1
|
Moustakas A. Crosstalk between TGF-β and EGF receptors via direct phosphorylation. J Cell Biol 2024; 223:e202403075. [PMID: 38506732 PMCID: PMC10955040 DOI: 10.1083/jcb.202403075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Aristidis Moustakas discusses work from Ye-Guang Chen and colleagues (https://doi.org/10.1083/jcb.202307138) on a new mechanism by which TGF-β modulates HER2 signaling in mammary epithelia.
Collapse
Affiliation(s)
- Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Moutabian H, Radi UK, Saleman AY, Adil M, Zabibah RS, Chaitanya MNL, Saadh MJ, Jawad MJ, Hazrati E, Bagheri H, Pal RS, Akhavan-Sigari R. MicroRNA-155 and cancer metastasis: Regulation of invasion, migration, and epithelial-to-mesenchymal transition. Pathol Res Pract 2023; 250:154789. [PMID: 37741138 DOI: 10.1016/j.prp.2023.154789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/25/2023]
Abstract
Among the leading causes of death globally has been cancer. Nearly 90% of all cancer-related fatalities are attributed to metastasis, which is the growing of additional malignant growths out of the original cancer origin. Therefore, a significant clinical need for a deeper comprehension of metastasis exists. Beginning investigations are being made on the function of microRNAs (miRNAs) in the metastatic process. Tiny non-coding RNAs called miRNAs have a crucial part in controlling the spread of cancer. Some miRNAs regulate migration, invasion, colonization, cancer stem cells' properties, the epithelial-mesenchymal transition (EMT), and the microenvironment, among other processes, to either promote or prevent metastasis. One of the most well-conserved and versatile miRNAs, miR-155 is primarily distinguished by overexpression in a variety of illnesses, including malignant tumors. It has been discovered that altered miR-155 expression is connected to a number of physiological and pathological processes, including metastasis. As a result, miR-155-mediated signaling pathways were identified as possible cancer molecular therapy targets. The current research on miR-155, which is important in controlling cancer cells' invasion, and metastasis as well as migration, will be summarized in the current work. The crucial significance of the lncRNA/circRNA-miR-155-mRNA network as a crucial regulator of carcinogenesis and a player in the regulation of signaling pathways or related genes implicated in cancer metastasis will be covered in the final section. These might provide light on the creation of fresh treatment plans for controlling cancer metastasis.
Collapse
Affiliation(s)
- Hossein Moutabian
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Mv N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | | | - Ebrahi Hazrati
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rashmi Saxena Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
3
|
C/EBPβ isoform-specific regulation of migration and invasion in triple-negative breast cancer cells. NPJ Breast Cancer 2022; 8:11. [PMID: 35042889 PMCID: PMC8766495 DOI: 10.1038/s41523-021-00372-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
The transcription factor C/EBPβ is a master regulator of mammary gland development and tissue remodelling during lactation. The CEBPB-mRNA is translated into three distinct protein isoforms named C/EBPβ-LAP1, -LAP2 and -LIP that are functionally different. The smaller isoform LIP lacks the N-terminal transactivation domains and is considered to act as an inhibitor of the transactivating LAP1/2 isoforms by competitive binding for the same DNA recognition sequences. Aberrantly high expression of LIP is associated with mammary epithelial proliferation and is found in grade III, estrogen receptor (ER) and progesterone (PR) receptor-negative human breast cancer. Here, we show that reverting the high LIP/LAP ratios in triple-negative breast cancer (TNBC) cell lines into low LIP/LAP ratios by overexpression of LAP reduces migration and matrix invasion of these TNBC cells. In addition, in untransformed MCF10A human mammary epithelial cells overexpression of LIP stimulates migration. Knockout of CEBPB in TNBC cells where LIP expression prevails, resulted in strongly reduced migration that was accompanied by a downregulation of genes involved in cell migration, extracellular matrix production and cytoskeletal remodelling, many of which are epithelial to mesenchymal transition (EMT) marker genes. Together, this study suggests that the LIP/LAP ratio is involved in regulating breast cancer cell migration and invasion. This study together with studies from others shows that understanding the functions the C/EBPβ-isoforms in breast cancer development may reveal new avenues of treatment.
Collapse
|
4
|
Huang G, Song C, Wang N, Qin T, Sui S, Obr A, Zeng L, Wood TL, Leroith D, Li M, Wu Y. RNA-binding protein CUGBP1 controls the differential INSR splicing in molecular subtypes of breast cancer cells and affects cell aggressiveness. Carcinogenesis 2020; 41:1294-1305. [PMID: 31958132 PMCID: PMC7513956 DOI: 10.1093/carcin/bgz141] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/18/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
The insulin receptor gene (INSR) undergoes alternative splicing to give rise to two functionally related, but also distinct, isoforms IR-A and IR-B, which dictate proliferative and metabolic regulations, respectively. Previous studies identified the RNA-binding protein CUGBP1 as a key regulator of INSR splicing. In this study, we show that the differential splicing of INSR occurs more frequently in breast cancer than in non-tumor breast tissues. In breast cancer cell lines, the IR-A:IR-B ratio varies in different molecular subtypes, knockdown or overexpression of CUGBP1 gene in breast cancer cells altered IR-A:IR-B ratio through modulation of IR-A expression, thereby reversed or enhanced the insulin-induced oncogenic behavior of breast cancer cells, respectively. Our data revealed the predominant mitogenic role of IR-A isoform in breast cancer and depicted a novel interplay between INSR and CUGBP1, implicating CUGBP1 and IR-A isoform as the potential therapeutic targets and biomarkers for breast cancer.
