1
|
Arvelo F, Sojo F. Transición epitelio – mesenquima y cáncer. INVESTIGACIÓN CLÍNICA 2023; 64:379-404. [DOI: 10.54817/ic.v64n3a10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Cancer cell migration and invasion are critical components of metastatic disease, the leading cause of death in cancer patients. The epithe-lium-mesenchyme-transition (EMT) and mesenchyme-epithelium-transition (MET) are pathways involved in cancer metastasis. This process involves the degradation of cell-cell and cell-extracellular matrix junctions and the subse-quent loss of regulation of binding proteins such as E-cadherin. Cells undergo a reorganization of the cytoskeleton. These alterations are associated with a change in cell shape from epithelial to mesenchymal morphology. Understand-ing EMT and MET’s molecular and cellular basis provides fundamental insights into cancer etiology and may lead to new therapeutic strategies. In this review, we discuss some of the regulatory mechanisms and pathological role of epitheli-al-mesenchymal plasticity, focusing on the knowledge about the complexity and dynamics of this phenomenon in cancer
Collapse
Affiliation(s)
- Francisco Arvelo
- Fundación Instituto de Estudios Avanzados-IDEA, Area Salud, Caracas-Venezuela. Laboratorio de Cultivo de Tejidos y Biología de Tumores, Instituto de Biología Experimental, Universidad Central de Venezuela, Caracas, Venezuela
| | - Felipe Sojo
- Fundación Instituto de Estudios Avanzados-IDEA, Area Salud, Caracas-Venezuela. Laboratorio de Cultivo de Tejidos y Biología de Tumores, Instituto de Biología Experimental, Universidad Central de Venezuela, Caracas, Venezuela
| |
Collapse
|
2
|
Leal-Orta E, Ramirez-Ricardo J, Garcia-Hernandez A, Cortes-Reynosa P, Salazar EP. Extracellular vesicles from MDA-MB-231 breast cancer cells stimulated with insulin-like growth factor 1 mediate an epithelial-mesenchymal transition process in MCF10A mammary epithelial cells. J Cell Commun Signal 2022; 16:531-546. [PMID: 34309795 PMCID: PMC9733745 DOI: 10.1007/s12079-021-00638-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) plays an important role in function and development of the mammary gland. However, high levels of IGF-1 has been associated with an increased risk of breast cancer development. Epithelial-mesenchymal transition (EMT) is a process where epithelial cells lose their epithelial characteristics and acquire a mesenchymal phenotype, which is considered one of the most important mechanisms in cancer initiation and promotion of metastasis. Extracellular vesicles (EVs) are released into the extracellular space by different cell types, which mediate intercellular communication and play an important role in different physiological and pathological processes, such as cancer. In this study, we demonstrate that EVs from MDA-MB-231 breast cancer cells stimulated with IGF-1 (IGF-1 EVs) decrease the levels of E-cadherin, increase the expression of vimentin and N-cadherin and stimulate the secretion of metalloproteinase-9 in mammary non-tumorigenic epithelial cells MCF10A. IGF-1 EVs also induce the expression of Snail1, Twist1 and Sip1, which are transcription factors involved in EMT. Moreover, IGF-1 EVs induce activation of ERK1/2, Akt1 and Akt2, migration and invasion. In summary, we demonstrate, for the first time, that IGF-1 EVs induce an EMT process in mammary non-tumorigenic epithelial cells MCF10A.
Collapse
Affiliation(s)
- Elizabeth Leal-Orta
- grid.512574.0Departamento de Biologia Celular, Cinvestav-IPN, 07360 Mexico City, Mexico
| | | | | | - Pedro Cortes-Reynosa
- grid.512574.0Departamento de Biologia Celular, Cinvestav-IPN, 07360 Mexico City, Mexico
| | - Eduardo Perez Salazar
- grid.512574.0Departamento de Biologia Celular, Cinvestav-IPN, 07360 Mexico City, Mexico
| |
Collapse
|
3
|
Rosendahl AH, Björner S, Ygland Rödström M, Jirström K, Borgquist S, Ingvar C, Pollak MN, Jernström H. Pre- and Postoperative Circulating IGF-I, IGFBP-3, and IGFBP-7 Levels in Relation to Endocrine Treatment and Breast Cancer Recurrence: A Nested Case-Control Study. Front Oncol 2021; 11:626058. [PMID: 33767994 PMCID: PMC7986849 DOI: 10.3389/fonc.2021.626058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/25/2021] [Indexed: 01/18/2023] Open
Abstract
Insulin-like growth factor-I (IGF-I) and its binding proteins (BPs) have been associated with breast cancer risk, especially high IGF-I concentrations and the biologically active fraction estimated as the IGF-I/IGFBP-3 molar ratio. The relation of circulating IGF-I and IGFBP-3 concentrations with risk of breast cancer recurrence has been less documented. In addition a new member to a sub-group of the IGFBP-superfamily was recently identified, the low affinity IGFBP-7. To date, the role of systemic IGFBP-7 in breast cancer progression has not been investigated. Our purpose was to establish whether circulating IGF-I, IGFBP-3, and IGFBP-7 levels are related to recurrence-risk in breast cancer. A case-control study was nested within the population-based BCBlood cohort of 853 breast cancer patients diagnosed 2002-2010 in Sweden and followed through 2012. In total, 95 patients with recurrence and 170 controls were matched