Molecular and Cellular Mechanisms of Cancers: Breast Cancer

A special issue of Cells (ISSN 2073-4409).

Deadline for manuscript submissions: closed (31 January 2020) | Viewed by 80384

Special Issue Editor

Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
Interests: cell growth; cell survival; cell death; apoptosis; cancer cells; developmental therapeutics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Breast cancer is a molecularly diverse disease which involves complex processes that result in initiation, progression, and metastasis. Traditionally, breast cancers are categorized into three major subgroups. These include the luminal subtype with hormone receptor positivity (HR+), the oncogene HER2 subtype (HER2+), and the triple-negative, basal subtype that lacks hormone receptors and HER2. Lately, additional subtypes within these broad categories have emerged. Together with the discovery of additional driver genes and their mutant variants, they underscore complex molecular mechanisms and pathways of breast carcinogenesis. There is an increasing realization that many oncogenes that control carcinogenesis exert profound effects on malignant transformation and distant metastases. Moreover, mutations in driver oncogenes, such as gene amplification and mutations and concomitant dysregulation of apoptotic pathways, often contribute to the emergence of resistant phenotypes that consequently affect the efficacy of current therapies and patient survival. Therefore, the targeting of such drivers and their downstream, oncogenic signaling nodes have been rationally pursued for many cancers, including breast cancers. For example, endocrine therapies or HER2 targeting have provided clinical benefits for treating luminal or HER2-positive subtypes of breast cancers, respectively. Additional, molecular mechanism-based targeted therapeutics have recently emerged. These include inhibitors of DNA repair PARP protein in a subset of a BRCA-mutant basal breast cancer subtype, or CDK4/6 inhibitors for HR+ HER2- advanced breast cancers. Although a novel class of immune check-point inhibitors have been approved for clinical use in many cancers, a number of clinical trials are currently underway to assess the potential therapeutic use of current immune check-point inhibitors as monotherapies or in combination with other targeted therapies in breast cancers. The primary focus of this topic will be cell signaling in cancer growth and survival, and current and novel strategies to target oncogenic drivers and their downstream signaling nodes. In addition, the reviews and research articles will explore the mechanisms of carcinogenesis and the ways in which tumor cells modulate these processes to optimize their survival and examine current clinical modalities aimed at exploiting these oncogenic targets to selectively inhibit tumor cells.

Prof. Arun K. Rishi
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • growth and survival
  • cancer
  • receptor signaling
  • signal transduction nodes: MAPKs, NF-kB
  • oncogenes
  • tumor metabolism
  • immune check-point signaling
  • epithelial-to-mesenchymal transition

Published Papers (12 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

16 pages, 1410 KiB  
Article
Vimentin Promotes the Aggressiveness of Triple Negative Breast Cancer Cells Surviving Chemotherapeutic Treatment
by Marie Winter, Samuel Meignan, Pamela Völkel, Pierre-Olivier Angrand, Valérie Chopin, Nadège Bidan, Robert-Alain Toillon, Eric Adriaenssens, Chann Lagadec and Xuefen Le Bourhis
Cells 2021, 10(6), 1504; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10061504 - 15 Jun 2021
Cited by 12 | Viewed by 3554
Abstract
Tremendous data have been accumulated in the effort to understand chemoresistance of triple negative breast cancer (TNBC). However, modifications in cancer cells surviving combined and sequential treatment still remain poorly described. In order to mimic clinical neoadjuvant treatment, we first treated MDA-MB-231 and [...] Read more.
Tremendous data have been accumulated in the effort to understand chemoresistance of triple negative breast cancer (TNBC). However, modifications in cancer cells surviving combined and sequential treatment still remain poorly described. In order to mimic clinical neoadjuvant treatment, we first treated MDA-MB-231 and SUM159-PT TNBC cell lines with epirubicin and cyclophosphamide for 2 days, and then with paclitaxel for another 2 days. After 4 days of recovery, persistent cells surviving the treatment were characterized at both cellular and molecular level. Persistent cells exhibited increased growth and were more invasive in vitro and in zebrafish model. Persistent cells were enriched for vimentinhigh sub-population, vimentin knockdown using siRNA approach decreased the invasive and sphere forming capacities as well as Akt phosphorylation in persistent cells, indicating that vimentin is involved in chemotherapeutic treatment-induced enhancement of TNBC aggressiveness. Interestingly, ectopic vimentin overexpression in native cells increased cell invasion and sphere formation as well as Akt phosphorylation. Furthermore, vimentin overexpression alone rendered the native cells resistant to the drugs, while vimentin knockdown rendered them more sensitive to the drugs. Together, our data suggest that vimentin could be considered as a new targetable player in the ever-elusive status of drug resistance and recurrence of TNBC. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

