test with Welch correction, p < 0.05)

Fig. 7. The abundance of adhesion proteins in ovarian adenocarcinoma cell lines totally represented (A) and located superficially (B). Expression levels of E-cadherin, N-cadherin, connexin-43 and integrin beta-1 denoted as relative fluorescence values calculated as a ratio of mean fluorescence intensity of cells stained with specific antibodies to mean fluorescence intensity of cells stained with antibodies of corresponding isotypic control. Data are presented as mean ± SD (n = 3). "*" indicates significant difference in RF between protein abundance in studied cell lines (Unpaired t-test with Welch correction, p < 0.05)

Discussion

Intercellular contact proteins are usually considered to be tumor suppressors, but there is more and more conflicting evidence of their involvement in the formation of an aggressive phenotype and triggering tumor progression and invasion. An intriguing potential reason of such controversy is the context in which these proteins function, e.g. a protein which is considered to be proliferation suppressor might

drive cell invasiveness or motility (Moh & Shen, 2009; Tang et al., 2018; Mulkearns-Hu-bert et al., 2020).

In present work we have analyzed the total abundance and the abundance of superficially localized representative proteins of actin-con-nected cell-cell and cell-ECM contacts in two ovarian carcinoma cell lines with different aggressiveness. In case of adherens junctions we have showed that E-cadherin was absent in

both studied cell lines. Concerning N-cadherin the studied cell cultures showed heterogeneity as a part of the cells did not express N-cadherin and the rest of the population expressed N-cad-herin at a relatively low level. Loss of E-cadherin, which is widely recognized as an epithelial marker, triggers the Epithelial-to-Mesen-chymal Transition (EMT) of cells and contributes to the development of an aggressive phe-notype. During EMT, there is a switch from the synthesis of E-cadherin to the synthesis of N-cadherin, which is a mesenchymal marker and is involved in intracellular signaling promoting invasion and metastasis (Gravdal et al., 2007; Loh et al., 2019). It is worth noting that the cells of the studied lines are characterized by the formation of loose three-dimensional aggregates in vitro; which is in line with an assumption of their mesenchymal state (Winner et al., 2016; Sokolova et al., 2019; Kutova et al., 2020).

Currently, more and more evidence is accumulating that EMT is not a binary process, but a continual one, implying a huge number of variants of the intermediate states of the cell, when it can express epithelial and mesenchymal markers simultaneously, which contributes to the development of cancer plasticity (Sha et al., 2019). It has been hypothesized and supported by mathematical modeling that such intermediate states can accelerate EMT and exacerbate its consequences (Goetz et al., 2020). The data on the intermediate position occupied by the SKOV-3 and SKOV-3.ip lines vary, because despite the fact that these lines are called intermediate mesenchymal, this can mean both the simultaneous presence of E-cadherin and N-cadherin (Rosso et al., 2017; Teng et al., 2015) or the absence of E-cadherin with presence of N-cadherin (Klymenko et al., 2017), which was observed in our work. Such inconsistency of the data might be due to the high plasticity of tumor cells, which results in subtle differences in the phenotype evoked by different cultivation conditions. It should also be noted that the representation of N-cadherin in our experiments was rather low. Simultaneous loss of adherens junctions' proteins has been described for very poorly differentiated hepatocellular carcinoma, which was accompanied by extremely low pa-

tient survival (Liu et al., 2015) and in exceptional cases of invasive lobular breast cancer characterized by tubular elements formation (Christgen et al., 2020). In addition, the observed difference in the amount of total N-cadherin and superficially localized can be explained by the transition of N-cadherin into a soluble form, which has pro-angiogenic properties and is at high level detected in biological fluids of cancer patients (Derycke et al., 2006 a, b).