on age and tumor characteristics. Plasma IGF analytes and tumor membrane IGF-I receptor (IGF-IRm) positivity were analyzed and recurrence-risk was evaluated with conditional logistic regression. Preoperative tertiles of IGF-I and IGFBP-3 were both positively associated with recurrence-risk, but not IGFBP-7. The trend was of borderline significance for IGF-I, T1:REF, T2 OR:1.6, T3 OR: 2.2 adjusted P trend=0.057 and significant for IGFBP-3 T1:REF, T2 OR:1.2, T3 OR: 2.1 adjusted P trend=0.042. The models were adjusted for age, anthropometric factors, smoking, and treatments. There was a significant interaction between IGFBP-7 and IGF-IRm positivity on recurrence, where the highest IGFBP-7 highest IGFBP-7 tertile conferred increased recurrence-risk in patients with IGF-IRm positive tumors but not in those with IGF-IRm negative tumors (P interaction=0.024). By the 1-year visit, age-adjusted IGF-I levels were reduced by 17% while IGFBP-3 and IGFBP-7 were stable. IGF-I levels were significantly reduced by radiotherapy in all patients and by tamoxifen in patients with ER+ tumors. Postoperative changes >10% (n=208) in IGF-I, IGFBP-3, IGFBP-7, or the IGF-I/IGFBP-3 ratio did not predict recurrence after adjustment for preoperative levels, age, anthropometric factors, smoking, and treatments. In conclusion, this study suggests that preoperative IGF-I and IGFBP-3 levels, but not postoperative changes, might provide independent prognostic information and influence breast cancer recurrence. The role of IGFBP-7 in breast cancer merits further study.
Collapse
Affiliation(s)
- Ann H Rosendahl
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Sofie Björner
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Maria Ygland Rödström
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Oncology and Therapeutic Pathology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden.,Department of Oncology, Aarhus University and Aarhus University Hospital, Aarhus, Denmark
| | - Christian Ingvar
- Department of Clinical Sciences Lund, Surgery, Lund University and Skåne University Hospital, Lund, Sweden
| | - Michael N Pollak
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Oncology McGill University, Montreal, QC, Canada
| | - Helena Jernström
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| |
Collapse
|
4
|
Zielinska HA, Daly CS, Alghamdi A, Bahl A, Sohail M, White P, Dean SR, Holly JMP, Perks CM. Interaction between GRP78 and IGFBP-3 Affects Tumourigenesis and Prognosis in Breast Cancer Patients. Cancers (Basel) 2020; 12:E3821. [PMID: 33352865 PMCID: PMC7767108 DOI: 10.3390/cancers12123821] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/26/2020] [Accepted: 12/15/2020] [Indexed: 01/09/2023] Open
Abstract
Insulin-like growth factor binding protein 3 (IGFBP-3) plays a key role in breast cancer progression and was recently shown to bind to the chaperone protein glucose-regulated protein 78 (GRP78); however, the clinical significance of this association remains poorly investigated. Here we report a direct correlation between the expression of GRP78 and IGFBP-3 in breast cancer cell lines and tumour sections. Kaplan-Meier survival plots revealed that patients with low GRP78 expression that are positive for IGFBP-3 had poorer survival rates than those with low IGFBP-3 levels, and we observed a similar trend in the publicly available METABRIC gene expression database. With breast cancer cells, in vitro IGFBP-3 enhanced induced apoptosis, however when GRP78 expression was silenced the actions of IGFBP-3 were switched from increasing to inhibiting ceramide (C2)-induced cell death and promoted cell invasion. Using immunofluorescence and cell surface biotinylation, we showed that knock-down of GRP78 negated the entry of IGFBP-3 into the cells. Together, our clinical and experimental results suggest that loss of GRP78 reduces IGFBP-3 entry into cells switching its actions to promote tumorigenesis and predicts a poor prognosis in breast cancer patients.
Collapse
Affiliation(s)
- Hanna A. Zielinska
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
| | - Carl S. Daly
- Faculty of Health Sciences, University of the West England, Bristol BS16 1QY, UK; (C.S.D.); (P.W.); (S.R.D.)
| | - Ahmad Alghamdi
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
- Faculty of Applied medical Sciences, Taif University, Taif, Saudi Arabia
| | - Amit Bahl
- Bristol Haematology and Oncology Centre, Department of Clinical Oncology, University Hospitals Bristol, Bristol BS2 8ED, UK;
| | - Muhammed Sohail
- Faculty of Life Sciences, School of Cellular and Molecular Medicine, Bristol University, Bristol BS8 1TD, UK;
| | - Paul White
- Faculty of Health Sciences, University of the West England, Bristol BS16 1QY, UK; (C.S.D.); (P.W.); (S.R.D.)
| | - Sarah R. Dean
- Faculty of Health Sciences, University of the West England, Bristol BS16 1QY, UK; (C.S.D.); (P.W.); (S.R.D.)
| | - Jeff M. P. Holly
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
| | - Claire M. Perks
- IGFs & Metabolic Endocrinology Group, Bristol Medical School, Translational Health Sciences, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; (H.A.Z.); (A.A.); (J.M.P.H.)