17 pages, 4418 KiB  
Article
Extracellular Acidosis Promotes Metastatic Potency via Decrease of the BMAL1 Circadian Clock Gene in Breast Cancer
by Yong-Jin Kwon, Eun-Bi Seo, Sun-Ho Kwon, Song-Hee Lee, Seul-Ki Kim, Sang Ki Park, Kyungjin Kim, SaeGwang Park, In-Chul Park, Jong-Wan Park and Sang-Kyu Ye
Cells 2020, 9(4), 989; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9040989 - 16 Apr 2020
Cited by 19 | Viewed by 3557
Abstract
Circadian oscillation is an essential process that influences many physiological and biological mechanisms and a decrease of circadian genes is associated with many diseases such as cancer. Despite many efforts to identify the detailed mechanism for decreasing circadian genes and recovering reduced circadian [...] Read more.
Circadian oscillation is an essential process that influences many physiological and biological mechanisms and a decrease of circadian genes is associated with many diseases such as cancer. Despite many efforts to identify the detailed mechanism for decreasing circadian genes and recovering reduced circadian genes in cancer, it is still largely unknown. We found that BMAL1 was reduced in tumor hypoxia-induced acidosis, and recovered by selectively targeting acidic pH in breast cancer cell lines. Surprisingly, BMAL1 was reduced by decrease of protein stability as well as inhibition of transcription under acidosis. In addition, melatonin significantly prevented acidosis-mediated decrease of BMAL1 by inhibiting lactate dehydrogenase-A during hypoxia. Remarkably, acidosis-mediated metastasis was significantly alleviated by BMAL1 overexpression in breast cancer cells. We therefore suggest that tumor hypoxia-induced acidosis promotes metastatic potency by decreasing BMAL1, and that tumor acidosis could be a target for preventing breast cancer metastasis by sustaining BMAL1. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Graphical abstract

12 pages, 5776 KiB  
Article
Basal-Type Breast Cancer Stem Cells Over-Express Chromosomal Passenger Complex Proteins
by Angela Schwarz-Cruz y Celis, Gisela Ceballos-Cancino, Karla Vazquez-Santillan, Magali Espinosa, Cecilia Zampedri, Ivan Bahena, Victor Ruiz, Vilma Maldonado and Jorge Melendez-Zajgla
Cells 2020, 9(3), 709; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9030709 - 13 Mar 2020
Cited by 9 | Viewed by 3221
Abstract
(1) Aim: In the present paper we analyzed the transcriptome of CSCs (Cancer Stem Cells), in order to find defining molecular processes of breast cancer. (2) Methods: We performed RNA-Seq from CSCs isolated from the basal cell line MDA-MB-468. Enriched processes and networks [...] Read more.
(1) Aim: In the present paper we analyzed the transcriptome of CSCs (Cancer Stem Cells), in order to find defining molecular processes of breast cancer. (2) Methods: We performed RNA-Seq from CSCs isolated from the basal cell line MDA-MB-468. Enriched processes and networks were studied using the IPA (Ingenuity Pathway Analysis) tool. Validation was performed with qRT-PCR and the analysis of relevant genes was evaluated by overexpression, flow cytometry and in vivo zebrafish studies. Finally, the clinical relevance of these results was assessed using reported cohorts. (3) Results: We found that CSCs presented marked differences from the non-CSCs, including enrichment in transduction cascades related to stemness, cellular growth, proliferation and apoptosis. Interestingly, CSCs overexpressed a module of co-regulated Chromosomal Passenger Proteins including BIRC5 (survivin), INCENP and AURKB. Overexpression of BIRC5 increased the number of CSCs, as assessed by in vitro and in vivo zebrafish xenotransplant analyses. Analysis of previously published cohorts showed that this co-regulated module was not only overexpressed in basal breast tumors but also associated with relapse-free and overall survival in these patients. (4) Conclusions: These results underline the importance of Cancer Stem Cells in breast cancer progression and point toward the possible use of chromosomal passenger proteins as prognostic factors. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