According to the obtained data gap junction protein connexin-43 was represented at relatively high level in both studied cell lines yet significantly prevailed in SKOV-3 cell line. Functional connexin-based gap junctions being properly organized provide Gap Junction Intercellular Communication (GJIC), which in turn maintains coordinated work of the cells within the tissue. Thus, connexins are considered to hinder tumor progression via GJIC (Krutov-skikh et al., 2002, Zefferino et al., 2019). Another plausible mechanism of connexin-medi-ated tumor suppression is the participation of connexins in intracellular signaling interfering with proliferative and invasive signals (Aasen et al., 2019; Mulkearns-Hubert et al., 2020). Concerning tumor suppressing potencies of connexin-43 it is reported that it can inhibit proliferation by affecting the Wingless-Integrated (Wnt)/ß-catenin pathway (Shima et al., 2006), decreasing the activity of proto-oncogene tyro-sine-protein kinase Src (acronym of 'sarcoma') or epidermal growth factor (EGF) (HerreroGonzalez et al., 2010; Qui et al., 2016) or by triggering apoptosis via binding to pro-apop-totic Bcl2-associated X protein (Bax) (Sun et al., 2012). Of note, our data indicate that the level of superficially localized connexin-43 did not exceed 20% of total abundance. It is possibly due to the disrupted connexin-43 trafficking to plasma membrane. Translocation to the cytoplasm was reported for connexin-43 and may be mediated by Wnt signaling pathway (Hou et al., 2019); it was also shown for other connexins (Krutovskikh et al., 1994; Thiagarajan et al., 2018). Aberrant localization of connexins which was observed in our study is reported to be associated with the triggering of EMT (Crespin et al., 2016; Kotini et al., 2018).

The most abundant of all observed proteins was integrin beta-1. The majority of research testifies that high levels of integrin beta-1 are represented in tumor cells compared to normal epithelium (Min et al., 2020). Up-regulated integrin beta-1 is associated with increased cancer cell survival, proliferation, migration and colony formation in vitro (Pardo et al., 2002; Chang et al., 2019) and with advanced stage of the tumor and lower life expectancy in patients (Lin et al., 2014; Lawson et al., 2010). Our data indicate that superficial localization of integrin beta-1 is low compared to total protein. It is interesting to note that higher total abundance of integrin beta-1 was detected in SKOV-3 cell line, yet superficially localized integrin beta-1 was more represented in SKOV-3.ip cell line. Superficially located integrins being activated are shown to promote tumors towards increased malignancy mainly via focal adhesion kinase (FAK) signaling axis (Yang et al., 2014; Zhang & Zou, 2015; Xu et al., 2017).

The profile of adhesion proteins expression appears to be more adequate means of tumor phenotype identification compared to assessment of individual proteins due to their complex crosstalk and reciprocal regulation. For example, it was shown that diapedesis-mediated tumor invasiveness was registered in tumors with simultaneous loss of E-cadherin and high expression of connexin-43 (Mol et al., 2007) or integrin beta-1 (Shu et al., 2013, Symowicz et al., 2007). It should be noted that protein expression and their representation on the cell surface is a dynamic process which is highly dependent on the interplay of microenvironmental context and ongoing physiological processes. For example, along with high expression of connexin-43, E-cadherin abundance can contin-

uously change over time during different phases of an invasion process (Mol et al., 2007; Xu et al., 2008).

Conclusions

In this work we have analyzed total content and superficially localized pool of adhesion proteins in two ovarian carcinoma cell lines. The absolute loss of epithelial marker E-cadherin and overall mislocalization of the rest analyzed proteins and hence their impaired adhesive function, allow us to speculate about mes-enchymal characteristics of the studied cell lines. At the same time, we have detected heterogeneity of the studied cell cultures in terms of N-cadherin and integrin beta-1 abundance. This is in line with the inconsistency of published data on SKOV-3 and SKOV-3.ip pheno-type and thus may be a strong evidence of cancer cells plasticity even in controllable conditions of laboratory maintenance. According to the obtained data SKOV-3.ip cell line possessed lower overall adhesion proteins abundance which is in line with a mainstream notion that loss of cellular adhesion underlies tumor aggressiveness. At the same time higher representation of superficially localized integrin beta-1 might be an additional sign of SKOV-3.ip superlative aggressiveness. Our data may be useful to expand an understanding of the nature of the exceeding aggressiveness of SKOV-3.ip cell line in comparison to SKOV-3.