Collapse
Affiliation(s)
- Gena Huang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Chen Song
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ning Wang
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
| | - Tao Qin
- Department of Pathology, Dalian Medical University, Dalian, Liaoning, China
| | - Silei Sui
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Alison Obr
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Li Zeng
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Cancer Institute of New Jersey, Newark, NJ, USA
| | - Derek Leroith
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
| | - Man Li
- Department of Breast Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
- National Center of Genetically Engineered Animal Models for International Research, Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provence Key Lab of Genome Engineered Animal Models, Dalian Medical University, Dalian, Liaoning, China
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn Mount Sinai School of Medicine, New York, NY, USA
- College of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
5
|
Jaques R, Xu S, Matsakas A. Evaluating Trastuzumab in the treatment of HER2 positive breast cancer. Histol Histopathol 2020; 35:1059-1075. [PMID: 32323293 DOI: 10.14670/hh-18-221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The transmembrane oncoprotein HER2 is encoded by ERBB2 gene and overexpressed in around 20% of invasive breast cancers. It can be specifically targeted by Trastuzumab (Herceptin®), a humanised IgG1 antibody. Trastuzumab has been regarded as one of the most effective therapeutic drugs targeted to HER2 positive cancers. However, there are drawbacks, notably cardiotoxicity and resistance, which have raised awareness in clinical use. Therefore, understanding the mechanism of action is vital to establish improved therapeutic strategies. Here we evaluate Trastuzumab application in the treatment of HER2 positive breast cancer, focusing on its mechanistic actions and clinical effectiveness. Alternative therapies targeting the HER2 receptor and its downstream anomalies will also be discussed, as these could highlight further targets that could be key to improving clinical outcomes.
Collapse
Affiliation(s)
- Ryan Jaques
- Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK.
| | - Sam Xu
- Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Antonios Matsakas
- Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| |
Collapse
|
6
|
Spike AJ, Rosen JM. C/EBPß Isoform Specific Gene Regulation: It's a Lot more Complicated than you Think! J Mammary Gland Biol Neoplasia 2020; 25:1-12. [PMID: 32078094 PMCID: PMC7694698 DOI: 10.1007/s10911-020-09444-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/06/2020] [Indexed: 11/29/2022] Open
Abstract
It has been almost 30 years since C/EBPß was discovered. Seminal studies have shown that C/EBPß is a master regulator of mammary gland development and has been shown to control and influence proliferation and differentiation through varying mechanisms. The single-exon C/EBPß mRNA yields at least three different protein isoforms which have diverse, specific, context-dependent, and often non-overlapping roles throughout development and breast cancer progression. These roles are dictated by a number of complex factors including: expression levels of other C/EBP family members and their stoichiometry relative to the isoform in question, binding site affinity, post-translational modifications, co-factor expression, and even hormone levels and lactogenic status. Here we summarize the historical work up to the latest findings in the field on C/EBPß in the mammary gland and in breast cancer. With the current emphasis on improving immunotherapy in breast cancer the role of specific C/EBPß isoforms in regulating specific chemokine and cytokine expression and the immune microenvironment will be of increasing interest.
Collapse
Affiliation(s)
- Aaron J Spike
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
7
|
Yeh HW, Lee SS, Chang CY, Lang YD, Jou YS. A New Switch for TGFβ in Cancer. Cancer Res 2019; 79:3797-3805. [PMID: 31300476 DOI: 10.1158/0008-5472.can-18-2019] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 10/17/2018] [Accepted: 05/08/2019] [Indexed: 11/16/2022]
Abstract
The TGFβ cytokine plays dichotomous roles during tumor progression. In normal and premalignant cancer cells, the TGFβ signaling pathway inhibits proliferation and promotes cell-cycle arrest and apoptosis. However, the activation of this pathway in late-stage cancer cells could facilitate the epithelial-to-mesenchymal transition, stemness, and mobile features to enhance tumorigenesis and metastasis. The opposite functions of TGFβ signaling during tumor progression make it a challenging target to develop anticancer interventions. Nevertheless, the recent discovery of cellular contextual determinants, especially the binding partners of the transcription modulators Smads, is critical to switch TGFβ responses from proapoptosis to prometastasis. In this review, we summarize the recently identified contextual determinants (such as PSPC1, KLF5, 14-3-3ζ, C/EBPβ, and others) and the mechanisms of how tumor cells manage the context-dependent autonomous TGFβ responses to potentiate tumor progression. With the altered expression of some contextual determinants and their effectors during tumor progression, the aberrant molecular prometastatic switch might serve as a new class of theranostic targets for developing anticancer strategies.