18 pages, 4790 KiB  
Article
CDK7 Inhibition Is Effective in all the Subtypes of Breast Cancer: Determinants of Response and Synergy with EGFR Inhibition
by Martina S. J. McDermott, Amanda C. Sharko, Jessica Munie, Susannah Kassler, Theresa Melendez, Chang-uk Lim and Eugenia V. Broude
Cells 2020, 9(3), 638; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9030638 - 06 Mar 2020
Cited by 24 | Viewed by 4592
Abstract
CDK7, a transcriptional cyclin-dependent kinase, is emerging as a novel cancer target. Triple-negative breast cancers (TNBC) but not estrogen receptor-positive (ER+) breast cancers have been reported to be uniquely sensitive to the CDK7 inhibitor THZ1 due to the inhibition of a cluster of [...] Read more.
CDK7, a transcriptional cyclin-dependent kinase, is emerging as a novel cancer target. Triple-negative breast cancers (TNBC) but not estrogen receptor-positive (ER+) breast cancers have been reported to be uniquely sensitive to the CDK7 inhibitor THZ1 due to the inhibition of a cluster of TNBC-specific genes. However, bioinformatic analysis indicates that CDK7 RNA expression is associated with negative prognosis in all the major subtypes of breast cancer. To further elucidate the effects of CDK7 inhibition in breast cancer, we profiled a panel of cell lines representing different breast cancer subtypes. THZ1 inhibited cell growth in all subtypes (TNBC, HER2+, ER+, and HER2+/ER+) with no apparent subtype selectivity. THZ1 inhibited CDK7 activity and induced G1 arrest and apoptosis in all the tested cell lines, but THZ1 sensitivity did not correlate with CDK7 inhibition or CDK7 expression levels. THZ1 sensitivity across the cell line panel did not correlate with TNBC-specific gene expression but it was found to correlate with the differential inhibition of three genes: CDKN1B, MYC and transcriptional coregulator CITED2. Response to THZ1 also correlated with basal CITED2 protein expression, a potential marker of CDK7 inhibitor sensitivity. Furthermore, all of the THZ1-inhibited genes examined were inducible by EGF but THZ1 prevented this induction. THZ1 had synergistic or additive effects when combined with the EGFR inhibitor erlotinib, with no outward selectivity for a particular subtype of breast cancer. These results suggest a potential broad utility for CDK7 inhibitors in breast cancer therapy and the potential for combining CDK7 and EGFR inhibitors. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