Acknowledgments

This research was financially supported by the Russian Science Foundation (project No. 19-74-20168). O.M.K. thanks the Russian Foundation for Basic Research for personal financial support (project No. 19-34-90159).

References

AASEN T., LEITHE E., GRAHAM S.V., KAMERITSCH P., MAYAN M.D., MESNIL M., POGODA K. & TABERNERO A. (2019): Connexins in cancer: bridging the gap to the clinic. Oncogene, 38, 44294451.

ALAGA K.C., CRAWFORD M., DAGNINO L., & LAIRD D.W. (2017): Aberrant Cx43 Expression and

Mislocalization in Metastatic Human Melanomas. Journal of Cancer, 8(7), 1123-1128. AMBROSI C., REN C., SPAGNOL G., CAVIN G., CONE A., GRINTSEVICH E E., SOSINSKY G.E. & SORGEN P.L. (2016): Connexin43 Forms Supramolecular Complexes through Non-Overlapping Binding Sites for Drebrin, Tubulin, and ZO-1. PloS one, 11(6), e0157073.

BONACQUISTI E E. & NGUYEN J. (2019): Connexin 43 (Cx43) in cancer: Implications for therapeutic approaches via gap junctions. Cancer Letters, 442, 439-444.

BRÜCHER B.L. & JAMALL I.S. (2014): Cell-cell communication in the tumor microenvironment, carcinogenesis, and anticancer treatment. Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology, 34(2), 213-243.

CAMPBELL H.K., MAIERS J.L. & DEMALI K.A. (2017): Interplay between tight junctions & adherens junctions. Experimental cell research, 358(1), 39-44.

CHANG Y.N., LIANG Y., GU W., WANG J., QIN Y., CHEN K., LI J., BAI X., ZHANG J. & XING G. (2019): Microfluidic Analysis for Separating and Measuring the Deformability of Cancer Cell Subpopulations. ACS Omega, 4(5), 8318-8323.

CHRISTGEN M., BARTELS S., VAN LUTTIKHUIZEN J.L., BUBLITZ J., RIEGER L.U., CHRISTGEN H., STARK H., SANDER B., LEHMANN U., STEINEMANN D., DERKSEN P. & KREIPE H. (2020). E-cadherin to P-cadherin switching in lobular breast cancer with tubular elements. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc, 33(12), 2483-2498.

CRESPIN S., FROMONT G., WAGER M., LEVILLAIN P., CRONIER L., MONVOISIN A., DEFAMIE N., & MESNIL M. (2016): Expression of a gap junction protein, connexin43, in a large panel of human gliomas: new insights. Cancer medicine, 5(8), 1742-1752.

DAR S., CHHINA J., MERT I., CHITALE D., BUEKERS T., KAUR H., GIRI S., MUNKARAH A. & RATTAN R. (2017): Bioenergetic Adaptations in Chemoresistant Ovarian Cancer Cells. Scientific reports, 7(1), 8760.

DERYCKE L., DE WEVER O., STOVE V., VANHOECKE B., DELANGHE J., DEPYPERE H. & BRACKE M. (2006): Soluble N-cadherin in human biological fluids. International journal of cancer, 119(12), 2895-2900.

DERYCKE L., MORBIDELLI L., ZICHE M., DE WEVER O., BRACKE M. & VAN AKEN E. (2006): Soluble N-cadherin fragment promotes angiogenesis. Clinical & experimental metastasis, 23(3-4), 187-201.

DOMINIAK A., CHELSTOWSKA B., OLEJARZ W. & NOWICKA G. (2020): Communication in the Cancer Microenvironment as a Target for Therapeutic Interventions. Cancers, 12(5), 1232.