Collapse
Affiliation(s)
- Hsi-Wen Yeh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Szu-Shuo Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Chieh-Yu Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Yaw-Dong Lang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuh-Shan Jou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
8
|
Ackermann T, Hartleben G, Müller C, Mastrobuoni G, Groth M, Sterken BA, Zaini MA, Youssef SA, Zuidhof HR, Krauss SR, Kortman G, de Haan G, de Bruin A, Wang ZQ, Platzer M, Kempa S, Calkhoven CF. C/EBPβ-LIP induces cancer-type metabolic reprogramming by regulating the let-7/LIN28B circuit in mice. Commun Biol 2019; 2:208. [PMID: 31240246 PMCID: PMC6572810 DOI: 10.1038/s42003-019-0461-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 05/13/2019] [Indexed: 12/18/2022] Open
Abstract
The transcription factors LAP1, LAP2 and LIP are derived from the Cebpb-mRNA through the use of alternative start codons. High LIP expression has been associated with human cancer and increased cancer incidence in mice. However, how LIP contributes to cellular transformation is poorly understood. Here we present that LIP induces aerobic glycolysis and mitochondrial respiration reminiscent of cancer metabolism. We show that LIP-induced metabolic programming is dependent on the RNA-binding protein LIN28B, a translational regulator of glycolytic and mitochondrial enzymes with known oncogenic function. LIP activates LIN28B through repression of the let-7 microRNA family that targets the Lin28b-mRNA. Transgenic mice overexpressing LIP have reduced levels of let-7 and increased LIN28B expression, which is associated with metabolic reprogramming as shown in primary bone marrow cells, and with hyperplasia in the skin. This study establishes LIP as an inducer of cancer-type metabolic reprogramming and as a regulator of the let-7/LIN28B regulatory circuit.
Collapse
Affiliation(s)
- Tobias Ackermann
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Götz Hartleben
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Christine Müller
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | | | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Britt A. Sterken
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Mohamad A. Zaini
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Sameh A. Youssef
- Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, NL-3584 CL Utrecht, the Netherlands
| | - Hidde R. Zuidhof
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Sara R. Krauss
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Gertrud Kortman
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Gerald de Haan
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Alain de Bruin
- Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, NL-3584 CL Utrecht, the Netherlands
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Matthias Platzer
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, D-07745 Jena, Germany
| | - Stefan Kempa
- Max Delbrück Center for Molecular Medicine, D-13092 Berlin, Germany
| | - Cornelis F. Calkhoven
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| |
Collapse
|
9
|
Adamo H, Hammarsten P, Hägglöf C, Dahl Scherdin T, Egevad L, Stattin P, Halin Bergström S, Bergh A. Prostate cancer induces C/EBPβ expression in surrounding epithelial cells which relates to tumor aggressiveness and patient outcome. Prostate 2019; 79:435-445. [PMID: 30536410 DOI: 10.1002/pros.23749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/08/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Implantation of rat prostate cancer cells into the normal rat prostate results in tumor-stimulating adaptations in the tumor-bearing organ. Similar changes are seen in prostate cancer patients and they are related to outcome. One gene previously found to be upregulated in the non-malignant part of tumor-bearing prostate lobe in rats was the transcription factor CCAAT/enhancer-binding protein-β (C/EBPβ). METHODS To explore this further, we examined C/EBPβ expression by quantitative RT-PCR, immunohistochemistry, and Western blot in normal rat prostate tissue surrounding slow-growing non-metastatic Dunning G, rapidly growing poorly metastatic (AT-1), and rapidly growing highly metastatic (MatLyLu) rat prostate tumors-and also by immunohistochemistry in a tissue microarray (TMA) from prostate cancer patients managed by watchful waiting. RESULTS In rats, C/EBPβ mRNA expression was upregulated in the surrounding tumor-bearing prostate lobe. In tumors and in the surrounding non-malignant prostate tissue, C/EBPβ was detected by immunohistochemistry in some epithelial cells and in infiltrating macrophages. The magnitude of glandular epithelial C/EBPβ expression in the tumor-bearing prostates was associated with tumor size, distance to the tumor, and metastatic capacity. In prostate cancer patients, high expression of C/EBPβ in glandular epithelial cells in the surrounding tumor-bearing tissue was associated with accumulation of M1 macrophages (iNOS+) and favorable outcome. High expression of C/EBPβ in tumor epithelial cells was associated with high Gleason score, high tumor cell proliferation, metastases, and poor outcome. CONCLUSIONS This study suggest that the expression of C/EBP-beta, a transcription factor mediating multiple biological effects, is differentially expressed both in the benign parts of the tumor-bearing prostate and in prostate tumors, and that alterations in this may be related to patient outcome.