| |
Collapse
|
5
|
Piperine Inhibits TGF-β Signaling Pathways and Disrupts EMT-Related Events in Human Lung Adenocarcinoma Cells. MEDICINES 2020; 7:medicines7040019. [PMID: 32276474 PMCID: PMC7235759 DOI: 10.3390/medicines7040019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
Abstract
Background: Piperine, an amide extracted from the Piper spices, exhibits strong anti-tumor properties. However, its effect on the epithelial–mesenchymal transition (EMT) process has never been investigated. Herein, we evaluate the toxic effect of piperine on lung adenocarcinoma (A549), breast adenocarcinoma (MDA-MB-231) and hepatocellular carcinoma (HepG2) cell lines, as well as its ability to inhibit EMT-related events induced by TGF-β1 treatment. Methods: The cell viability was investigated by MTT assay. Protein expression was evaluated by Western blot. Gene expression was monitored by real-time PCR. Zymography assay was employed to detect metalloproteinase (MMP) activity in conditioned media. Cell motility was assessed by the wound-healing and phagokinetic gold sol assays. Results: The results revealed that piperine was cytotoxic in concentrations over 100 µM, showing IC50 values for HepG2, MDA-MB-231 and A549 cell lines of 214, 238 and 198 µM, respectively. In order to investigate whether piperine would reverse the TGF-β1 induced-EMT, the A549 cell line was pretreated with sublethal concentrations of the natural amide followed by the addition of TGF-β1. Besides disrupting EMT-related events, piperine also inhibited both ERK 1/2 and SMAD 2 phosphorylation. Conclusions: These results suggest that piperine might be further used in therapeutic strategies for metastatic cancer and EMT-related disorders.
Collapse
|
6
|
Lende TH, Austdal M, Varhaugvik AE, Skaland I, Gudlaugsson E, Kvaløy JT, Akslen LA, Søiland H, Janssen EAM, Baak JPA. Influence of pre-operative oral carbohydrate loading vs. standard fasting on tumor proliferation and clinical outcome in breast cancer patients ─ a randomized trial. BMC Cancer 2019; 19:1076. [PMID: 31703648 PMCID: PMC6842165 DOI: 10.1186/s12885-019-6275-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022] Open
Abstract
Background Conflicting results have been reported on the influence of carbohydrates in breast cancer. Objective To determine the influence of pre-operative per-oral carbohydrate load on proliferation in breast tumors. Design Randomized controlled trial. Setting University hospital with primary and secondary care functions in South-West Norway. Patients Sixty-one patients with operable breast cancer from a population-based cohort. Intervention Per-oral carbohydrate load (preOp™) 18 and 2–4 h before surgery (n = 26) or standard pre-operative fasting with free consumption of tap water (n = 35). Measurements The primary outcome was post-operative tumor proliferation measured by the mitotic activity index (MAI). The secondary outcomes were changes in the levels of serum insulin, insulin-c-peptide, glucose, IGF-1, and IGFBP3; patients’ well-being, and clinical outcome over a median follow-up of 88 months (range 33–97 months). Results In the estrogen receptor (ER) positive subgroup (n = 50), high proliferation (MAI ≥ 10) occurred more often in the carbohydrate group (CH) than in the fasting group (p = 0.038). The CH group was more frequently progesterone receptor (PR) negative (p = 0.014). The CH group had a significant increase in insulin (+ 24.31 mIE/L, 95% CI 15.34 mIE/L to 33.27 mIE/L) and insulin c-peptide (+ 1.39 nM, 95% CI 1.03 nM to 1.77 nM), but reduced IGFBP3 levels (− 0.26 nM; 95% CI − 0.46 nM to − 0.051 nM) compared to the fasting group. CH-intervention ER-positive patients had poorer relapse-free survival (73%) than the fasting group (100%; p = 0.012; HR = 9.3, 95% CI, 1.1 to 77.7). In the ER-positive patients, only tumor size (p = 0.021; HR = 6.07, 95% CI 1.31 to 28.03) and the CH/fasting subgrouping (p = 0.040; HR = 9.30, 95% CI 1.11 to 77.82) had independent prognostic value. The adverse clinical outcome of carbohydrate loading occurred only in T2 patients with relapse-free survival of 100% in the fasting group vs. 33% in the CH group (p = 0.015; HR = inf). The CH group reported less pain on days 5 and 6 than the control group (p < 0.001) but otherwise exhibited no factors related to well-being. Limitation Only applicable to T2 tumors in patients with ER-positive breast cancer. Conclusions Pre-operative carbohydrate load increases proliferation and PR-negativity in ER-positive patients and worsens clinical outcome in ER-positive T2 patients. Trial registration CliniTrials.gov; NCT03886389. Retrospectively registered March 22, 2019.
Collapse
Affiliation(s)
- Tone Hoel Lende
- Department of Breast & Endocrine Surgery, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway. .,Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway.