23 pages, 2640 KiB  
Article
Identification of PARP-1, Histone H1 and SIRT-1 as New Regulators of Breast Cancer-Related Aromatase Promoter I.3/II
by Alexander Kaiser, Thomas Krüger, Gabriele Eiselt, Joachim Bechler, Olaf Kniemeyer, Otmar Huber and Martin Schmidt
Cells 2020, 9(2), 427; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9020427 - 12 Feb 2020
Cited by 10 | Viewed by 3543
Abstract
Paracrine interactions between malignant estrogen receptor positive (ER+) breast cancer cells and breast adipose fibroblasts (BAFs) stimulate estrogen biosynthesis by aromatase in BAFs. In breast cancer, mainly the cAMP-responsive promoter I.3/II-region mediates excessive aromatase expression. A rare single nucleotide variant (SNV) [...] Read more.
Paracrine interactions between malignant estrogen receptor positive (ER+) breast cancer cells and breast adipose fibroblasts (BAFs) stimulate estrogen biosynthesis by aromatase in BAFs. In breast cancer, mainly the cAMP-responsive promoter I.3/II-region mediates excessive aromatase expression. A rare single nucleotide variant (SNV) in this promoter region, which caused 70% reduction in promoter activity, was utilized for the identification of novel regulators of aromatase expression. To this end, normal and mutant promoter activities were measured in luciferase reporter gene assays. DNA-binding proteins were captured by DNA-affinity and identified by mass spectrometry. The DNA binding of proteins was analyzed using electrophoretic mobility shift assays, immunoprecipitation-based in vitro binding assays and by chromatin immunoprecipitation in BAFs in vivo. Protein expression and parylation were analyzed by western blotting. Aromatase activities and RNA-expression were measured in BAFs. Functional consequences of poly (ADP-ribose) polymerase-1 (PARP-1) knock-out, rescue or overexpression, respectively, were analyzed in murine embryonic fibroblasts (MEFs) and the 3T3-L1 cell model. In summary, PARP-1 and histone H1 (H1) were identified as critical regulators of aromatase expression. PARP-1-binding to the SNV-region was crucial for aromatase promoter activation. PARP-1 parylated H1 and competed with H1 for DNA-binding, thereby inhibiting its gene silencing action. In MEFs (PARP-1 knock-out and wild-type) and BAFs, PARP-1-mediated induction of the aromatase promoter showed bi-phasic dose responses in overexpression and inhibitor experiments, respectively. The HDAC-inhibitors butyrate, panobinostat and selisistat enhanced promoter I.3/II-mediated gene expression dependent on PARP-1-activity. Forskolin stimulation of BAFs increased promoter I.3/II-occupancy by PARP-1, whereas SIRT-1 competed with PARP-1 for DNA binding but independently activated the promoter I.3/II. Consistently, the inhibition of both PARP-1 and SIRT-1 increased the NAD+/NADH-ratio in BAFs. This suggests that cellular NAD+/NADH ratios control the complex interactions of PARP-1, H1 and SIRT-1 and regulate the interplay of parylation and acetylation/de-acetylation events with low NAD+/NADH ratios (reverse Warburg effect), promoting PARP-1 activation and estrogen synthesis in BAFs. Therefore, PARP-1 inhibitors could be useful in the treatment of estrogen-dependent breast cancers. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

17 pages, 4760 KiB  
Article
Tannic Acid Promotes TRAIL-Induced Extrinsic Apoptosis by Regulating Mitochondrial ROS in Human Embryonic Carcinoma Cells
by Nipin Sp, Dong Young Kang, Eun Seong Jo, Alexis Rugamba, Wan Seop Kim, Yeong-Min Park, Dae-Yong Hwang, Ji-Seung Yoo, Qing Liu, Kyoung-Jin Jang and Young Mok Yang
Cells 2020, 9(2), 282; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9020282 - 23 Jan 2020
Cited by 34 | Viewed by 4789
Abstract
Human embryonic carcinoma (EC; NCCIT) cells have self-renewal ability and pluripotency. Cancer stem cell markers are highly expressed in NCCIT cells, imparting them with the pluripotent nature to differentiate into other cancer types, including breast cancer. As one of the main cancer stem [...] Read more.
Human embryonic carcinoma (EC; NCCIT) cells have self-renewal ability and pluripotency. Cancer stem cell markers are highly expressed in NCCIT cells, imparting them with the pluripotent nature to differentiate into other cancer types, including breast cancer. As one of the main cancer stem cell pathways, Wnt/β-catenin is also overexpressed in NCCIT cells. Thus, inhibition of these pathways defines the ability of a drug to target cancer stem cells. Tannic acid (TA) is a natural polyphenol present in foods, fruits, and vegetables that has anti-cancer activity. Through Western blotting and PCR, we demonstrate that TA inhibits cancer stem cell markers and the Wnt/β-catenin signaling pathway in NCCIT cells and through a fluorescence-activated cell sorting analysis we demonstrated that TA induces sub-G1 cell cycle arrest and apoptosis. The mechanism underlying this is the induction of mitochondrial reactive oxygen species (ROS) (mROS), which then induce the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated extrinsic apoptosis pathway instead of intrinsic mitochondrial apoptosis pathway. Moreover, ribonucleic acid sequencing data with TA in NCCIT cells show an elevation in TRAIL-induced extrinsic apoptosis, which we confirm by Western blotting and real-time PCR. The induction of human TRAIL also proves that TA can induce extrinsic apoptosis in NCCIT cells by regulating mROS. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