Collapse
Affiliation(s)
- Hanibal Adamo
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Peter Hammarsten
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Christina Hägglöf
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Tove Dahl Scherdin
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Pär Stattin
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| |
Collapse
|
10
|
ALK-mediated Tyr95 phosphorylation of Smad4 impairs its transcription activity and the tumor suppressive activity of TGF-β. SCIENCE CHINA-LIFE SCIENCES 2019; 62:431-432. [DOI: 10.1007/s11427-019-9506-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 12/28/2022]
|
11
|
Shi Q, Chen YG. The functional switch of TGF-β signaling in breast cancer. Oncotarget 2019; 10:1604-1605. [PMID: 30899430 PMCID: PMC6422195 DOI: 10.18632/oncotarget.26715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/14/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Qiaoni Shi
- Ye-Guang Chen: The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ye-Guang Chen
- Ye-Guang Chen: The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Huang F, Shi Q, Li Y, Xu L, Xu C, Chen F, Wang H, Liao H, Chang Z, Liu F, Zhang XHF, Feng XH, Han JDJ, Luo S, Chen YG. HER2/EGFR-AKT Signaling Switches TGFβ from Inhibiting Cell Proliferation to Promoting Cell Migration in Breast Cancer. Cancer Res 2018; 78:6073-6085. [PMID: 30171053 DOI: 10.1158/0008-5472.can-18-0136] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/15/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022]
Abstract
TGFβ signaling inhibits cell proliferation to block cancer initiation, yet it also enhances metastasis to promote malignancy during breast cancer development. The mechanisms underlying these differential effects are still unclear. Here, we report that HER2/EGFR signaling switches TGFβ function in breast cancer cells from antiproliferation to cancer promotion. Inhibition of HER2/EGFR activity attenuated TGFβ-induced epithelial-mesenchymal transition and migration but enhanced the antiproliferative activity of TGFβ. Activation of HER2/EGFR induced phosphorylation of Smad3 at Ser208 of the linker region through AKT, which promoted the nuclear accumulation of Smad3 and subsequent expression of the genes related to EMT and cell migration. In contrast, HER2/EGFR signaling had no effects on the nuclear localization of Smad2. Knockdown of Smad3, but not Smad2, blocked TGFβ-induced breast cancer cell migration. We observed a positive correlation between the nuclear localization of Smad3 and HER2 activation in advanced human breast cancers. Our results demonstrate a key role for HER2/EGFR in differential regulation of Smad3 activity to shift TGFβ function from antitumorigenic to protumorigenic during breast cancer development.Significance: TGFβ signaling can shift from inhibiting to promoting breast cancer development via HER2/EGFR AKT-mediated phosphorylation of Smad3 at S208, enhancing its nuclear accumulation and upregulation of EMT-related genes.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/21/6073/F1.large.jpg Cancer Res; 78(21); 6073-85. ©2018 AACR.
Collapse
Affiliation(s)
- Fei Huang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qiaoni Shi
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuzhen Li
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Linlin Xu
- The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Chi Xu
- Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fenfang Chen
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Hongwei Liao
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zai Chang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Fang Liu
- Center for Advanced Biotechnology and Medicine, Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing-Dong J Han
- Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shiwen Luo
- The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
13
|
Müller C, Zidek LM, Ackermann T, de Jong T, Liu P, Kliche V, Zaini MA, Kortman G, Harkema L, Verbeek DS, Tuckermann JP, von Maltzahn J, de Bruin A, Guryev V, Wang ZQ, Calkhoven CF. Reduced expression of C/EBPβ-LIP extends health and lifespan in mice. eLife 2018; 7:34985. [PMID: 29708496 PMCID: PMC5986274 DOI: 10.7554/elife.34985] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 04/27/2018] [Indexed: 02/06/2023] Open
Abstract
Ageing is associated with physical decline and the development of age-related diseases such as metabolic disorders and cancer. Few conditions are known that attenuate the adverse effects of ageing, including calorie restriction (CR) and reduced signalling through the mechanistic target of rapamycin complex 1 (mTORC1) pathway. Synthesis of the metabolic transcription factor C/EBPβ-LIP is stimulated by mTORC1, which critically depends on a short upstream open reading frame (uORF) in the Cebpb-mRNA. Here, we describe that reduced C/EBPβ-LIP expression due to genetic ablation of the uORF delays the development of age-associated phenotypes in mice. Moreover, female C/EBPβΔuORF mice display an extended lifespan. Since LIP levels increase upon aging in wild type mice, our data reveal an important role for C/EBPβ in the aging process and suggest that restriction of LIP expression sustains health and fitness. Thus, therapeutic strategies targeting C/EBPβ-LIP may offer new possibilities to treat age-related diseases and to prolong healthspan. The risks of major diseases including type II diabetes, cancer and Alzheimer’s are linked to the biological process of ageing. By finding ways to slow ageing, we can help more people to live longer healthier lives while avoiding these illnesses. Placing some animals on a diet that contains only two-thirds as many calories as they would normally eat can improve their fitness during old age and delay the onset of many age-related problems. It is unrealistic to expect people to control their diet to this extent, yet there may be other ways to bring about the same