| | - Marie Austdal
- Department of Research, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Anne Elin Varhaugvik
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Pathology, Helse Møre og Romsdal HF, P.O. Box 1600, N-6026, Ålesund, Norway
| | - Ivar Skaland
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Einar Gudlaugsson
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway
| | - Jan Terje Kvaløy
- Department of Research, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Mathematics and Physics, University of Stavanger, P.O. Box 8600 Forus, N-4036, Stavanger, Norway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway.,Gades Institute, Laboratory Medicine Pathology, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway
| | - Håvard Søiland
- Department of Breast & Endocrine Surgery, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5012, Bergen, Norway
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,Department of Mathematics and Physics, University of Stavanger, P.O. Box 8600 Forus, N-4036, Stavanger, Norway
| | - Jan P A Baak
- Department of Pathology, Stavanger University Hospital, Helse Stavanger HF, P.O. Box 8100, N-4068, Stavanger, Norway.,, Risavegen 66, N-4056, Tananger, Norway.,, Vierhuysen 6, 1921 SB, Akersloot, Netherlands
| |
Collapse
|
7
|
Holly JMP, Biernacka K, Perks CM. Systemic Metabolism, Its Regulators, and Cancer: Past Mistakes and Future Potential. Front Endocrinol (Lausanne) 2019; 10:65. [PMID: 30809194 PMCID: PMC6380210 DOI: 10.3389/fendo.2019.00065] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/23/2019] [Indexed: 12/28/2022] Open
Abstract
There has been a resurgence of interest in cancer metabolism; primarily in the resetting of metabolism within malignant cells. Metabolism within cells has always been a tightly regulated process; initially in protozoans due to metabolic enzymes, and the intracellular signaling pathways that regulate these, being directly sensitive to the availability of nutrients. With the evolution of metazoans many of these controls had been overlaid by extra-cellular regulators that ensured coordinated regulation of metabolism within the community of cells that comprised the organism. Central to these systemic regulators is the insulin/insulin-like growth factor (IGF) system that throughout evolution has integrated the control of tissue growth with metabolic status. Oncological interest in the main systemic metabolic regulators greatly subsided when pharmaceutical strategies designed to treat cancers failed in the clinic. During the same period, however the explosion of new information from genetics has revealed the complexity and heterogeneity of advanced cancers and helped explain the problems of managing cancer when it reaches such a stage. Evidence has also accumulated implying that the setting of the internal environment determines whether cancers progress to advanced disease and metabolic status is clearly an important component of this local ecology. We are in the midst of an epidemic of metabolic disorders and there is considerable research into strategies for controlling metabolism. Integrating these new streams of information suggests new possibilities for cancer prevention; both primary and secondary.
Collapse
Affiliation(s)
- Jeff M. P. Holly
- Faculty of Medicine, School of Translational Health Science, University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | | | | |
Collapse
|
8
|
Negri T, Brich S, Bozzi F, Volpi CV, Gualeni AV, Stacchiotti S, De Cecco L, Canevari S, Gloghini A, Pilotti S. New transcriptional-based insights into the pathogenesis of desmoplastic small round cell tumors (DSRCTs). Oncotarget 2018; 8:32492-32504. [PMID: 28415643 PMCID: PMC5464804 DOI: 10.18632/oncotarget.16477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/13/2017] [Indexed: 12/14/2022] Open
Abstract
To gain new insights into desmoplastic small round cell tumors (DSRCTs) by means of gene expression profiling (GEP). Formalin-fixed, paraffin-embedded surgical specimens obtained from seven pretreated DSRCT patients were interrogated using GEP complemented by immunohistochemistry, a cancer stem cell array, and miRNA in situ hybridisation, including the combined chimera modules miRNA-200/ZEB1 and miRNA-34/SLUG. The chimera modules divided the cases into three classes that respectively recapitulated the traits of mesenchymal epithelial reverse transition (MErT), epithelial mesenchymal transition (EMT), and hybrid/partial EMT. This indicates a close correlation between the reprogramming governed by EMT regulators and DSRCT biology, which was further confirmed by miRNA-21 and is consistent with the broad morphological spectrum of DSRCTs. Starting from the miRNA-200/ZEB1 axis, we also found that DSRCTs carry a signature of immunological ignorance that is not responsive to PD-L1 blockade. Evidence that the up-regulation of miRNA-200 and E-cadherin, and quite a high level of miRNA-21 expression segregate with the MErT supports the idea that, in addition to the hybrid/partial state, MErT is also enriched in stemness: the androgen-positive cases, whose stemness traits were confirmed by stem cell arrays, all fell into these two classes. Our findings also confirmed that tumoral cell PDGFRA expression correlates with desmoplasia, and demonstrated the co-expression of PDGFRA and ISLR/Meflin, another marker of pluripotency. Despite the limited number of cases, these findings provide unexpectedly relevant information concerning the pathogenesis of DSRCTs, and prove the validity of miRNA-based chimera circuit modelling in the clinico-pathological setting.
Collapse
Affiliation(s)
- Tiziana Negri
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Brich
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,MOSE-DEA, University of Trieste, Trieste, Italy
| | - Fabio Bozzi
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara V Volpi
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ambra V Gualeni
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor and Rare Cancer Medical Oncology Unit, Cancer Medicine Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics and Bioinformatics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Canevari
- Department of Experimental Oncology and Molecular Medicine, Functional Genomics and Bioinformatics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Department of Diagnostic Pathology and Laboratory Medicine, Laboratory of Experimental Molecular Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
9
|
Hou LK, Yu Y, Xie YG, Wang J, Mao JF, Zhang B, Wang X, Cao XC. miR-340 and ZEB1 negative feedback loop regulates TGF-β- mediated breast cancer progression. Oncotarget 2018; 7:26016-26. [PMID: 27036021 PMCID: PMC5041961 DOI: 10.18632/oncotarget.8421] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 03/06/2016] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs act as key regulators in carcinogenesis and progression in various cancers. In present study, we explored the role of miR-340 in the breast cancer progression. Our results showed that overexpression of miR-340 inhibits breast cancer cell proliferation and invasion, whereas depletion of miR-340 promotes breast cancer progression. Molecularly, ZEB1 was identified as a target gene of miR-340 and miR-340 suppressed the expression of ZEB1 by directly binding to the 3′-UTR of ZEB1. Furthermore, ZEB1 transcriptionally suppresses miR-340 expression. The negative feedback loop regulated TGF-β-mediated breast cancer progression. In conclusion, our data suggested that miR-340 acted as a tumor suppressor in breast cancer progression.
