1
|
Khaledian B, Thibes L, Shimono Y. Adipocyte regulation of cancer stem cells. Cancer Sci 2023; 114:4134-4144. [PMID: 37622414 PMCID: PMC10637066 DOI: 10.1111/cas.15940] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/26/2023] Open
Abstract
Cancer stem cells (CSCs) are a highly tumorigenic subpopulation of the cancer cells within a tumor that drive tumor initiation, progression, and therapy resistance. In general, stem cell niche provides a specific microenvironment in which stem cells are present in an undifferentiated and self-renewable state. CSC niche is a specialized tumor microenvironment for CSCs which provides cues for their maintenance and propagation. However, molecular mechanisms for the CSC-niche interaction remain to be elucidated. We have revealed that adipsin (complement factor D) and its downstream effector hepatocyte growth factor are secreted from adipocytes and enhance the CSC properties in breast cancers in which tumor initiation and progression are constantly associated with the surrounding adipose tissue. Considering that obesity, characterized by excess adipose tissue, is associated with an increased risk of multiple cancers, it is reasonably speculated that adipocyte-CSC interaction is similarly involved in many types of cancers, such as pancreas, colorectal, and ovarian cancers. In this review, various molecular mechanisms by which adipocytes regulate CSCs, including secretion of adipokines, extracellular matrix production, biosynthesis of estrogen, metabolism, and exosome, are discussed. Uncovering the roles of adipocytes in the CSC niche will propose novel strategies to treat cancers, especially those whose progression is linked to obesity.
Collapse
Affiliation(s)
- Behnoush Khaledian
- Department of BiochemistryFujita Health University School of MedicineToyoakeAichiJapan
| | - Lisa Thibes
- Department of BiochemistryFujita Health University School of MedicineToyoakeAichiJapan
| | - Yohei Shimono
- Department of BiochemistryFujita Health University School of MedicineToyoakeAichiJapan
| |
Collapse
|
2
|
Caruso A, Gelsomino L, Panza S, Accattatis FM, Naimo GD, Barone I, Giordano C, Catalano S, Andò S. Leptin: A Heavyweight Player in Obesity-Related Cancers. Biomolecules 2023; 13:1084. [PMID: 37509120 PMCID: PMC10377641 DOI: 10.3390/biom13071084] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Obesity, defined as the abnormal or excessive expansion of white adipose tissue, has reached pandemic proportions and is recognized as an important health concern since it is a common root for several comorbidities, including malignancies. Indeed, the current knowledge of the white adipose tissue, which shifts its role from an energy storage tissue to an important endocrine and metabolic organ, has opened up new avenues for the discovery of obesity's effects on tumor biology. In this review, we will report the epidemiological studies concerning the strong impact of obesity in several types of cancer and describe the mechanisms underlying the heterotypic signals between cancer cell lines and adipocytes, with particular emphasis on inflammation, the insulin/IGF-1 axis, and adipokines. Among the adipokines, we will further describe the in vitro, in vivo, and clinical data concerning the role of leptin, recognized as one of the most important mediators of obesity-associated cancers. In fact, leptin physiologically regulates energy metabolism, appetite, and reproduction, and several studies have also described the role of leptin in affecting cancer development and progression. Finally, we will summarize the newest pharmacological strategies aimed at mitigating the protumorigenic effects of leptin, underlining their mechanisms of action.
Collapse
Affiliation(s)
- Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Felice Maria Accattatis
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| |
Collapse
|
3
|
Ward AV, Matthews SB, Fettig LM, Riley D, Finlay-Schultz J, Paul KV, Jackman M, Kabos P, MacLean PS, Sartorius CA. Estrogens and Progestins Cooperatively Shift Breast Cancer Cell Metabolism. Cancers (Basel) 2022; 14:1776. [PMID: 35406548 PMCID: PMC8996926 DOI: 10.3390/cancers14071776] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 12/15/2022] Open
Abstract
Metabolic reprogramming remains largely understudied in relation to hormones in estrogen receptor (ER) and progesterone receptor (PR) positive breast cancer. In this study, we investigated how estrogens, progestins, or the combination, impact metabolism in three ER and PR positive breast cancer cell lines. We measured metabolites in the treated cells using ultra-performance liquid chromatography coupled with mass spectrometry (UPLC-MS). Top metabolic processes upregulated with each treatment involved glucose metabolism, including Warburg effect/glycolysis, gluconeogenesis, and the pentose phosphate pathway. RNA-sequencing and pathway analysis on two of the cell lines treated with the same hormones, found estrogens target oncogenes, such as MYC and PI3K/AKT/mTOR that control tumor metabolism, while progestins increased genes associated with fatty acid metabolism, and the estrogen/progestin combination additionally increased glycolysis. Phenotypic analysis of cell energy metabolism found that glycolysis was the primary hormonal target, particularly for the progestin and estrogen-progestin combination. Transmission electron microscopy found that, compared to vehicle, estrogens elongated mitochondria, which was reversed by co-treatment with progestins. Progestins promoted lipid storage both alone and in combination with estrogen. These findings highlight the shift in breast cancer cell metabolism to a more glycolytic and lipogenic phenotype in response to combination hormone treatment, which may contribute to a more metabolically adaptive state for cell survival.
Collapse
Affiliation(s)
- Ashley V. Ward
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.V.W.); (S.B.M.); (L.M.F.); (D.R.); (J.F.-S.)
| | - Shawna B. Matthews
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.V.W.); (S.B.M.); (L.M.F.); (D.R.); (J.F.-S.)
| | - Lynsey M. Fettig
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.V.W.); (S.B.M.); (L.M.F.); (D.R.); (J.F.-S.)
| | - Duncan Riley
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.V.W.); (S.B.M.); (L.M.F.); (D.R.); (J.F.-S.)
| | - Jessica Finlay-Schultz
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.V.W.); (S.B.M.); (L.M.F.); (D.R.); (J.F.-S.)
| | - Kiran V. Paul
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.V.P.); (P.K.)
| | - Matthew Jackman
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.J.); (P.S.M.)
| | - Peter Kabos
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.V.P.); (P.K.)
| | - Paul S. MacLean
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (M.J.); (P.S.M.)
| | - Carol A. Sartorius
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (A.V.W.); (S.B.M.); (L.M.F.); (D.R.); (J.F.-S.)