effects. Calorie restriction affects the activity of many different genes; for example, it causes a gene that produces a protein known as Liver-enriched Inhibitory Protein (LIP for short) to shut down. LIP controls the activity of many genes involved in metabolism, so it could be a key target for drugs to control ageing. Müller, Zidek et al. used mice that are unable to produce LIP to study this protein’s effect on ageing. The life expectancy of female mice lacking LIP increased by up to 20%. These mice were leaner, fitter, more resistant to cancer, had stronger immune systems and controlled their blood sugar levels better than normal mice. Male mice that lacked LIP did not live longer but did experience some ageing-related benefits. Genetic analysis also showed that gene activity particularly of metabolic genes is more robust in old female LIP-deficient mice and thus more similar to young control mice than old control mice. The results presented by Müller, Zidek et al. suggest that targeting the activity of the LIP gene could help to slow the ageing process. It is not yet clear whether shutting off LIP has similar beneficial effects in humans. Further research is also needed to investigate why female mice gain more benefits from a lack of LIP than males do.
Collapse
Affiliation(s)
- Christine Müller
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands.,Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Laura M Zidek
- Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Tobias Ackermann
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Tristan de Jong
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Peng Liu
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Verena Kliche
- Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Mohamad Amr Zaini
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Gertrud Kortman
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Liesbeth Harkema
- Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Dineke S Verbeek
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan P Tuckermann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | | | - Alain de Bruin
- Dutch Molecular Pathology Centre, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Cornelis F Calkhoven
- European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands.,Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| |
Collapse
|
14
|
Cifdaloz M, Osterloh L, Graña O, Riveiro-Falkenbach E, Ximénez-Embún P, Muñoz J, Tejedo C, Calvo TG, Karras P, Olmeda D, Miñana B, Gómez-López G, Cañon E, Eyras E, Guo H, Kappes F, Ortiz-Romero PL, Rodríguez-Peralto JL, Megías D, Valcárcel J, Soengas MS. Systems analysis identifies melanoma-enriched pro-oncogenic networks controlled by the RNA binding protein CELF1. Nat Commun 2017; 8:2249. [PMID: 29269732 PMCID: PMC5740069 DOI: 10.1038/s41467-017-02353-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 11/23/2017] [Indexed: 12/19/2022] Open
Abstract
Melanomas are well-known for their altered mRNA expression profiles. Yet, the specific contribution of mRNA binding proteins (mRBPs) to melanoma development remains unclear. Here we identify a cluster of melanoma-enriched genes under the control of CUGBP Elav-like family member 1 (CELF1). CELF1 was discovered with a distinct prognostic value in melanoma after mining the genomic landscape of the 692 known mRBPs across different cancer types. Genome-wide transcriptomic, proteomic, and RNA-immunoprecipitation studies, together with loss-of-function analyses in cell lines, and histopathological evaluation in clinical biopsies, revealed an intricate repertoire of CELF1-RNA interactors with minimal overlap with other malignancies. This systems approach uncovered the oncogene DEK as an unexpected target and downstream effector of CELF1. Importantly, CELF1 and DEK were found to represent early-induced melanoma genes and adverse indicators of overall patient survival. These results underscore novel roles of CELF1 in melanoma, illustrating tumor type-restricted functions of RBPs in cancer.
Collapse
Affiliation(s)
- Metehan Cifdaloz
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - Lisa Osterloh
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | | | - Erica Riveiro-Falkenbach
- Instituto de Investigación i+12, Hospital 12 de Octubre Medical School, Universidad Complutense, 28041, Madrid, Spain
| | | | | | - Cristina Tejedo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - Tonantzin G Calvo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - Panagiotis Karras
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - Belén Miñana
- Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
| | | | - Estela Cañon
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain
| | - Eduardo Eyras
- Department of Experimental and Health Sciences, Universidad Pompeu Fabra, Barcelona, 08002, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08010, Spain
| | - Haihong Guo
- Institute of Biochemistry and Molecular Biology; Medical School, RWTH Aachen University, Aachen, 52074, Germany
| | - Ferdinand Kappes
- Institute of Biochemistry and Molecular Biology; Medical School, RWTH Aachen University, Aachen, 52074, Germany
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, No. 111, Ren Ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park (SIP), Suzhou, 215123, China
| | - Pablo L Ortiz-Romero
- Instituto de Investigación i+12, Hospital 12 de Octubre Medical School, Universidad Complutense, 28041, Madrid, Spain
| | - Jose L Rodríguez-Peralto
- Instituto de Investigación i+12, Hospital 12 de Octubre Medical School, Universidad Complutense, 28041, Madrid, Spain
| | - Diego Megías
- Confocal Microscopy Unit, (CNIO), Madrid, 28029, Spain
| | - Juan Valcárcel
- Centre de Regulació Genòmica (CRG), The Barcelona Institute of Science and Technology, Barcelona, 08003, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08010, Spain
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), 28029, Madrid, Spain.