Collapse
Affiliation(s)
- Li-Kun Hou
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Yue Yu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Ye-Gong Xie
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Jie Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Jie-Fei Mao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Bin Zhang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Xin Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| | - Xu-Chen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin 300060, China
| |
Collapse
|
10
|
Onstenk W, Sieuwerts AM, Mostert B, Lalmahomed Z, Bolt-de Vries JB, van Galen A, Smid M, Kraan J, Van M, de Weerd V, Ramírez-Moreno R, Biermann K, Verhoef C, Grünhagen DJ, IJzermans JNM, Gratama JW, Martens JWM, Foekens JA, Sleijfer S. Molecular characteristics of circulating tumor cells resemble the liver metastasis more closely than the primary tumor in metastatic colorectal cancer. Oncotarget 2018; 7:59058-59069. [PMID: 27340863 PMCID: PMC5312295 DOI: 10.18632/oncotarget.10175] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/29/2016] [Indexed: 12/21/2022] Open
Abstract
Background CTCs are a promising alternative for metastatic tissue biopsies for use in precision medicine approaches. We investigated to what extent the molecular characteristics of circulating tumor cells (CTCs) resemble the liver metastasis and/or the primary tumor from patients with metastatic colorectal cancer (mCRC). Results The CTC profiles were concordant with the liver metastasis in 17/23 patients (74%) and with the primary tumor in 13 patients (57%). The CTCs better resembled the liver metastasis in 13 patients (57%), and the primary tumor in five patients (22%). The strength of the correlations was not associated with clinical parameters. Nine genes (CDH1, CDH17, CDX1, CEACAM5, FABP1, FCGBP, IGFBP3, IGFBP4, and MAPT) displayed significant differential expressions, all of which were downregulated, in CTCs compared to the tissues in the 23 patients. Patients and Methods Patients were retrospectively selected from a prospective study. Using the CellSearch System, CTCs were enumerated and isolated just prior to liver metastasectomy. A panel of 25 CTC-specific genes was measured by RT-qPCR in matching CTCs, primary tumors, and liver metastases. Spearman correlation coefficients were calculated and considered as continuous variables with r=1 representing absolute concordance and r= -1 representing absolute discordance. A cut-off of r>0.1 was applied in order to consider profiles to be concordant. Conclusions In the majority of the patients, CTCs reflected the molecular characteristics of metastatic cells better than the primary tumors. Genes involved in cell adhesion and epithelial-to-mesenchymal transition were downregulated in the CTCs. Our results support the use of CTC characterization as a liquid biopsy for precision medicine.
Collapse
Affiliation(s)
- Wendy Onstenk
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Anieta M Sieuwerts
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Bianca Mostert
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Zarina Lalmahomed
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joan B Bolt-de Vries
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Anne van Galen
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Marcel Smid
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Jaco Kraan
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Mai Van
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Vanja de Weerd
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Raquel Ramírez-Moreno
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Katharina Biermann
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dirk J Grünhagen
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan N M IJzermans
- Department of Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jan W Gratama
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - John W M Martens
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - John A Foekens
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Erasmus MC Cancer Institute, Department of Medical Oncology and Cancer Genomics Netherlands, Rotterdam, The Netherlands
| |
Collapse
|
11
|
Daubriac J, Han S, Grahovac J, Smith E, Hosein A, Buchanan M, Basik M, Boucher Y. The crosstalk between breast carcinoma-associated fibroblasts and cancer cells promotes RhoA-dependent invasion via IGF-1 and PAI-1. Oncotarget 2017. [PMID: 29535813 PMCID: PMC5828213 DOI: 10.18632/oncotarget.23735] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Carcinoma-associated fibroblasts (CAFs) can remodel the extracellular matrix to promote cancer cell invasion, but the paracrine signaling between CAFs and cancer cells that regulates tumor cell migration remains to be identified. To determine how the interaction between CAFs and cancer cells modulates the invasiveness of cancer cells, we developed a 3-dimensional co-culture model composed of breast cancer (BC) MDA-MB-231 cell spheroids embedded in a collagen gel with and without CAFs. We found that the crosstalk between CAFs and cancer cells promotes invasion by stimulating the scattering of MDA-MB-231 cells, which was dependent on RhoA/ROCK/phospho MLC signaling in cancer cells but independent of RhoA in CAFs. The activation of RhoA/ROCK in cancer cells activates MLC and increases migration, while the genetic-down-regulation of RhoA and pharmacological inhibition of ROCK reduced cell scattering and invasion. Two distinct mechanisms induced the activation of the RhoA/ROCK pathway in MDA-MB-231 cells, the secretion of IGF-1 by CAFs and the upregulation of PAI-1 in cancer cells. In an orthotopic model of BC, IGF-1R inhibition decreased the incidence of lung metastasis, while Y27632-inhibition of ROCK enhanced the lung metastasis burden, which was associated with an increased recruitment of CAFs and expression of PAI-1. Thus the crosstalk between CAFs and BC cells increases the secretion of IGF-1 in CAFs and PAI-1 activity in cancer cells. Both IGF1 and PAI-1 activate RhoA/ROCK signaling in cancer cells, which increases cell scattering and invasion.