14 pages, 2741 KiB  
Article
Glucose Transporter 3 Is Essential for the Survival of Breast Cancer Cells in the Brain
by Min-Hsun Kuo, Wen-Wei Chang, Bi-Wen Yeh, Yeh-Shiu Chu, Yueh-Chun Lee and Hsueh-Te Lee
Cells 2019, 8(12), 1568; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8121568 - 04 Dec 2019
Cited by 26 | Viewed by 3782
Abstract
Breast cancer brain metastasis commonly occurs in one-fourth of breast cancer patients and is associated with poor prognosis. Abnormal glucose metabolism is found to promote cancer metastasis. Moreover, the tumor microenvironment is crucial and plays an active role in the metabolic adaptations and [...] Read more.
Breast cancer brain metastasis commonly occurs in one-fourth of breast cancer patients and is associated with poor prognosis. Abnormal glucose metabolism is found to promote cancer metastasis. Moreover, the tumor microenvironment is crucial and plays an active role in the metabolic adaptations and survival of cancer cells. Glucose transporters are overexpressed in cancer cells to increase glucose uptake. The glucose transporter 3 (GLUT3) is a high-affinity glucose transporter that is highly expressed in mammalian neurons. GLUT3 is also overexpressed in several malignant brain tumors. However, the role of GLUT3 in breast cancer brain metastasis remains unknown. The results of the present study demonstrated that GLUT3 is highly overexpressed in brain metastatic breast cancers and mediates glucose metabolic reprogramming. Furthermore, knockdown of cAMP-response element binding protein (CREB) could directly regulate GLUT3 expression in brain metastatic breast cancer cells. Notably, we verified and provided a novel role of GLUT3 in mediating glucose metabolism and assisting breast cancer cells to survive in the brain to promote brain metastasis. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

19 pages, 3417 KiB  
Article
The Peptide ERα17p Is a GPER Inverse Agonist that Exerts Antiproliferative Effects in Breast Cancer Cells
by Rosamaria Lappano, Christophe Mallet, Bruno Rizzuti, Fedora Grande, Giulia Raffaella Galli, Cillian Byrne, Isabelle Broutin, Ludivine Boudieu, Alain Eschalier, Yves Jacquot and Marcello Maggiolini
Cells 2019, 8(6), 590; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8060590 - 14 Jun 2019
Cited by 17 | Viewed by 3760
Abstract
The inhibition of the G protein-coupled estrogen receptor (GPER) offers promising perspectives for the treatment of breast tumors. A peptide corresponding to part of the hinge region/AF2 domain of the human estrogen receptor α (ERα17p, residues 295–311) exerts anti-proliferative effects in various breast [...] Read more.
The inhibition of the G protein-coupled estrogen receptor (GPER) offers promising perspectives for the treatment of breast tumors. A peptide corresponding to part of the hinge region/AF2 domain of the human estrogen receptor α (ERα17p, residues 295–311) exerts anti-proliferative effects in various breast cancer cells including those used as triple negative breast cancer (TNBC) models. As preliminary investigations have evoked a role for the GPER in the mechanism of action of this peptide, we focused our studies on this protein using SkBr3 breast cancer cells, which are ideal for GPER evaluation. ERα17p inhibits cell growth by targeting membrane signaling. Identified as a GPER inverse agonist, it co-localizes with GPER and induces the proteasome-dependent downregulation of GPER. It also decreases the level of pEGFR (phosphorylation of epidermal growth factor receptor), pERK1/2 (phosphorylation of extracellular signal-regulated kinase), and c-fos. ERα17p is rapidly distributed in mice after intra-peritoneal injection and is found primarily in the mammary glands. The N-terminal PLMI motif, which presents analogies with the GPER antagonist PBX1, reproduces the effect of the whole ERα17p. Thus, this motif seems to direct the action of the entire peptide, as highlighted by docking and molecular dynamics studies. Consequently, the tetrapeptide PLMI, which can be claimed as the first peptidic GPER disruptor, could open new avenues for specific GPER modulators. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