| |
Collapse
|
15
|
Seoane J, Gomis RR. TGF-β Family Signaling in Tumor Suppression and Cancer Progression. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022277. [PMID: 28246180 DOI: 10.1101/cshperspect.a022277] [Citation(s) in RCA: 365] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) induces a pleiotropic pathway that is modulated by the cellular context and its integration with other signaling pathways. In cancer, the pleiotropic reaction to TGF-β leads to a diverse and varied set of gene responses that range from cytostatic and apoptotic tumor-suppressive ones in early stage tumors, to proliferative, invasive, angiogenic, and oncogenic ones in advanced cancer. Here, we review the knowledge accumulated about the molecular mechanisms involved in the dual response to TGF-β in cancer, and how tumor cells evolve to evade the tumor-suppressive responses of this signaling pathway and then hijack the signal, converting it into an oncogenic factor. Only through the detailed study of this complexity can the suitability of the TGF-β pathway as a therapeutic target against cancer be evaluated.
Collapse
Affiliation(s)
- Joan Seoane
- Translational Research Program, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Roger R Gomis
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.,Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
16
|
Interplay between TGF-β signaling and receptor tyrosine kinases in tumor development. SCIENCE CHINA-LIFE SCIENCES 2017; 60:1133-1141. [DOI: 10.1007/s11427-017-9173-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 09/13/2017] [Indexed: 12/12/2022]
|
17
|
Vlasova-St Louis I, Bohjanen PR. Post-transcriptional regulation of cytokine and growth factor signaling in cancer. Cytokine Growth Factor Rev 2016; 33:83-93. [PMID: 27956133 DOI: 10.1016/j.cytogfr.2016.11.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/28/2016] [Indexed: 12/11/2022]
Abstract
Cytokines and growth factors regulate cell proliferation, differentiation, migration and apoptosis, and play important roles in coordinating growth signal responses during development. The expression of cytokine genes and the signals transmitted through cytokine receptors are tightly regulated at several levels, including transcriptional and post-transcriptional levels. A majority of cytokine mRNAs, including growth factor transcripts, contain AU-rich elements (AREs) in their 3' untranslated regions that control gene expression by regulating mRNA degradation and changing translational rates. In addition, numerous proteins involved in transmitting signals downstream of cytokine receptors are regulated at the level of mRNA degradation by GU-rich elements (GREs) found in their 3' untranslated regions. Abnormal stabilization and overexpression of ARE or GRE-containing transcripts had been observed in many malignancies, which is a consequence of the malfunction of RNA-binding proteins. In this review, we briefly summarize the role of AREs and GREs in regulating mRNA turnover to coordinate cytokine and growth factor expression, and we describe how dysregulation of mRNA degradation mechanisms contributes to the development and progression of cancer.
Collapse
Affiliation(s)
| | - Paul R Bohjanen
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA; Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
18
|
Bohjanen PR, Moua ML, Guo L, Taye A, Vlasova-St Louis IA. Altered CELF1 binding to target transcripts in malignant T cells. RNA (NEW YORK, N.Y.) 2015; 21:1757-1769. [PMID: 26249002 PMCID: PMC4574752 DOI: 10.1261/rna.049940.115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/29/2015] [Indexed: 06/04/2023]
Abstract
The RNA-binding protein, CELF1, binds to a regulatory sequence known as the GU-rich element (GRE) and controls a network of mRNA transcripts that regulate cellular activation, proliferation, and apoptosis. We performed immunoprecipitation using an anti-CELF1 antibody, followed by identification of copurified transcripts using microarrays. We found that CELF1 is bound to a distinct set of target transcripts in the H9 and Jurkat malignant T-cell lines, compared with primary human T cells. CELF1 was not phosphorylated in resting normal T cells, but in malignant T cells, phosphorylation of CELF1 correlated with its inability to bind to GRE-containing mRNAs that served as CELF1 targets in normal T cells. Lack of binding by CELF1 to these mRNAs in malignant T cells correlated with stabilization and increased expression of these transcripts. Several of these GRE-containing transcripts that encode regulators of cell growth were also stabilized and up-regulated in primary tumor cells from patients with T-cell acute lymphoblastic leukemia. Interestingly, transcripts encoding numerous suppressors of cell proliferation that served as targets of CELF1 in malignant T cells, but not normal T cells, exhibited accelerated degradation and reduced expression in malignant compared with normal T cells, consistent with the known function of CELF1 to mediate degradation of bound transcripts. Overall, CELF1 dysfunction in malignant T cells led to the up-regulation of a subset of GRE-containing transcripts that promote cell growth and down-regulation of another subset that suppress cell growth, producing a net effect that would drive a malignant phenotype.