Collapse
Affiliation(s)
- Julien Daubriac
- Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shiwei Han
- Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jelena Grahovac
- Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eve Smith
- Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Abdel Hosein
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montreal, Canada
| | - Marguerite Buchanan
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montreal, Canada
| | - Mark Basik
- Lady Davis Institute for Medical Research, Sir Mortimer B. Davis Jewish General Hospital, McGill University, Montreal, Canada
| | - Yves Boucher
- Edwin L. Steele Laboratory for Tumor Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Oncogenic function of the homeobox A13-long noncoding RNA HOTTIP-insulin growth factor-binding protein 3 axis in human gastric cancer. Oncotarget 2017; 7:36049-36064. [PMID: 27144338 PMCID: PMC5094982 DOI: 10.18632/oncotarget.9102] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/11/2016] [Indexed: 12/11/2022] Open
Abstract
To study the mechanisms of gastric tumorigenesis, we have established CSN cell line from human normal gastric mucosa, and CS12, a tumorigenic and invasive gastric cancer cell line from CSN passages. Many stem cell markers were expressed in both CSN and CS12 cells, but LGR5 and NANOG were expressed only in CS12 cells. Increased expression of homeobox A13 (HoxA13) and its downstream cascades was significant for the tumorigenic activity of CS12 cells, and was associated with recruitment of E2F-1 to HoxA13 promoter accompanied with increased trimethylation of histone H3 lysine 4 (H3K4me3) at the hypomethylated E2F motifs. Knockdown of HoxA13 caused the downregulation of long non-coding RNA HOTTIP and insulin growth factor-binding protein 3 (IGFBP-3) genes, indicating that both were targets of HoxA13. Concurrent regulation of HoxA13-HOTTIP was mediated by the mixed lineage leukemia-WD repeat domain 5 complex, which caused the trimethylation of H3K4 and then stimulated cell proliferation. HoxA13 transactivated the IGFBP-3 promoter through the HOX-binding site. Activation of IGFBP-3 stimulated the oncogenic potential and invasion activity. Increased expression of HoxA13 (63.2%) and IGFBP-3 (28.6%) was detected in human gastric cancer tissues and was found in the gastric cancer data of The Cancer Genome Atlas. Taken together, the HoxA13–HOTTIP–IGFBP-3 cascade is critical for the carcinogenic characteristics of CS12 cells.
Collapse
|
13
|
Dong Y, Liu Y, Jiang A, Li R, Yin M, Wang Y. MicroRNA-335 suppresses the proliferation, migration, and invasion of breast cancer cells by targeting EphA4. Mol Cell Biochem 2017; 439:95-104. [PMID: 28795314 DOI: 10.1007/s11010-017-3139-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/02/2017] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that exert their functions by targeting specific mRNA sequences. Many studies have demonstrated that miRNAs are crucial for cancer progression, during which they can act as either oncogenes or tumor suppressors. Previous research has shown that miR-335 is downregulated in breast cancer, and it has been shown to be a breast cancer suppressor. In addition, emerging evidence indicates that erythropoietin-producing hepatocellular A4 (EphA4) is implicated in cancer cell proliferation, migration, and invasion. However, little is known about the relationship between miR-335 and EphA4 in breast cancer. In the present study, we used bioinformatic and biochemical analyses to demonstrate that EphA4 is a direct downstream target of miR-335 in human breast cancer MCF-7 and MDA-MB-23 cells and revealed that miR-335 negatively regulates the expression of EphA4 in these cells. Further investigation revealed that miR-335 overexpression inhibits MCF-7 and MDA-MB-231 cell proliferation and that this inhibition is attenuated by EphA4 coexpression. Similarly, miR-335 overexpression also inhibited growth and downregulated EphA4 expression in tumors in nude mice. Moreover, our results demonstrated that miR-335 overexpression suppresses migration and invasion in MCF-7 and MDA-MB-231 cells, an effect that was reversed by EphA4 overexpression. These findings confirmed that EphA4 is a direct target gene of miR-335 and that miR-335 suppresses breast cancer cell proliferation and motility in part by directly inhibiting EphA4 expression.
Collapse
Affiliation(s)
- Yilong Dong
- School of Medicine, Yunnan University, 2 Cuihu Bei Road, Kunming, 650091, Yunnan, People's Republic of China
| | - Yang Liu
- School of Medicine, Yunnan University, 2 Cuihu Bei Road, Kunming, 650091, Yunnan, People's Republic of China
| | - Aimei Jiang
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650031, Yunnan, People's Republic of China
| | - Ruiqian Li
- The Third Affiliated Hospital of Kunming Medical University, 519 Kunzhou Road, Kunming, 650031, Yunnan, People's Republic of China
| | - Min Yin
- School of Medicine, Yunnan University, 2 Cuihu Bei Road, Kunming, 650091, Yunnan, People's Republic of China
| | - Yanmei Wang
- The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming, 650031, Yunnan, People's Republic of China.