| |
Collapse
|
4
|
Sekar S, Subbamanda Y, Pullaguri N, Sharma A, Sahu C, Kumar R, Bhargava A. Isoform-specific expression of T-type voltage-gated calcium channels and estrogen receptors in breast cancer reveals specific isoforms that may be potential targets. CURRENT RESEARCH IN BIOTECHNOLOGY 2022. [DOI: 10.1016/j.crbiot.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
5
|
Abstract
Prolactin coordinates with the ovarian steroids to orchestrate mammary development and lactation, culminating in nourishment and an increasingly appreciated array of other benefits for neonates. Its central activities in mammary epithelial growth and differentiation suggest that it plays a role(s) in breast cancer, but it has been challenging to identify its contributions, essential for incorporation into prevention and treatment approaches. Large prospective epidemiologic studies have linked higher prolactin exposure to increased risk, particularly for ER+ breast cancer in postmenopausal women. However, it has been more difficult to determine its actions and clinical consequences in established tumors. Here we review experimental data implicating multiple mechanisms by which prolactin may increase the risk of breast cancer. We then consider the evidence for role(s) of prolactin and its downstream signaling cascades in disease progression and treatment responses, and discuss how new approaches are beginning to illuminate the biology behind the seemingly conflicting epidemiologic and experimental studies of prolactin actions across diverse breast cancers.
Collapse
|
6
|
Zheng Y, Karnoub AE. Endocrine regulation of cancer stem cell compartments in breast tumors. Mol Cell Endocrinol 2021; 535:111374. [PMID: 34242715 DOI: 10.1016/j.mce.2021.111374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cancer cells within breast tumors exist within a hierarchy in which only a small and rare subset of cells is able to regenerate growths with the heterogeneity of the original tumor. These highly malignant cancer cells, which behave like stem cells for new cancers and are called "cancer stem cells" or CSCs, have also been shown to possess increased resistance to therapeutics, and represent the root cause underlying therapy failures, persistence of residual disease, and relapse. As >90% of cancer deaths are due to refractory tumors, identification of critical molecular drivers of the CSC-state would reveal vulnerabilities that can be leveraged in designing therapeutics that eradicate advanced disease and improve patient survival outcomes. An expanding and complex body of work has now described the exquisite susceptibility of CSC pools to the regulatory influences of local and systemic hormones. Indeed, breast CSCs express a plethora of hormonal receptors, which funnel hormonal influences over every aspect of breast neoplasia - be it tumor onset, growth, survival, invasion, metastasis, or therapy resistance - via directly impacting CSC behavior. This article is intended to shed light on this active area of investigation by attempting to provide a systematic and comprehensive overview of the available evidence directly linking hormones to breast CSC biology.
Collapse
Affiliation(s)
- Yurong Zheng
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Antoine E Karnoub
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard Stem Cell Institute, Cambridge, MA, 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
7
|
Schuler LA, Murdoch FE. Endogenous and Therapeutic Estrogens: Maestro Conductors of the Microenvironment of ER+ Breast Cancers. Cancers (Basel) 2021; 13:3725. [PMID: 34359625 PMCID: PMC8345134 DOI: 10.3390/cancers13153725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/25/2022] Open
Abstract
Estrogen receptor alpha (ERα) marks heterogeneous breast cancers which display a repertoire of somatic genomic mutations and an immune environment that differs from other breast cancer subtypes. These cancers also exhibit distinct biological behaviors; despite an overall better prognosis than HER2+ or triple negative breast cancers, disseminated dormant cells can lead to disease recurrence decades after the initial diagnosis and treatment. Estrogen is the best studied driver of these cancers, and antagonism or reduction of estrogen activity is the cornerstone of therapeutic approaches. In addition to reducing proliferation of ERα+ cancer cells, these treatments also alter signals to multiple other target cells in the environment, including immune cell subpopulations, cancer-associated fibroblasts, and endothelial cells via several distinct estrogen receptors. In this review, we update progress in our understanding of the stromal cells populating the microenvironments of primary and metastatic ER+ tumors, the effects of estrogen on tumor and stromal cells to modulate immune activity and the extracellular matrix, and net outcomes in experimental and clinical studies. We highlight new approaches that will illuminate the unique biology of these cancers, provide the foundation for developing new treatment and prevention strategies, and reduce mortality of this disease.
Collapse
Affiliation(s)
- Linda A. Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | | |
Collapse
|
8
|
Gustafsson A, Garre E, Leiva MC, Salerno S, Ståhlberg A, Landberg G. Patient-derived scaffolds as a drug-testing platform for endocrine therapies in breast cancer. Sci Rep 2021; 11:13334. [PMID: 34172801 PMCID: PMC8233392 DOI: 10.1038/s41598-021-92724-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/08/2021] [Indexed: 12/26/2022] Open
Abstract
Three-dimensional cell culture platforms based on decellularised patient-based microenvironments provide in vivo-like growth conditions allowing cancer cells to interact with intact structures and components of the surrounding tissue. A patient-derived scaffold (PDS) model was therefore evaluated as a testing platform for the endocrine therapies (Z)-4-Hydroxytamoxifen (4OHT) and fulvestrant as well as the CDK4/6-inhibitor palbociclib, monitoring the treatment responses in breast cancer cell lines MCF7 and T47D adapted to the patient-based microenvironments. MCF7 cells growing in PDSs showed increased resistance to 4OHT and fulvestrant treatment (100- and 20-fold) compared to 2D cultures. Quantitative PCR analyses of endocrine treated cancer cells in PDSs revealed upregulation of pluripotency markers further supported by increased self-renewal capacity in sphere formation assays. When comparing different 3D growth platforms including PDS, matrigel, gelatin sponges and 3D-printed hydrogels, 3D based cultures showed slightly varying responses to fulvestrant and palbociclib whereas PDS and matrigel cultures showed more similar gene expression profiles for 4OHT treatment compared to the other platforms. The results support that the PDS technique maximized to provide a multitude of smaller functional PDS replicates from each primary breast cancer, is an up-scalable patient-derived drug-testing platform available for gene expression profiling and downstream functional assays.