Collapse
Affiliation(s)
- Paul R Bohjanen
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA Department of Microbiology, University of Minnesota, Minneapolis, Minnesota 55455, USA Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Mai Lee Moua
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Liang Guo
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Ammanuel Taye
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Irina A Vlasova-St Louis
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, USA Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
19
|
Pavlovic M, Arnal-Estapé A, Rojo F, Bellmunt A, Tarragona M, Guiu M, Planet E, Garcia-Albéniz X, Morales M, Urosevic J, Gawrzak S, Rovira A, Prat A, Nonell L, Lluch A, Jean-Mairet J, Coleman R, Albanell J, Gomis RR. Enhanced MAF Oncogene Expression and Breast Cancer Bone Metastasis. J Natl Cancer Inst 2015; 107:djv256. [PMID: 26376684 PMCID: PMC4681582 DOI: 10.1093/jnci/djv256] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There are currently no biomarkers for early breast cancer patient populations at risk of bone metastasis. Identification of mediators of bone metastasis could be of clinical interest. METHODS A de novo unbiased screening approach based on selection of highly bone metastatic breast cancer cells in vivo was used to determine copy number aberrations (CNAs) associated with bone metastasis. The CNAs associated with bone metastasis were examined in independent primary breast cancer datasets with annotated clinical follow-up. The MAF gene encoded within the CNA associated with bone metastasis was subjected to gain and loss of function validation in breast cancer cells (MCF7, T47D, ZR-75, and 4T1), its downstream mechanism validated, and tested in clinical samples. A multivariable Cox cause-specific hazard model with competing events (death) was used to test the association between 16q23 or MAF and bone metastasis. All statistical tests were two-sided. RESULTS 16q23 gain CNA encoding the transcription factor MAF mediates breast cancer bone metastasis through the control of PTHrP. 16q23 gain (hazard ratio (HR) for bone metastasis = 14.5, 95% confidence interval (CI) = 6.4 to 32.9, P < .001) as well as MAF overexpression (HR for bone metastasis = 2.5, 95% CI = 1.7 to 3.8, P < .001) in primary breast tumors were specifically associated with risk of metastasis to bone but not to other organs. CONCLUSIONS These results suggest that MAF is a mediator of breast cancer bone metastasis. 16q23 gain or MAF protein overexpression in tumors may help to select patients at risk of bone relapse.
Collapse
Affiliation(s)
- Milica Pavlovic
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Anna Arnal-Estapé
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Federico Rojo
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Anna Bellmunt
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Maria Tarragona
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Marc Guiu
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Evarist Planet
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Xabier Garcia-Albéniz
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Mónica Morales
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Jelena Urosevic
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Sylwia Gawrzak
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Ana Rovira
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Aleix Prat
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Lara Nonell
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Ana Lluch
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Joël Jean-Mairet
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Robert Coleman
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Joan Albanell
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG)
| | - Roger R Gomis
- Oncology Program (MP, AAE, AB, MT, MG, XGA, MM, JU, SG, RRG) and Biostatistics and Bioinformatics Unit (EP), Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain; Cancer Research Program (FR, AR, JA) and Microarray Analysis Service (LN), IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain; Pathology Department, IIS-Fundación Jimenez Diaz, Madrid, Spain (FR); Medical Oncology Service, Hospital del Mar, Barcelona, Spain (AR, JA); Department of Oncology and Hematology, Hospital Clínico Universitario, Valencia, Spain (AL); Valencia Central University, Spain (AL); Inbiomotion, Barcelona, Spain (JJM); Sheffield Cancer Research Centre, Sheffield, UK (RC); Universitat Pompeu Fabra, Barcelona, Spain (JA); Translational Genomics, Vall d'Hebron Insitute of Oncology, Barcelona, Spain (AP); Department of Epidemiology, Harvard School of Public Health, Boston, MA (XGA); Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain (RRG).