| |
Collapse
|
14
|
Morimoto-Kamata R, Yui S. Insulin-like growth factor-1 signaling is responsible for cathepsin G-induced aggregation of breast cancer MCF-7 cells. Cancer Sci 2017; 108:1574-1583. [PMID: 28544544 PMCID: PMC5543509 DOI: 10.1111/cas.13286] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 12/20/2022] Open
Abstract
Cathepsin G (CG), a neutrophil serine protease, induces cell migration and multicellular aggregation of human breast cancer MCF-7 cells in a process that is dependent on E-cadherin and CG enzymatic activity. While these tumor cell aggregates can cause tumor emboli that could represent intravascular growth and extravasation into the surrounding tissues, resulting in metastasis, the molecular mechanism underlying this process remains poorly characterized. In this study, we aimed to identify the signaling pathway that is triggered during CG-mediated stimulation of cell aggregation. Screening of a library of compounds containing approximately 90 molecular-targeting drugs revealed that this process was suppressed by the insulin-like growth factor-1 (IGF-1) receptor (IGF-1R)-specific kinase inhibitor OSI-906, as well as the multikinase inhibitors axitinib and sunitinib. Antibody array analysis, which is capable of detecting tyrosine phosphorylation of 49 distinct receptor tyrosine kinases, and the results of immunoprecipitation studies indicated that IGF-1R is phosphorylated in response to CG treatment. Notably, IGF-1R neutralization via treatment with a specific antibody or silencing of IGF-1R expression through siRNA transfection suppressed cell aggregation. Furthermore, CG treatment of MCF-7 cells resulted in increased release of IGF-1 into the medium for 24 h, while antibody-mediated IGF-1 neutralization partially prevented CG-induced cell aggregation. These results demonstrate that autocrine IGF-1 signaling is partly responsible for the cell aggregation induced by CG.
Collapse
Affiliation(s)
- Riyo Morimoto-Kamata
- Department of Pharma-Sciences, Laboratory of Host Defense, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Satoru Yui
- Department of Pharma-Sciences, Laboratory of Host Defense, Teikyo University, Itabashi-ku, Tokyo, Japan
| |
Collapse
|
15
|
Yao J, Zhang P, Li J, Xu W. MicroRNA-215 acts as a tumor suppressor in breast cancer by targeting AKT serine/threonine kinase 1. Oncol Lett 2017; 14:1097-1104. [PMID: 28693279 DOI: 10.3892/ol.2017.6200] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 02/07/2017] [Indexed: 01/15/2023] Open
Abstract
There are accumulating reports that microRNAs are dysregulated in a number of human cancer types, and that they may function as tumor suppressors or oncogenes in tumorigenesis and tumor development. microRNA-215 (miR-215) has been identified as a tumor suppressor in epithelial ovarian, pancreatic, non-small cell lung and colon cancer, whereas it may act as an oncogene in gastric and cervical cancer. The role of miR-215 in breast cancer carcinogenesis and progression has yet to be elucidated. In the present study, the expression level of miR-215 was determined in breast cancer tissues and cell lines using the reverse transcription-quantitative polymerase chain reaction. The effects of miR-215 overexpression on proliferation and the invasive capacity of breast cancer cells were assessed using MTT and cell invasion assays. The results revealed that miR-215 was significantly downregulated in breast cancer tissues and cell lines. Restoration of miR-215 expression inhibited the proliferation and invasion of breast cancer cells. The underlying molecular mechanism for the suppression of proliferation and invasion by miR-215 was investigated. AKT serine/threonine kinase 1 (AKT1) was validated as a novel direct target of miR-215, and the effect of AKT1 small interfering RNA mimicked the effect of miR-215 overexpression in breast cancer cells. These results indicated that miR-215 acted as a tumor suppressor, and that its downregulation in tumor tissues may contribute to the carcinogenesis and progression of breast cancer, indicating that miR-215 may be a novel therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jian Yao
- Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ping Zhang
- Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jin Li
- Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wei Xu
- Department of Integrated Traditional Chinese and Western Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
16
|
Metformin may protect nondiabetic breast cancer women from metastasis. Clin Exp Metastasis 2016; 33:339-57. [PMID: 26902691 DOI: 10.1007/s10585-016-9782-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 02/17/2016] [Indexed: 02/02/2023]
Abstract
Metformin, a widely prescribed oral hypoglycemic agent, has recently received a big interest because of its potential antitumorigenic effects in different cancer types. The present study investigated the impact of adding metformin to breast cancer adjuvant therapy in nondiabetic women on, insulin like growth factor-1 (IGF-1), IGF binding protein-3 (IGFBP-3), insulin, fasting blood glucose (FBG), the molar ratio of IGF-1 to IGFBP-3, homeostatic model assessment of insulin resistance (HOMA-IR) and metastasis. 102 women with newly diagnosed breast cancer were divided into 2 main groups, a control group and a metformin group. All women were treated with adjuvant therapy, according to the protocols of Ministry of Health and Population and National Cancer Institute, Egypt. Moreover, the women in the metformin group received 850 mg of metformin twice daily. Blood samples were collected at baseline, after chemotherapy (CT), after 6 months of hormonal therapy (6-HT) and 12 months of hormonal therapy (12-HT) for analysis of serum IGF-1, IGFBP-3, insulin, FBG and cancer antigen 15-3 (CA15-3). Metformin resulted in a significant reduction of IGF-1, IGF-1: IGFBP-3 molar ratio, insulin, FBG and HOMA-IR. On the other hand, metformin caused a significant increase of IGFBP-3. Moreover, metformin significantly decreased the numbers of metastatic cases after 6-HT. Metformin may have potential antitumor and antimetastatic effects that need further clinical investigations. This may be attributed to either the significant increase of the apoptotic inducer IGFBP-3 or/and the significant reduction of mitogenic insulin, IGF-1, free bioactive IGF-1, FBG and HOMA-IR.