Collapse
Affiliation(s)
- Anna Gustafsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Elena Garre
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Maria Carmen Leiva
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Simona Salerno
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41390, Gothenburg, Sweden
- Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, 41390, Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, 41390, Gothenburg, Sweden
| | - Göran Landberg
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41390, Gothenburg, Sweden.
| |
Collapse
|
9
|
Mohebi M, Sattari A, Ghafouri-Fard S, Modarressi MH, Kholghi-Oskooei V, Taheri M. Expression profiling revealed up-regulation of three lncRNAs in breast cancer samples. Exp Mol Pathol 2020; 117:104544. [PMID: 32976818 DOI: 10.1016/j.yexmp.2020.104544] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 09/12/2020] [Accepted: 09/19/2020] [Indexed: 01/31/2023]
Abstract
Long non-coding RNAs (lncRNAs) have been vastly investigated for their critical roles in the pathogenesis of breast cancer. Yet, the expression pattern and clinical significance of three lncRNAs namely CTBP1AS2, LINC-ROR and SPRY4-IT1 in breast cancer are not completely clarified. In the present investigation, we assessed expression of these lncRNAs in breast cancer tissues and paired non-cancerous specimens from the same patients using quantitative real time PCR. Notably, expression of CTBP1AS2, LINC-ROR and SPRY4-IT1 were upregulated in breast cancer tissues compared with non-cancerous tissues (ER = 17.62, P value<0.000; ER = 4.62, P value = 0.001 and ER = 3.47, P value = 0.005, respectively). Relative expression of LINC-ROR in tumoral tissues compared with non-tumoral tissues was associated with a history of hormone replacement therapy (P = 0.04). Expression levels of CTBP1AS2, LINC-ROR and SPRY4-IT1 were significantly correlated with each other in both tumoral and non-tumoral tissues. The strongest correlations were detected between CTBP1AS2/ LINC-ROR and CTBP1AS2/ SPRY4-IT1 pairs in non-tumoral tissues. CTBP1AS2 and SPRY4-IT1 had the best sensitivity (80%) and specificity (64%) values, respectively. Based on AUC values, the best diagnostic power belonged to CTBP1AS2. The current study potentiates CTBP1AS2, LINC-ROR and SPRY4-IT1 as putative contributors in the pathogenesis of breast cancer and suggests these lncRNAs as candidates for functional analysis in this kind of cancer.
Collapse
Affiliation(s)
- Mehdi Mohebi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Sattari
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Vahid Kholghi-Oskooei
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Bilyi DD, Gerdeva AА, Samoiliuk VV, Suslova NI, Yevtushenko ID. A modern look at the molecular-biological mechanisms of breast tumours in dogs. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
High morbidity and increase in the number of registrations of breast tumours in dogs, their wide application as biological models, discussion of numerous questions of oncogenesis, and the lack of a uniform/unified methodological approach to the study of molecular and biological mechanisms of treatment of cancer determine the relevance of the problem of cancer both in humans and in our domestic companions. The analysis of publications allowed us to establish the following patterns of carcinogenesis. The peculiarities of the biological behaviour of breast tumours depend on their pathomorphological structure. Genetic predisposition to breast cancer is characteristic only in the single breed aspect. Environmental factors are of critical relevance to carcinogenesis : chemical pollutants initiate oncogenesis indirectly – by altering the expression of several receptors, impaired endocrine balance and direct mutagenic effects. Reproductive status plays a key role in the initiation and progression of breast tumours by reducing the expression of estrogen, progesterone and prolactin receptor genes. The inflammatory response that accompanies the neoplasia process is characterized by increased production of cytokines, cyclooxygenase-2, interleukins (IL-1, IL-6, IL-8), vascular endothelial growth receptors, and impaired hemostatic status (oxidative stress), which promotes progression of disease. In breast cancer in dogs, genomic instability leads to genomic aberrations, and subsequently, mutations that support the proliferation, survival and dissemination of neoplastic cells. The initiation and progression of mammary gland tumours is provided by cancer stem cells by disrupting the regulation of precursor cell self-renewal, which also predispose to resistance to chemotherapeutic agents, radiation, and hormonal cancer therapy. The analysis of the publications revealed the major markers of carcinogenesis that could potentially be used as biological targets for the design of modern diagnostic strategies and high-performance therapeutic protocols.
Collapse
|
11
|
Hermawan A, Putri H. Bioinformatics Studies Provide Insight into Possible Target and Mechanisms of Action of Nobiletin against Cancer Stem Cells. Asian Pac J Cancer Prev 2020; 21:611-620. [PMID: 32212785 PMCID: PMC7437309 DOI: 10.31557/apjcp.2020.21.3.611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Nobiletin treatment on MDA-MB 231 cells reduces the expression of CXC chemokine receptor type 4 (CXCR4), which is highly expressed in cancer stem cell populations in tumor patients. However, the mechanisms of nobiletin in cancer stem cells (CSCs) remain elusive. This study was aimed to explore the potential target and mechanisms of nobiletin in cancer stem cells using bioinformatics approaches. METHODS Gene expression profiles by public COMPARE predicting the sensitivity of tumor cells to nobiletin. Functional annotations on gene lists are carried out with The Database for Annotation, Visualization and Integrated Discovery (DAVID) v6.8, and WEB-based GEne SeT Analysis Toolkit (WebGestalt). The protein-protein interaction (PPI) network was analyzed by STRING-DB and visualized by Cytoscape. RESULTS Microarray analyses reveal many genes involved in protein binding, transcriptional and translational activity. Pathway enrichment analysis revealed breast cancer regulation of estrogen signaling and Wnt/ß-catenin by nobiletin. Moreover, three hub genes, i.e. ESR1, NCOA3, and RPS6KB1 and one significant module were filtered out and selected from the PPI network. CONCLUSION Nobiletin might serve as a lead compound for the development of CSCs-targeted drugs by targeting estrogen and Wnt/ß-catenin signaling. Further studies are needed to explore the full therapeutic potential of nobiletin in cancer stem cells. .