FARAHANI E., PATRA H.K., JANGAMREDDY J.R., RASHEDI I., KAWALEC M., RAO PARITI R.K., BATAKIS P. & WIECHEC E. (2014): Cell adhesion molecules and their relation to (cancer) cell sternness. Carcinogenesis, 35(4), 747-59.

GALLUZZI L. & THOMAS C. (2020a): Actin Cytoskeleton in Cancer Progression and Metastasis. International Review of Cell and Molecular Biology, 355(Pt A), 254.

GALLUZZI L. & THOMAS C. (2020b): Actin Cytoskeleton in Cancer Progression and Metastasis. International Review of Cell and Molecular Biology, 356(Pt B), 330.

GARROD D., & CHIDGEY M. (2008): Desmosome structure, composition and function. Biochimica et Bi-ophysica Acta, 1778, 572-587.

GOETZ H., MELENDEZ-ALVAREZ J.R., CHEN L. & TIAN X. J. (2020): A plausible accelerating function of intermediate states in cancer metastasis. PLoS computational biology, 16(3), e1007682.

GRAVDAL K., HALVORSEN O.J., HAUKAAS S.A. & AKSLEN L A. (2007): A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clinical cancer research: an official journal of the American Association^ for Cancer Research, 13(23), 7003-7011.

HARRIS T. J. & TEPASS U. (2010): Adherens junctions: from molecules to morphogenesis. Nature reviews. Molecular cell biology, 11(7), 502-514.

HATZFELD M., KEIL R. & MAGIN T.M. (2017): Desmosomes and Intermediate Filaments: Their Consequences for Tissue Mechanics. Cold Spring Harbor perspectives in biology, 9(6), a029157.

HERRERO-GONZALEZ S., GANGOSO E., GIAUME C., NAUS, C.C., MEDINA J.M. & TABERNERO A. (2010): Connexin43 inhibits the oncogenic activity of c-Src in C6 glioma cells. Oncogene, 29, 57125723.

HERVÉ J.C., BOURMEYSTER N., SARROUILHE D. & DUFFY H.S. (2007): Gap junctional complexes: from partners to functions. Progress in biophysics and molecular biology, 94(1-2), 29-65.

HOU X., KHAN M.R.A., TURMAINE M., THRASIVOULOU C., BECKER D.L. & AHMED A. (2019): Wnt signaling regulates cytosolic translocation of connexin 43. American Journal of Physiology-Regulatory, 317, R248-R261.

HUNG M.C., ZHANG X., YAN D.H., ZHANG H.Z., HE G.P., ZHANG T.Q. & SHI D.R. (1992): Aberrant expression of the c-erbB-2/neu protooncogene in ovarian cancer. Cancer letters, 61(2), 95-103.

HYNES R.O. (1992): Integrins: versatility, modulation, and signaling in cell adhesion. Cell, 69(1), 11-25.

KHAN Z.S. & HUSSAIN F. (2020): Shear Stress Increases V-H + -ATPase and Acidic Vesicle Number Density, and p-mTORC2 Activation in Prostate Cancer Cells. Cellular and molecular bioengineering, 13(6), 591-604.

KLYMENKO Y., JOHNSON J., BOS B., LOMBARD R., CAMPBELL L., LOUGHRAN E. & STACK M.S. (2017): Heterogeneous Cadherin Expression and Multicellular Aggregate Dynamics in Ovarian Cancer Dissemination. Neoplasia (New York, N.Y.), 19(7), 549-563.

KOTINI M., BARRIGA E.H., LESLIE J., GENTZEL M., RAUSCHENBERGER V., SCHAMBONY A. & MAYOR R. (2018): Gap junction protein connexin-43 is a direct transcriptional regulator ofN-cadherin in vivo. Nature communications, 9(1), 3846.

KRUTOVSKIKH V., PICCOLI C. & YAMASAKI H. (2002): Gap junction intercellular communication propagates cell death in cancerous cells. Oncogene, 21, 1989-1999.