Review

Jump to: Research

15 pages, 2202 KiB  
Review
Role of Secreted Frizzled-Related Protein 1 in Early Mammary Gland Tumorigenesis and Its Regulation in Breast Microenvironment
by Alisson Clemenceau, Caroline Diorio and Francine Durocher
Cells 2020, 9(1), 208; https://0-doi-org.brum.beds.ac.uk/10.3390/cells9010208 - 14 Jan 2020
Cited by 11 | Viewed by 3867
Abstract
In mice, the lack of secreted frizzled-related protein 1 (SFRP1) is responsible for mammogenesis and hyperplasia, while, in bovines, its overexpression is associated with post-lactational mammary gland involution. Interestingly, there are no reports dealing with the role of SFRP1 in female [...] Read more.
In mice, the lack of secreted frizzled-related protein 1 (SFRP1) is responsible for mammogenesis and hyperplasia, while, in bovines, its overexpression is associated with post-lactational mammary gland involution. Interestingly, there are no reports dealing with the role of SFRP1 in female involution. However, SFRP1 dysregulation is largely associated with human tumorigenesis in the literature. Indeed, the lack of SFRP1 is associated with both tumor development and patient prognosis. Considering the increased risk of breast tumor development associated with incomplete mammary gland involution, it is crucial to demystify the “grey zone” between physiological age-related involution and tumorigenesis. In this review, we explore the functions of SFRP1 involved in the breast involution processes to understand the perturbations driven by the disappearance of SFRP1 in mammary tissue. Moreover, we question the presence of recurrent microcalcifications identified by mammography. In bone metastases from prostate primary tumor, overexpression of SFRP1 results in an osteolytic response of the tumor cells. Hence, we explore the hypothesis of an osteoblastic differentiation of mammary cells induced by the lack of SFRP1 during lobular involution, resulting in a new accumulation of hydroxyapatite crystals in the breast tissue. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

33 pages, 2540 KiB  
Review
MicroRNAs Contribute to Breast Cancer Invasiveness
by Ivana Fridrichova and Iveta Zmetakova
Cells 2019, 8(11), 1361; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8111361 - 31 Oct 2019
Cited by 105 | Viewed by 8957
Abstract
Cancer statistics in 2018 highlight an 8.6 million incidence in female cancers, and 4.2 million cancer deaths globally. Moreover, breast cancer is the most frequent malignancy in females and twenty percent of these develop metastasis. This provides only a small chance for successful [...] Read more.
Cancer statistics in 2018 highlight an 8.6 million incidence in female cancers, and 4.2 million cancer deaths globally. Moreover, breast cancer is the most frequent malignancy in females and twenty percent of these develop metastasis. This provides only a small chance for successful therapy, and identification of new molecular markers for the diagnosis and prognostic prediction of metastatic disease and development of innovative therapeutic molecules are therefore urgently required. Differentially expressed microRNAs (miRNAs) in cancers cause multiple changes in the expression of the tumorigenesis-promoting genes which have mostly been investigated in breast cancers. Herein, we summarize recent data on breast cancer-specific miRNA expression profiles and their participation in regulating invasive processes, in association with changes in cytoskeletal structure, cell-cell adhesion junctions, cancer cell-extracellular matrix interactions, tumor microenvironments, epithelial-to-mesenchymal transitions and cancer cell stem abilities. We then focused on the epigenetic regulation of individual miRNAs and their modified interactions with other regulatory genes, and reviewed the function of miRNA isoforms and exosome-mediated miRNA transfer in cancer invasiveness. Although research into miRNA’s function in cancer is still ongoing, results herein contribute to improved metastatic cancer management. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