Collapse
|
17
|
Goldhamer AC, Klaper M, Foorohar A, Myers TR. Water-only fasting and an exclusively plant foods diet in the management of stage IIIa, low-grade follicular lymphoma. BMJ Case Rep 2015; 2015:bcr-2015-211582. [PMID: 26655228 DOI: 10.1136/bcr-2015-211582] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Follicular lymphoma (FL), the second most common non-Hodgkin's lymphoma (NHL), is well characterised by a classic histological appearance and an indolent course. Current treatment protocols for FL range from close observation to immunotherapy, chemotherapy and/or radiotherapies. We report the case of a 42-year-old woman diagnosed by excisional biopsy with stage IIIa, grade 1 FL. In addition to close observation, the patient underwent a medically supervised, 21-day water-only fast after which enlarged lymph nodes were substantially reduced in size. The patient then consumed a diet of minimally processed plant foods free of added sugar, oil and salt (SOS), and has remained on the diet since leaving the residential facility. At 6 and 9-month follow-up visits, the patient's lymph nodes were non-palpable and she remained asymptomatic. This case establishes a basis for further studies evaluating water-only fasting and a plant foods, SOS-free diet as a treatment protocol for FL.
Collapse
Affiliation(s)
- Alan C Goldhamer
- Department of Nutritional Medicine, TrueNorth Health Center, Santa Rosa, California, USA
| | - Michael Klaper
- Department of Nutritional Medicine, TrueNorth Health Center, Santa Rosa, California, USA
| | - Afsoon Foorohar
- Department of Nutritional Medicine, TrueNorth Health Center, Santa Rosa, California, USA
| | - Toshia R Myers
- Department of Nutritional Medicine, TrueNorth Health Center, Santa Rosa, California, USA
| |
Collapse
|
18
|
Xue J, Chen Z, Gu X, Zhang Y, Zhang W. MicroRNA-148a inhibits migration of breast cancer cells by targeting MMP-13. Tumour Biol 2015; 37:1581-90. [PMID: 26298724 DOI: 10.1007/s13277-015-3926-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is a threat to the health of women, and metastasis of breast cancer cells plays an important role in the deterioration of breast cancer. MicroRNAs play a critical role in the tumorigenesis and development of breast cancer. MicroRNA-148a (miR-148a) is associated with the growth and metastasis of tumor cells. In the present study, we investigated the role of miR-148a in migration of breast cancer cells as well as the underlying mechanism. MiR-148a was found to inhibit the proliferation and migration of breast cancer cells. To further explore the mechanism through which miR-148a plays its antitumor role, matrix metalloproteinase-13 (MMP-13) was identified as a target of miR-148a by western blot and luciferase reporter assay. Moreover, silence of MMP-13 mimicked the effect of miR-148a, whereas overexpression of MMP-13 rescued the impaired migration caused by miR-148a. Our study demonstrates that miR-148a inhibits the migration of breast cancer cells by targeting MMP-13 and also lays theoretical foundation for further exploration for the function of miR-148a.
Collapse
Affiliation(s)
- Jinqi Xue
- The Seventh Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Zhiguang Chen
- Department of Spine and Joint Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Xi Gu
- The Seventh Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Yang Zhang
- The Seventh Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China
| | - Wenhai Zhang
- The Seventh Department of General Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
19
|
Coughlin SS, Smith SA. The Insulin-like Growth Factor Axis, Adipokines, Physical Activity, and Obesity in Relation to Breast Cancer Incidence and Recurrence. ACTA ACUST UNITED AC 2015; 4:24-31. [PMID: 26251693 PMCID: PMC4524449 DOI: 10.5539/cco.v4n2p24] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Obesity, a risk factor for the development of postmenopausal breast cancer and certain other cancer types, has also been associated with poorer response to cancer therapy and cancer recurrence. The insulin-like growth factor (IGF) axis also influences cancer risk. Methods In this commentary, we consider the literature on IGF and its binding proteins and the risk of breast cancer, along with effects of obesity, adipokines, and insulin resistance on breast cancer, and the potential for lifestyle interventions to address weight gain and physical inactivity among at-risk women. Results Greater body fatness is associated with a higher risk of postmenopausal breast cancer. The association may be explained, in part, by hyperinsulinemia and alterations in adipokines and estrogens. Nutrition, energy balance, and levels of physical activity are determinants of IGF bioactivity. Alterations in the IGF axis can increase cancer risk and progression. Results from epidemiologic studies indicate that higher circulating levels of IGF-I are associated with an increased risk of breast cancer. Conclusions Intervention studies are needed to determine how to sustain the positive effects of exercise over time and to identify the optimal mode, intensity, frequency, duration, and timing of exercise for breast cancer survivors, including important subgroups of survivors such as African American and Hispanic women. Future epidemiologic studies of the relationships between the IGF axis and breast cancer should include adequate numbers of African American women, Hispanic women, and other minority women who have been underrepresented in studies completed to date.
Collapse
Affiliation(s)
- Steven S Coughlin
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Selina A Smith
- Institute of Public and Preventive Health, and Department of Family Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|