KUTOVA O.M., SENCHA L.M., POSPELOV A.D., DOBRYNINA O.E., BRILKINA A.A., CHERKASOVA E.I. & BALALAEVA I.V. (2020). Comparative Analysis of Cell-Cell Contact Abundance in Ovarian Carcinoma Cells Cultured in Two- and Three-Dimensional In Vitro Models. Biology, 9(12), 446.

LAWSON M.H., CUMMINGS N.M., RASSL D.M., VOWLER S.L., WICKENS M., HOWAT W.J., BREN-TON J.D., MURPHY G. & RINTOUL R.C. (2010): Bcl-2 and ß1-integrin predict survival in a tissue microarray of small cell lung cancer. British journal of cancer, 103(11), 1710-1715.

LIN H C., WU C.L., CHEN Y.L., HUANG J.S., WONG T.Y. & YUAN K. (2014): High-level ß1-integrin expression in a subpopulation of highly tumorigenic oral cancer cells. Clinical Oral Investigations, 18(4), 1277-1284.

LIU Y.A., LIANG B.Y., GUAN Y., YOU J., ZHU L., CHEN X.P. & HUANG Z.Y. (2015): Loss of N-cadherin is associated with loss of E-cadherin expression and poor outcomes of liver resection in hepatocellular carcinoma. The Journal of surgical research, 194(1), 167-176.

LOH C.Y., CHAI J.Y., TANG T.F., WONG W.F., SETHI G., SHANMUGAM M.K., CHONG P.P. & LOOI C.Y. (2019): The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells, 8(10), 1118.

MIN W., ZOU C., DAI D., ZUO Q., CHEN C., XU J., LI Y. & YUE Z. (2020): Integrin Beta 1 Promotes Glioma Cell Proliferation by Negatively Regulating the Notch Pathway. Journal of Oncology, 15, 8297017.

MOH M.C. & SHEN S. (2009): The roles of cell adhesion molecules in tumor suppression and cell migration: a new paradox. Cell adhesion & migration, 3(4), 334-336.

MOL A.J., GELDOF A.A., MEIJER G.A., VAN DER POEL H.G. & VAN MOORSELAAR R.J. (2007): New experimental markers for early detection of high-risk prostate cancer: role of cell-cell adhesion and cell migration. Journal of cancer research and clinical oncology, 133(10), 687-695.

MOU Y.Y., ZHAO G.Q., LIN J.Y., ZHAO J., LIN H., HU L.T., XU Q., WANG Q. & SUN W.R. (2011): Expression of connexin 43 and E-cadherin in choroidal melanoma. International journal of ophthalmology, 4(2), 156-161.

MULKEARNS-HUBERT E E., REIZES O. & LATHIA J.D. (2020): Connexins in Cancer: Jekyll or Hyde? Biomolecules, 10(12), 1654.

NEOPHYTOU C M., PANAGI M., STYLIANOPOULOS T. & PAPAGEORGIS P. (2021): The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities. Cancers, 13(9), 2053.

NIELSEN M.S., AXELSEN L.N., SORGEN PL., VERMA V., DELMAR M. & HOLSTEIN-RATHLOU N.H. (2012): Gap junctions. Comprehensive Physiology, 2, 1981-2035.

NISHIMURA M. (2003): Suppression of gap junctional intercellular communication via 5' CpG island meth-ylation in promoter region of E-cadherin gene in endometrial cancer cells. Carcinogenesis, 24(10), 1615-1623.

PARDO O.E., ARCARO A., SALERNO G., RAGUZ S., DOWNWARD J. & SECKL M.J. (2002): Fibroblast growth factor-2 induces translational regulation of Bcl-XL and Bcl-2 via a MEK-dependent pathway: correlation with resistance to etoposide-induced apoptosis. Journal of Biological Chemistry, 277, 12040-12046.