32 pages, 2359 KiB  
Review
Mechanisms of Chemotherapy Resistance in Triple-Negative Breast Cancer—How We Can Rise to the Challenge
by Milica Nedeljković and Ana Damjanović
Cells 2019, 8(9), 957; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8090957 - 22 Aug 2019
Cited by 427 | Viewed by 26322
Abstract
Triple-negative (TNBC) is the most lethal subtype of breast cancer owing to high heterogeneity, aggressive nature, and lack of treatment options. Chemotherapy remains the standard of care for TNBC treatment, but unfortunately, patients frequently develop resistance. Accordingly, in recent years, tremendous effort has [...] Read more.
Triple-negative (TNBC) is the most lethal subtype of breast cancer owing to high heterogeneity, aggressive nature, and lack of treatment options. Chemotherapy remains the standard of care for TNBC treatment, but unfortunately, patients frequently develop resistance. Accordingly, in recent years, tremendous effort has been made into elucidating the mechanisms of TNBC chemoresistance with the goal of identifying new molecular targets. It has become evident that the development of TNBC chemoresistance is multifaceted and based on the elaborate interplay of the tumor microenvironment, drug efflux, cancer stem cells, and bulk tumor cells. Alterations of multiple signaling pathways govern these interactions. Moreover, TNBC’s high heterogeneity, highlighted in the existence of several molecular signatures, presents a significant obstacle to successful treatment. In the present, in-depth review, we explore the contribution of key mechanisms to TNBC chemoresistance as well as emerging strategies to overcome them. We discuss novel anti-tumor agents that target the components of these mechanisms and pay special attention to their current clinical development while emphasizing the challenges still ahead of successful TNBC management. The evidence presented in this review outlines the role of crucial pathways in TNBC survival following chemotherapy treatment and highlights the importance of using combinatorial drug strategies and incorporating biomarkers in clinical studies. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Graphical abstract

19 pages, 719 KiB  
Review
The Effects of Adipocytes on the Regulation of Breast Cancer in the Tumor Microenvironment: An Update
by Dinh-Toi Chu, Thuy Nguyen Thi Phuong, Nguyen Le Bao Tien, Dang-Khoa Tran, Tran-Thuy Nguyen, Vo Van Thanh, Thuy Luu Quang, Le Bui Minh, Van Huy Pham, Vo Truong Nhu Ngoc, Kushi Kushekhar and Thien Chu-Dinh
Cells 2019, 8(8), 857; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8080857 - 08 Aug 2019
Cited by 65 | Viewed by 8435
Abstract
Obesity is a global pandemic and it is well evident that obesity is associated with the development of many disorders including many cancer types. Breast cancer is one of that associated with a high mortality rate. Adipocytes, a major cellular component in adipose [...] Read more.
Obesity is a global pandemic and it is well evident that obesity is associated with the development of many disorders including many cancer types. Breast cancer is one of that associated with a high mortality rate. Adipocytes, a major cellular component in adipose tissue, are dysfunctional during obesity and also known to promote breast cancer development both in vitro and in vivo. Dysfunctional adipocytes can release metabolic substrates, adipokines, and cytokines, which promote proliferation, progression, invasion, and migration of breast cancer cells. The secretion of adipocytes can alter gene expression profile, induce inflammation and hypoxia, as well as inhibit apoptosis. It is known that excessive free fatty acids, cholesterol, triglycerides, hormones, leptin, interleukins, and chemokines upregulate breast cancer development. Interestingly, adiponectin is the only adipokine that has anti-tumor properties. Moreover, adipocytes are also related to chemotherapeutic resistance, resulting in the poorer outcome of treatment and advanced stages in breast cancer. Evaluation of the adipocyte secretion levels in the circulation can be useful for prognosis and evaluation of the effectiveness of cancer therapy in the patients. Therefore, understanding about functions of adipocytes as well as obesity in breast cancer may reveal novel targets that support the development of new anti-tumor therapy. In this systemic review, we summarize and update the effects of secreted factors by adipocytes on the regulation of breast cancer in the tumor microenvironment. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Breast Cancer)
Show Figures

Figure 1

Back to TopTop