Collapse
Affiliation(s)
| | - Herwandhani Putri
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
12
|
López-Mateo I, Alonso-Merino E, Suarez-Cabrera C, Park JW, Cheng SY, Alemany S, Paramio JM, Aranda A. Thyroid Hormone Receptor β Inhibits Self-Renewal Capacity of Breast Cancer Stem Cells. Thyroid 2020; 30:116-132. [PMID: 31760908 PMCID: PMC6998057 DOI: 10.1089/thy.2019.0175] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: A subpopulation of cancer stem cells (CSCs) with capacity for self-renewal is believed to drive initiation, progression, and relapse of breast tumors. Methods: Since the thyroid hormone receptor β (TRβ) appears to suppress breast tumor growth and metastasis, we have analyzed the possibility that TRβ could affect the CSC population using MCF-7 cells grown under adherent conditions or as mammospheres, as well as inoculation into immunodeficient mice. Results: Treatment of TRβ-expressing MCF-7 cells (MCF7-TRβ cells) with the thyroid hormone triiodothyronine (T3) decreased significantly CD44+/CD24- and ALDH+ cell subpopulations, the efficiency of mammosphere formation, the self-renewal capacity of CSCs in limiting dilution assays, the expression of the pluripotency factors in the mammospheres, and tumor initiating capacity in immunodeficient mice, indicating that the hormone reduces the CSC population present within the bulk MCF7-TRβ cultures. T3 also decreased migration and invasion, a hallmark of CSCs. Transcriptome analysis showed downregulation of the estrogen receptor alpha (ERα) and ER-responsive genes by T3. Furthermore, among the T3-repressed genes, there was an enrichment in genes containing binding sites for transcription factors that are key determinants of luminal-type breast cancers and are required for ER binding to chromatin. Conclusion: We demonstrate a novel role of TRβ in the biology of CSCs that may be related to its action as a tumor suppressor in breast cancer.
Collapse
Affiliation(s)
- Irene López-Mateo
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
| | - Elvira Alonso-Merino
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
| | | | - Jeong Won Park
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland
| | - Susana Alemany
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
| | - Jesús M. Paramio
- Molecular Oncology Unit, CIEMAT, Madrid, Spain
- Institute of Biomedical Research, Hosp Univ. “12 de Octubre,” Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ana Aranda
- Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols,” Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Address correspondence to: Ana Aranda, PhD, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Instituto de Investigaciones Biomédicas “Alberto Sols”, Arturo Duperier 4, Madrid 28029, Spain
| |
Collapse
|
13
|
Flores-Ramírez I, Baranda-Avila N, Langley E. Breast Cancer Stem Cells and Sex Steroid Hormones. Curr Stem Cell Res Ther 2019; 14:398-404. [PMID: 30095060 DOI: 10.2174/1574888x13666180810121415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Breast cancer stem cells (BCSCs) are a small population of tumor-initiating cells that express stem cell-associated markers. In recent years, their properties and mechanisms of regulation have become the focus of intense research due to their intrinsic resistance to conventional cancer therapies. This review describes breast cancer stem cell origin, signaling pathways involved in self-renewal, such as Wnt, Notch and Hedgehog, biomarkers linked to stemness, and the role of sex steroid hormones in BCSC regulation.
Collapse
Affiliation(s)
- Iván Flores-Ramírez
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, México.,Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| | - Noemi Baranda-Avila
- Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| | - Elizabeth Langley
- Departamento de Investigacion Basica, Instituto Nacional de Cancerologia, Av. San Fernando No. 22, Col. Seccion XVI, Tlalpan 14080, CDMX, Mexico
| |
Collapse
|
14
|
Cenciarini ME, Proietti CJ. Molecular mechanisms underlying progesterone receptor action in breast cancer: Insights into cell proliferation and stem cell regulation. Steroids 2019; 152:108503. [PMID: 31562879 DOI: 10.1016/j.steroids.2019.108503] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The ovarian steroid hormone progesterone and its nuclear receptor, the Progesterone Receptor (PR), play an essential role in the regulation of cell proliferation and differentiation in the mammary gland. In addition, experimental and clinical evidence demonstrate their critical role in controlling mammary gland tumorigenesis and breast cancer development. When bound to its ligand, the main action of PR is as a transcription factor, which regulates the expression of target genes networks. PR also activates signal transduction pathways through a rapid or non-genomic mechanism in breast cancer cells, an event that is fully integrated with its genomic effects. This review summarizes the molecular mechanisms of the ligand-activated PR actions that drive epithelial cell proliferation and the regulation of the stem cell population in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Mauro E Cenciarini
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina.
| |
Collapse
|
15
|
Wanandi SI, Syahrani RA, Arumsari S, Wideani G, Hardiany NS. Profiling of Gene Expression Associated with Stemness and Aggressiveness of ALDH1A1-Expressing Human Breast Cancer Cells. Malays J Med Sci 2019; 26:38-52. [PMID: 31728117 PMCID: PMC6839666 DOI: 10.21315/mjms2019.26.5.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/17/2019] [Indexed: 01/06/2023] Open
Abstract
Background It has been widely reported that breast cancer aggressiveness may be driven by breast cancer stem cells (BCSCs). BCSCs display stemness properties that include self-renewal, tumourigenicity and pluripotency. The regulation of gene expression may have important roles in BCSC stemness and aggressiveness. Thus, the aim of this study was to examine the stemness and aggressiveness gene expression profile of BCSCs compared to MCF-7 and MDA-MB-231 breast cancer cells. Methods Human ALDH1+ BCSCs were grown in serum-free Dulbecco’s Modified Eagle Medium (DMEM)/F12, while MCF-7 and MDA-MB-231 were cultured in DMEM supplemented with 10% foetal bovine serum under standard conditions. Total RNA was extracted using the Tripure Isolation Reagent. The relative mRNA expressions of OCT4, ALDH1A1 and CD44 associated with stemness as well as TGF-β1, TβR1, ERα1 and MnSOD associated with aggressiveness in BCSCs and MCF-7 cells were determined using the quantitative real-time PCR (qRT-PCR). Results The mRNA expressions of OCT4 (5.19-fold ± 0.338; P = 0.001), ALDH1A1 (3.67-fold ± 0.523; P = 0.006), CD44 (2.65-fold ± 0.307; P = 0.006), TGF-β1 (22.89-fold ± 6.840; P = 0.015), TβR1 (3.74-fold ± 1.446; P = 0.045) and MnSOD (4.6-fold ± 1.096; P = 0.014) were higher in BCSCs than in MCF-7 but were almost similar to MDA-MB-231 cells. In contrast, the ERα1 expression of BCSCs (0.97-fold ± 0.080; P = 0.392) was similar to MCF-7 cells, indicating that BSCSs are oestrogen-dependent breast cancer cells. Conclusion The oestrogen-dependent BCSCs express stemness and aggressiveness genes at a higher level compared to oestrogen-dependent MCF-7 but are almost similar to oestrogen-independent MDA-MB-231 cells.