QIU X., CHENG J.C., KLAUSEN C., CHANG H.M., FAN Q. & LEUNG PC. (2016): EGF-Induced Con-nexin43 Negatively Regulates Cell Proliferation in Human Ovarian Cancer. Journal of Cellular Physiology, 231, 111-119.

ROSSO M., MAJEM B., DEVIS L., LAPYCKYJ L., BESSO M.J., LLAURADÓ M., ABASCAL M.F., MATOS M L., LANAU L., CASTELLVÍ J., SÁNCHEZ J.L., PÉREZ BENAVENTE A., GIL-MORENO A., REVENTÓS J., SANTAMARIA MARGALEF A., RIGAU M. & VAZQUEZ-LEVIN M.H. (2017): E-cadherin: A determinant molecule associated with ovarian cancer progression, dissemination and aggressiveness. PloS one, 12(9), e0184439.

SANDQUIST E.J., SOMJI S., DUNLEVY JR., GARRETT S.H., ZHOU X.D., SLUSSER-NORE A. & SENS D.A. (2016): Loss of N-Cadherin Expression in Tumor Transplants Produced From As+3- and Cd+2-Transformed Human Urothelial (UROtsa) Cell Lines. PloS one, 11(5), e0156310.

SERAYA-BAREKET C., WEISZ A., SHINDERMAN-MAMAN E., TEPER-ROTH S., STAMLER D., AR-BIB, N., KADAN Y., FISHMAN A., KIDRON D., EDELSTEIN E., ELLIS M. & ASHUR-FABIAN O. (2020): The identification of nuclear avP3 integrin in ovarian cancer: non-paradigmal localization with cancer promoting actions. Oncogenesis, 9(7), 69.

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

SHA Y., HAENSEL D., GUTIERREZ G., DU H., DAI X. & NIE Q. (2019): Intermediate cell states in epithelial-to-mesenchymal transition. Physical biology, 16(2), 021001.

SHIMA K., MURAMATSU T., ABIKO Y., YAMAOKA Y., SASAKI H. & SHIMONO M. (2006): Connexin 43 transfection in basaloid squamous cell carcinoma cells. Oncology Reports, 16, 285-288.

SHU H., CHEN H., YANG B., CHANG Z., XIONG M. & CHEN W. (2013): Aberrant expression of E-cadherin and integrin p-1 in trophoblasts is associated with malignant gestational trophoblastic diseases. International journal of gynecological cancer: official journal of the International Gynecological Cancer Society, 23(4), 749-754.

SOKOLOVA E., KUTOVA O., GRISHINA A., POSPELOV A., GURYEV E., SCHULGA A., DEYEV S., & BALALAEVA I. (2019). Penetration Efficiency of Antitumor Agents in Ovarian Cancer Spheroids: The Case of Recombinant Targeted Toxin DARPin-LoPE and the Chemotherapy Drug, Doxorubicin. Pharmaceutics, 11(5), 219.

SONG D., YANG D., POWELL C.A. & WANG X. (2019): Cell-cell communication: old mystery and new opportunity. Cell biology and toxicology, 35(2), 89-93.

SONNENSCHEIN C. & SOTO A.M. (2020): Over a century of cancer research: Inconvenient truths and promising leads. PLoS biology, 18(4), e3000670.

SUN Q., KANEHIRA K. & TANIGUCHI A. (2018): PEGylated TiO2 nanoparticles mediated inhibition of cell migration via integrin beta 1. Science and technology of advanced materials, 19(1), 271-281.

SUN Y., ZHAO X., YAO Y., QI X., YUAN Y. & HU Y. (2012): Connexin 43 interacts with Bax to regulate apoptosis of pancreatic cancer through a gap junction-independent pathway. International journal of oncology, 41(3), 941-948.

SYMOWICZ J., ADLEY B P., GLEASON K.J., JOHNSON J.J., GHOSH S., FISHMAN D.A., HUDSON L.G. & STACK M.S. (2007): Engagement of collagen-binding integrins promotes matrix metallopro-teinase-9-dependent E-cadherin ectodomain shedding in ovarian carcinoma cells. Cancer research, 67(5), 2030-2039.