Collapse
Affiliation(s)
- Septelia Inawati Wanandi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Molecular Biology and Proteomics Core Facilities, IMERI-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Resda Akhra Syahrani
- Molecular Biology and Proteomics Core Facilities, IMERI-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Sekar Arumsari
- Molecular Biology and Proteomics Core Facilities, IMERI-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Gita Wideani
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Novi Silvia Hardiany
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Molecular Biology and Proteomics Core Facilities, IMERI-Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
16
|
Yousefnia S, Ghaedi K, Seyed Forootan F, Nasr Esfahani MH. Characterization of the stemness potency of mammospheres isolated from the breast cancer cell lines. Tumour Biol 2019; 41:1010428319869101. [DOI: 10.1177/1010428319869101] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Stemness phenotype mammospheres established from cell lines and tissues taken from autopsy can be used to test and to identify the most sensitive drugs for chemotherapy. Therefore, the aim of the present study was isolation and characterization of cancer stem cells derived from MCF7, MDA-MB231, and SKBR3 breast cancer cell lines to demonstrate the stemness phenotypes of mammospheres generated for further their applications in therapeutic approaches. In this study, two luminal subtypes of cell lines, MCF7 and SKBR3 and a basal subtype cell line, MDA-MB-231, were chosen. Mammosphere culturing was implemented for breast cancer stem cells isolation and mammosphere formation efficiency. At the next step, CD44+/CD24– cell ratio, Oct4 and Nanog mRNA levels, proliferation rate, migration rate of mammospheres, and drug resistance (in third passage) were evaluated. In addition, tumorigenicity of mammospheres in the chick embryo model was evaluated and compared through the chick chorioallantoic membrane assay. Among mammospheres formed in all three cell lines, MCF7 had the highest mammosphere formation efficiency. CD24 marker (a differentiation marker for the breast cancer cells) was significantly reduced in the mammospheres generated from MCF7 and SKBR3, during three passages. Also, Oct4 and Nanog transcript levels were significantly higher in all three types of mammospheres, as compared with their cell lines. Proliferation, migration rate, and drug resistance of mammospheres generated from all three cell lines were found to be significantly higher. Tumorigenicity of MCF7 mammospheres was confirmed through tumor size measurement. Also, tumorigenicity of MCF7 and SKBR3 mammospheres was confirmed through more migration from ectoderm to mesoderm and endoderm. We succeeded to establish the technology that can be extended to tissue in the future. We have demonstrated a number of mammospheres can be generated from cell lines. Also, cells with different molecular features showed different stemness phenotypes.
Collapse
Affiliation(s)
- Saghar Yousefnia
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Kamran Ghaedi
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Academic Center for Education, Culture and Research, Isfahan, Iran
| | - Farzad Seyed Forootan
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Academic Center for Education, Culture and Research, Isfahan, Iran
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Mohammad Hossein Nasr Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, Academic Center for Education, Culture and Research, Isfahan, Iran
| |
Collapse
|
17
|
Nasr M, Farghaly M, Elsaba T, El-Mokhtar M, Radwan R, Elsabahy M, Abdelkareem A, Fakhry H, Mousa N. Resistance of primary breast cancer cells with enhanced pluripotency and stem cell activity to sex hormonal stimulation and suppression. Int J Biochem Cell Biol 2018; 105:84-93. [PMID: 30359767 DOI: 10.1016/j.biocel.2018.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023]
Abstract
Female sex steroid hormones have a fundamental role in breast cancer. Meanwhile, current evidence supports the contribution of breast cancer stem cells in carcinogenesis, metastasis, and resistance to cytotoxic chemotherapy. Nevertheless, the interaction between breast cancer stem cells with sex hormones or key hormonal antagonists remains elusive. OBJECTIVE To investigate the effect of diverse sex hormonal stimulation and suppression regimens on the proliferation of a primary human breast cancer cells with stem cell activity. METHODS Cells were exposed to estradiol, progesterone, letrozole, ulipristal acetate, or a combination of ulipristal acetate-letrozole, continually for 6 months. Additionally, nanoparticle-linked letrozole and ulipristal acetate formulations were included in a subsequent short-term exposure study. Phenotypic, pathologic, and functional characteristics of unexposed cells were investigated. RESULTS The proliferation of breast cancer cells was comparable among all hormonal stimulation and suppression groups (P= 0.8). In addition, the nanoparticle encapsulated hormonal antagonists were not able to overcome the observed resistance of cells. Cell characterization showed a mesenchymal-like phenotype overexpressing three master pluripotency markers (Oct 4, SOX2, and Nanog), and 92% of cells were expressing ALDH1A1. Notably, the CD44 high/CD24 low cell population presented only 0.97%-5.4% over repeat analyses. Most cells lacked the expression of mesenchymal markers; however, they showed differentiation into osteogenic and adipogenic lineages. Upon transfer to serum-free culture, the long-term maintained mesenchymal-like cancer cells showed remarkable morphologic plasticity as they switched promptly into an epithelial-like phenotype with significant mammosphere formation capacity (P= 0.008). CONCLUSION Breast cancer cells can develop a pluripotent program with enhanced stemness activity that may together contribute to universal resistance to sex hormonal stimulation or deprivation. Isolation and characterization of patient-derived breast cancer stem cells in large clinical studies is therefore crucial to identify new targets for endocrine therapies, potentially directed towards stemness and pluripotency markers. Such direction may help overcoming endocrine resistance and draw attention to breast cancer stem cells' behaviour under endogenous and exogenous sex hormones throughout a woman's reproductive life.