TAKADA Y., YE X. & SIMON S. (2007): The integrins. Genome biology, 8(5), 215.

TANG M., YUE P., IP P.P., HUANG R.L., LAI H.C., CHEUNG A., TSE K.Y., NGAN H. & WONG A. (2018): Soluble E-cadherin promotes tumor angiogenesis and localizes to exosome surface. Nature communications, 9(1), 2270.

TENG L., PENG S., GUO H., LIANG H., XU Z., SU Y. & GAO L. (2015). Conditioned media from human ovarian cancer endothelial progenitor cells induces ovarian cancer cell migration by activating epithelial-to-mesenchymal transition. Cancer gene therapy, 22(11), 518-523.

TSUKITA S., FURUSE M. & ITOH M. (2001): Multifunctional strands in tight junctions. Nature Reviews Molecular Cell Biology, 2, 285-293.

VICENTE-MANZANARES M., CHOI C.K. & HORWITZ A.R. (2009): Integrins in cell migration--the actin connection. Journal of cell science, 122(Pt 2), 199-206.

WANG X. & LI S. (2014): Protein mislocalization: mechanisms, functions and clinical applications in cancer. Biochimica et biophysica acta, 1846(1), 13-25.

WELLS A., YATES C. & SHEPARD C.R. (2008): E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomas. Clinical & experimental metastasis, 25(6), 621-628.

WINNER K.K., STEINKAMP MP., LEE R.J., SWAT M., MULLER C.Y., MOSES M.E., JIANG Y. & WILSON B.S. (2016): Spatial Modeling of Drug Delivery Routes for Treatment of Disseminated Ovarian Cancer. Cancer research, 76(6), 1320-1334.

XU H.T., LI Q.C., ZHANG Y.X., ZHAO Y., LIU Y., YANG Z.Q. & WANG E. H. (2008): Connexin 43 recruits E-cadherin expression and inhibits the malignant behaviour of lung cancer cells. Folia histo-chemica et cytobiologica, 46(3), 315-321.

XU J., ZHENG T., HONG W., YE H., HU C. & ZHENG Y. (2018): Mechanism for the Decision of Ovarian Surface Epithelial Stem Cells to Undergo Neo-Oogenesis or Ovarian Tumorigenesis. Cell Physiol Bio-chem, 50(1), 214-232.

YANG Z., ZHOU X., LIU Y., GONG C., WEI X., ZHANG T., MA D. & GAO Q. (2014). Activation of integrin pi mediates the increased malignant potential of ovarian cancer cells exerted by inflammatory cytokines. Anti-cancer agents in medicinal chemistry, 14(7), 955-962.

YU D., WOLF J.K., SCANLON M., PRICE J.E. & HUNG M.C. (1993): Enhanced c-erbB-2/neu expression in human ovarian cancer cells correlates with more severe malignancy that can be suppressed by E1A. Cancer Research, 53(4), 891-898.

ZEFFERINO R., PICCOLI C., GIOIA S. D., CAPITANIO N. & CONESE M. (2019): Gap Junction Intercellular Communication in the Carcinogenesis Hallmarks: Is This a Phenomenon or Epiphenomenon? Cells, 8(8), 896.

ZEMLJIC -HARPF A.E., GODOY J.C., PLATOSHYN O., ASFAW E.K., BUSIJA A.R., DOMENIGHETTI A.A. & ROSS R.S. (2014): Vinculin directly binds zonula occludens-1 and is essential for stabilizing connexin-43-containing gap junctions in cardiac myocytes. Journal of cell science, 127(Pt 5), 11041116.

ZHANG L. & ZOU W. (2015). Inhibition of integrin pi decreases the malignancy of ovarian cancer cells and potentiates anticancer therapy via the FAK/STAT1 signaling pathway. Molecular medicine reports, 12(6), 7869-7876.

i Надоели баннеры? Вы всегда можете отключить рекламу.