Collapse
Affiliation(s)
| | | | - Tarek Elsaba
- South Egypt Cancer Institute, Assiut University, Egypt
| | | | - Radwa Radwan
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt
| | - Mahmoud Elsabahy
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt; Laboratory for Synthetic-Biologic Interactions, Department of Chemistry, Texas A&M University, College Station, TX, USA
| | | | | | - Noha Mousa
- Zewail City of Science and Technology, Egypt.
| |
Collapse
|
18
|
Pelekanou V, Notas G, Athanasouli P, Alexakis K, Kiagiadaki F, Peroulis N, Kalyvianaki K, Kampouri E, Polioudaki H, Theodoropoulos P, Tsapis A, Castanas E, Kampa M. BCMA (TNFRSF17) Induces APRIL and BAFF Mediated Breast Cancer Cell Stemness. Front Oncol 2018; 8:301. [PMID: 30131941 PMCID: PMC6091000 DOI: 10.3389/fonc.2018.00301] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/18/2018] [Indexed: 01/16/2023] Open
Abstract
Recent advances in cancer immunology revealed immune-related properties of cancer cells as novel promising therapeutic targets. The two TNF superfamily members, APRIL (TNFSF13), and BAFF (TNFSF13B), which are type II membrane proteins, released in active forms by proteolytic cleavage and are primarily involved in B-lymphocyte maturation, have also been associated with tumor growth and aggressiveness in several solid tumors, including breast cancer. In the present work we studied the effect of APRIL and BAFF on epithelial to mesenchymal transition, migration, and stemness of breast cancer cells. Our findings show that both molecules increase epithelial to mesenchymal transition and migratory capacity of breast cancer cells, as well as cancer stem cell numbers, by increasing the expression of pluripotency genes such as ALDH1A1, KLF4, and NANOG. These effects are mediated by their common receptor BCMA (TNFRSF17) and the JNK signaling pathway. Interestingly, transcriptional data analysis from breast cancer cells and patients revealed that androgens can increase APRIL transcription and subsequently, in an autocrine/paracrine manner, enhance its pluripotency effect. In conclusion, our data suggest a possible role of APRIL and BAFF in breast cancer disease progression and provide evidence for a new possible mechanism of therapy resistance, that could be particularly relevant in aromatase inhibitors-treated patients, were local androgen is increased.
Collapse
Affiliation(s)
- Vasiliki Pelekanou
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Paraskevi Athanasouli
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Konstantinos Alexakis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Fotini Kiagiadaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Nikolaos Peroulis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Konstantina Kalyvianaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Errika Kampouri
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Hara Polioudaki
- Department of Biochemistry, School of Medicine, University of Crete, Heraklion, Greece
| | | | - Andreas Tsapis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
19
|
Gelsomino L, Panza S, Giordano C, Barone I, Gu G, Spina E, Catalano S, Fuqua S, Andò S. Mutations in the estrogen receptor alpha hormone binding domain promote stem cell phenotype through notch activation in breast cancer cell lines. Cancer Lett 2018; 428:12-20. [DOI: 10.1016/j.canlet.2018.04.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/17/2018] [Accepted: 04/19/2018] [Indexed: 12/21/2022]
|
20
|
Jokela TA, Engelsen AST, Rybicka A, Pelissier Vatter FA, Garbe JC, Miyano M, Tiron C, Ferariu D, Akslen LA, Stampfer MR, Lorens JB, LaBarge MA. Microenvironment-Induced Non-sporadic Expression of the AXL and cKIT Receptors Are Related to Epithelial Plasticity and Drug Resistance. Front Cell Dev Biol 2018; 6:41. [PMID: 29719832 PMCID: PMC5913284 DOI: 10.3389/fcell.2018.00041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/23/2018] [Indexed: 12/13/2022] Open
Abstract
The existence of rare cancer cells that sporadically acquire drug-tolerance through epigenetic mechanisms is proposed as one mechanism that drives cancer therapy failure. Here we provide evidence that specific microenvironments impose non-sporadic expression of proteins related to epithelial plasticity and drug resistance. Microarrays of robotically printed combinatorial microenvironments of known composition were used to make cell-based functional associations between microenvironments, which were design-inspired by normal and tumor-burdened breast tissues, and cell phenotypes. We hypothesized that specific combinations of microenvironment constituents non-sporadically impose the induction of the AXL and cKIT receptor tyrosine kinase proteins, which are known to be involved in epithelial plasticity and drug-tolerance, in an isogenic human mammary epithelial cell (HMEC) malignant progression series. Dimension reduction analysis reveals type I collagen as a dominant feature, inducing expression of both markers in pre-stasis finite lifespan HMECs, and transformed non-malignant and malignant immortal cell lines. Basement membrane-associated matrix proteins, laminin-111 and type IV collagen, suppress AXL and cKIT expression in pre-stasis and non-malignant cells. However, AXL and cKIT are not suppressed by laminin-111 in malignant cells. General linear models identified key factors, osteopontin, IL-8, and type VIα3 collagen, which significantly upregulated AXL and cKIT, as well as a plasticity-related gene expression program that is often observed in stem cells and in epithelial-to-mesenchymal-transition. These factors are co-located with AXL-expressing cells in situ in normal and breast cancer tissues, and associated with resistance to paclitaxel. A greater diversity of microenvironments induced AXL and cKIT expression consistent with plasticity and drug-tolerant phenotypes in tumorigenic cells compared to normal or immortal cells, suggesting a reduced perception of microenvironment specificity in malignant cells. Microenvironment-imposed reprogramming could explain why resistant cells are seemingly persistent and rapidly adaptable to multiple classes of drugs. These results support the notion that specific microenvironments drive drug-tolerant cellular phenotypes and suggest a novel interventional avenue for preventing acquired therapy resistance.
Collapse
Affiliation(s)
- Tiina A. Jokela
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Population Sciences, Center for Cancer and Aging, City of Hope, Duarte, CA, United States
| | - Agnete S. T. Engelsen
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Agata Rybicka
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | - James C. Garbe
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Masaru Miyano
- Department of Population Sciences, Center for Cancer and Aging, City of Hope, Duarte, CA, United States
| | - Crina Tiron
- Regional Institute of Oncology, Iasi, Romania
| | - Dan Ferariu
- Regional Institute of Oncology, Iasi, Romania
| | - Lars A. Akslen
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Martha R. Stampfer
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - James B. Lorens
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Mark A. LaBarge
- Department of Population Sciences, Center for Cancer and Aging, City of Hope, Duarte, CA, United States
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
21
|
Chen KHE, Bustamante K, Nguyen V, Walker AM. Involvement of miR-106b in tumorigenic actions of both prolactin and estradiol. Oncotarget 2018; 8:36368-36382. [PMID: 28422740 PMCID: PMC5482661 DOI: 10.18632/oncotarget.16755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023] Open
Abstract
Prolactin promotes a variety of cancers by an array of different mechanisms. Here, we have investigated prolactin's inhibitory effect on expression of the cell cycle-regulating protein, p21. Using a miRNA array, we identified a number of miRNAs upregulated by prolactin treatment, but one in particular that was strongly induced by prolactin and predicted to bind to the 3′UTR of p21 mRNA, miR-106b. By creating a p21 mRNA 3′UTR-luciferase mRNA construct, we demonstrated degradation of the construct in response to prolactin in human breast, prostate and ovarian cancer cell lines. Increased expression of miR-106b replicated, and anti-miR-106b counteracted, the effects of prolactin on degradation of the 3′UTR construct, p21 mRNA levels, and cell proliferation in breast (T47D) and prostate (PC3) cancer cells. Increased expression of miR-106b also stimulated migration of the very epithelioid T47D cell line. By contrast, anti-miR-106b dramatically decreased expression of the mesenchymal markers, SNAIL-2, TWIST-2, VIMENTIN, and FIBRONECTIN. Using signaling pathway inhibitors and the 3′UTR construct, induction of miR-106b by prolactin was determined to be mediated through the MAPK/ERK and PI3K/Akt pathways and not through Jak2/Stat5 in both T47D and PC3 cells. Prolactin activation of MAPK/ERK and PI3K/Akt also activates ERα in the absence of an ERα ligand. 17β-estradiol promoted degradation of the construct in both cell lines and pre-incubation in the estrogen antagonist, Fulvestrant, blocked the ability of both prolactin and 17β-estradiol to induce the construct-degrading activity. Together, these data support a convergence of the prolactin and 17β-estradiol miR-106b-elevating signaling pathways at ERα.
Collapse
Affiliation(s)
- Kuan-Hui Ethan Chen
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Karissa Bustamante
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Vi Nguyen
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| | - Ameae M Walker
- Division of Biomedical Sciences, University of California, Riverside, CA 92521, USA
| |
Collapse
|
22
|
Alferez DG, Simões BM, Howell SJ, Clarke RB. The Role of Steroid Hormones in Breast and Effects on Cancer Stem Cells. CURRENT STEM CELL REPORTS 2018; 4:81-94. [PMID: 29600163 PMCID: PMC5866269 DOI: 10.1007/s40778-018-0114-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Purpose of Review This review will discuss how the steroid hormones, estrogen and progesterone, as well as treatments that target steroid receptors, can regulate cancer stem cell (CSC) activity. The CSC theory proposes a hierarchical organization in tumors where at its apex lies a subpopulation of cancer cells endowed with self-renewal and differentiation capacity. Recent Findings In breast cancer (BC), CSCs have been suggested to play a key role in tumor maintenance, disease progression, and the formation of metastases. In preclinical models of BC, only a few CSCs are required sustain tumor re-growth, especially after conventional anti-endocrine treatments. CSCs include therapy-resistant clones that survive standard of care treatments like chemotherapy, irradiation, and hormonal therapy. Summary The relevance of hormones for both normal mammary gland and BC development is well described, but it was only recently that the activities of hormones on CSCs have been investigated, opening new directions for future BC treatments and CSCs.
Collapse
Affiliation(s)
- Denis G. Alferez
- Breast Biology Group, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Cancer Research Centre, Wilmslow Road, Manchester, M20 4GJ UK
| | - Bruno M. Simões
- Breast Biology Group, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Cancer Research Centre, Wilmslow Road, Manchester, M20 4GJ UK
| | - Sacha J. Howell
- Breast Biology Group, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Cancer Research Centre, Wilmslow Road, Manchester, M20 4GJ UK
- Department of Medical Oncology, The University of Manchester, The Christie NHS Foundation Trust, Manchester, M20 4BX UK
| | - Robert B. Clarke
- Breast Biology Group, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Cancer Research Centre, Wilmslow Road, Manchester, M20 4GJ UK
| |
Collapse
|
23
|
Shea MP, O'Leary KA, Fakhraldeen SA, Goffin V, Friedl A, Wisinski KB, Alexander CM, Schuler LA. Antiestrogen Therapy Increases Plasticity and Cancer Stemness of Prolactin-Induced ERα + Mammary Carcinomas. Cancer Res 2018; 78:1672-1684. [PMID: 29363543 DOI: 10.1158/0008-5472.can-17-0985] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 10/31/2017] [Accepted: 01/19/2018] [Indexed: 12/24/2022]
Abstract
Although antiestrogen therapies are successful in many patients with estrogen receptor alpha-positive (ERα+) breast cancer, 25% to 40% fail to respond. Although multiple mechanisms underlie evasion of these treatments, including tumor heterogeneity and drug-resistant cancer stem cells (CSC), further investigations have been limited by the paucity of preclinical ERα+ tumor models. Here, we examined a mouse model of prolactin-induced aggressive ERα+ breast cancer, which mimics the epidemiologic link between prolactin exposure and increased risk for metastatic ERα+ tumors. Like a subset of ERα+ patient cancers, the prolactin-induced adenocarcinomas contained two major tumor subpopulations that expressed markers of normal luminal and basal epithelial cells. CSC activity was distributed equally across these two tumor subpopulations. Treatment with the selective estrogen receptor downregulator (SERD), ICI 182,780 (ICI), did not slow tumor growth, but induced adaptive responses in CSC activity, increased markers of plasticity including target gene reporters of Wnt/Notch signaling and epithelial-mesenchymal transition, and increased double-positive (K8/K5) cells. In primary tumorsphere cultures, ICI stimulated CSC self-renewal and was able to overcome the dependence of self-renewal upon Wnt or Notch signaling individually, but not together. Our findings demonstrate that treatment of aggressive mixed lineage ERα+ breast cancers with a SERD does not inhibit growth, but rather evokes tumor cell plasticity and regenerative CSC activity, predicting likely negative impacts on patient tumors with these characteristics.Significance: This study suggests that treatment of a subset of ERα+ breast cancers with antiestrogen therapies may not only fail to slow growth but also promote aggressive behavior by evoking tumor cell plasticity and regenerative CSC activity. Cancer Res; 78(7); 1672-84. ©2018 AACR.
Collapse
Affiliation(s)
- Michael P Shea
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kathleen A O'Leary
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Saja A Fakhraldeen
- Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Vincent Goffin
- Inserm Unit 1151, Institut Necker Enfants Malades (INEM), Université Paris Descartes, Paris, France
| | - Andreas Friedl
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kari B Wisinski
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Caroline M Alexander
- Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin.,University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin. .,University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
24
|
Martignani E, Accornero P, Miretti S, Baratta M. Bovine Mammary Organoids: A Model to Study Epithelial Mammary Cells. Methods Mol Biol 2018; 1817:137-144. [PMID: 29959710 DOI: 10.1007/978-1-4939-8600-2_14] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bovine mammary organoids are cell aggregates that are produced by an association of a mechanical and an enzymatic dissociation of mammary gland tissue. They provide a useful source to isolate mammary epithelial cells, but can also be frozen as an intermediate dissociation step.Due to the strong cell-cell interactions among epithelial cells, the production and isolation of organoids is an efficient way to remove unwanted cell population of non-epithelial origin like fibroblasts.
Collapse
Affiliation(s)
- Eugenio Martignani
- Department of Veterinary Science, University of Turin, Grugliasco, TO, Italy.
| | - Paolo Accornero
- Department of Veterinary Science, University of Turin, Grugliasco, TO, Italy
| | - Silvia Miretti
- Department of Veterinary Science, University of Turin, Grugliasco, TO, Italy
| | - Mario Baratta
- Department of Veterinary Science, University of Turin, Grugliasco, TO, Italy
| |
Collapse
|
25
|
Ables ET, Drummond-Barbosa D. Steroid Hormones and the Physiological Regulation of Tissue-Resident Stem Cells: Lessons from the Drosophila Ovary. CURRENT STEM CELL REPORTS 2017; 3:9-18. [PMID: 28458991 PMCID: PMC5407287 DOI: 10.1007/s40778-017-0070-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Stem cells respond to local paracrine signals; more recently, however, systemic hormones have also emerged as key regulators of stem cells. This review explores the role of steroid hormones in stem cells, using the Drosophila germline stem cell as a centerpiece for discussion. RECENT FINDINGS Stem cells sense and respond directly and indirectly to steroid hormones, which regulate diverse sets of target genes via interactions with nuclear hormone receptors. Hormone-regulated networks likely integrate the actions of multiple systemic signals to adjust the activity of stem cell lineages in response to changes in physiological status. SUMMARY Hormones are inextricably linked to animal physiology, and can control stem cells and their local niches. Elucidating the molecular mechanisms of hormone signaling in stem cells is essential for our understanding of the fundamental underpinnings of stem cell biology, and for informing new therapeutic interventions against cancers or for regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth T. Ables
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
26
|
Atashgaran V, Wrin J, Barry SC, Dasari P, Ingman WV. Dissecting the Biology of Menstrual Cycle-Associated Breast Cancer Risk. Front Oncol 2016; 6:267. [PMID: 28083513 PMCID: PMC5183603 DOI: 10.3389/fonc.2016.00267] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022] Open
Abstract
Fluctuations in circulating estrogen and progesterone across the menstrual cycle lead to increased breast cancer susceptibility in women; however, the biological basis for this increased risk is not well understood. Estrogen and progesterone have important roles in normal mammary gland development, where they direct dynamic interactions among the hormonally regulated mammary epithelial, stromal, and immune cell compartments. The continuous fluctuations of estrogen and progesterone over a woman’s reproductive lifetime affect the turnover of mammary epithelium, stem cells, and the extracellular matrix, as well as regulate the phenotype and function of mammary stromal and immune cells, including macrophages and regulatory T cells. Collectively, these events may result in genome instability, increase the chance of random genetic mutations, dampen immune surveillance, and promote tolerance in the mammary gland, and thereby increase the risk of breast cancer initiation. This article reviews the current status of our understanding of the molecular and the cellular changes that occur in the mammary gland across the menstrual cycle and how continuous menstrual cycling may increase breast cancer susceptibility in women.
Collapse
Affiliation(s)
- Vahid Atashgaran
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Adelaide, SA, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Joseph Wrin
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Adelaide, SA, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Simon Charles Barry
- The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia; Molecular Immunology Laboratory, Discipline of Pediatrics, University of Adelaide, North Adelaide, SA, Australia
| | - Pallave Dasari
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Adelaide, SA, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Wendy V Ingman
- Discipline of Surgery, School of Medicine, The Queen Elizabeth Hospital, University of Adelaide, Adelaide, SA, Australia; The Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
27
|
Grimm SL, Hartig SM, Edwards DP. Progesterone Receptor Signaling Mechanisms. J Mol Biol 2016; 428:3831-49. [PMID: 27380738 DOI: 10.1016/j.jmb.2016.06.020] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/25/2016] [Accepted: 06/27/2016] [Indexed: 12/27/2022]
Abstract
Progesterone receptor (PR) is a master regulator in female reproductive tissues that controls developmental processes and proliferation and differentiation during the reproductive cycle and pregnancy. PR also plays a role in progression of endocrine-dependent breast cancer. As a member of the nuclear receptor family of ligand-dependent transcription factors, the main action of PR is to regulate networks of target gene expression in response to binding its cognate steroid hormone, progesterone. This paper summarizes recent advances in understanding the structure-function properties of the receptor protein and the tissue/cell-type-specific PR signaling pathways that contribute to the biological actions of progesterone in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Sandra L Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA.
| |
Collapse
|
28
|
A Role for Notch Signalling in Breast Cancer and Endocrine Resistance. Stem Cells Int 2016; 2016:2498764. [PMID: 26880941 PMCID: PMC4736972 DOI: 10.1155/2016/2498764] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/20/2015] [Indexed: 12/16/2022] Open
Abstract
Over the past decade, there has been growing interest in the Notch signalling pathway within the breast cancer field. This interest stemmed initially from the observation that Notch signalling is aberrantly activated in breast cancer and its effects on various cellular processes including proliferation, apoptosis, and cancer stem cell activity. However more recently, elevated Notch signalling has been correlated with therapy resistance in oestrogen receptor-positive breast cancer. As a result, inhibiting Notch signalling with therapeutic agents is being explored as a promising treatment option for breast cancer patients.
Collapse
|
29
|
Affiliation(s)
- Dean G Tang
- Department of Epigenetics and Molecular CarcinogenesisThe University of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China Department of Epigenetics and Molecular CarcinogenesisThe University of Texas MD Anderson Cancer Center, Science Park, Park Road 1C, Smithville, Texas 78957, USACancer Stem Cell InstituteResearch Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| |
Collapse
|