| |
Collapse
|
20
|
Wang X, Jiao W, Zhao Y, Zhang L, Yao R, Wang Y, Wang M, Luo Y, Zhao J. CUG-binding protein 1 (CUGBP1) expression and prognosis of brain metastases from non-small cell lung cancer. Thorac Cancer 2015; 7:32-8. [PMID: 26816536 PMCID: PMC4718132 DOI: 10.1111/1759-7714.12268] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 03/29/2015] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The brain is a frequent site of metastases from non-small cell lung cancer (NSCLC). The purpose of this study was to detect the expression of CUG-binding protein 1 (CUGBP1) messenger ribonucleic acid (mRNA) and Ki-67 in metastasized brain tissue from NSCLC and determine the relationship between CUGBP1 and brain metastases. METHODS The expression of CUGBP1 mRNA and Ki-67 in metastasized brain tissue from NSCLC was investigated by semiquantitative polymerase chain reaction and immunohistochemistry, respectively. The expression of CUGBP1 and Ki-67 in metastasized brain tissue from NSCLC was related to clinical characteristics, as assessed using the chi-square test. The prognostic significance was assessed by univariate and multivariate analyses using the Cox hazard model. RESULTS The expression of CUGBP1 mRNA and Ki-67 was overexpressed in metastasized brain tissue from NSCLC and was correlated with differentiation. In addition, by both univariate and multivariate survival analyses, CUGBP1 expression, Ki-67 expression, and age were noted to be independent indicators of a shorter postsurgical survival. CONCLUSION The expression of CUGBP1 is an important factor in the development of brain metastases from NSCLC.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Wenjie Jiao
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Yandong Zhao
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Liangdong Zhang
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Ruyong Yao
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Yongjie Wang
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Mingzhao Wang
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Yiren Luo
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| | - Jinpeng Zhao
- Department of Thoracic Surgery The Affiliated Hospital of Qingdao University Qingdao China
| |
Collapse
|
21
|
Perino A, Pols TWH, Nomura M, Stein S, Pellicciari R, Schoonjans K. TGR5 reduces macrophage migration through mTOR-induced C/EBPβ differential translation. J Clin Invest 2014; 124:5424-36. [PMID: 25365223 DOI: 10.1172/jci76289] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
The bile acid-responsive G protein-coupled receptor TGR5 is involved in several metabolic processes, and recent studies suggest that TGR5 activation may promote pathways that are protective against diet-induced diabetes. Here, we investigated the role of macrophage-specific TGR5 signaling in protecting adipose tissue from inflammation and associated insulin resistance. Examination of adipose tissue from obese mice lacking macrophage Tgr5 revealed enhanced inflammation, increased chemokine expression, and higher macrophage numbers compared with control obese animals. Moreover, macrophage-specific deletion of Tgr5 exacerbated insulin resistance in obese animals. Conversely, pharmacological activation of TGR5 markedly decreased LPS-induced chemokine expression in primary macrophages. This reduction was mediated by AKT-dependent activation of mTOR complex 1, which in turn induced the differential translation of the dominant-negative C/EBPβ isoform, liver inhibitory protein (LIP). Overall, these studies reveal a signaling pathway downstream of TGR5 that modulates chemokine expression in response to high-fat diet and suggest that targeting this pathway has the potential to be therapeutically exploited for prevention of chronic inflammatory diseases and type 2 diabetes mellitus.
Collapse
|
22
|
Miura Y, Hagiwara N, Radisky DC, Hirai Y. CCAAT/enhancer binding protein beta (C/EBPβ) isoform balance as a regulator of epithelial-mesenchymal transition in mouse mammary epithelial cells. Exp Cell Res 2014; 327:146-55. [PMID: 24881817 DOI: 10.1016/j.yexcr.2014.05.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/03/2014] [Accepted: 05/21/2014] [Indexed: 12/22/2022]
Abstract
Activation of the epithelial-mesenchymal transition (EMT) program promotes cell invasion and metastasis, and is reversed through mesenchymal-epithelial transition (MET) after formation of distant metastases. Here, we show that an imbalance of gene products encoded by the transcriptional factor C/EBPβ, LAP (liver-enriched activating protein) and LIP (liver-enriched inhibitory protein), can regulate both EMT- and MET-like phenotypic changes in mouse mammary epithelial cells. By using tetracycline repressive LIP expression constructs, we found that SCp2 cells, a clonal epithelial line of COMMA1-D cells, expressed EMT markers, lost the ability to undergo alveolar-like morphogenesis in 3D Matrigel, and acquired properties of benign adenoma cells. Conversely, we found that inducible expression of LAP in SCg6 cells, a clonal fibroblastic line of COMMA1-D cells, began to express epithelial keratins with suppression of proliferation. The overexpression of the C/EBPβ gene products in these COMMA1-D derivatives was suppressed by long-term cultivation on tissue culture plastic, but gene expression was maintained in cells grown on Matrigel or exposed to proteasome inhibitors. Thus, imbalances of C/EBPβ gene products in mouse mammary epithelial cells, which are affected by contact with basement membrane, are defined as a potential regulator of metastatic potential.
Collapse
Affiliation(s)
- Yuka Miura
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan
| | - Natsumi Hagiwara
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32225 USA
| | - Yohei Hirai
- Department of Bioscience, Graduate School of Science and Technology, Kwansei Gakuin University, Hyogo, 2-1 Gakuen, Sanda 669-1337 Japan.
| |
Collapse
|
23
|
Park BH, Kook S, Lee S, Jeong JH, Brufsky A, Lee BC. An isoform of C/EBPβ, LIP, regulates expression of the chemokine receptor CXCR4 and modulates breast cancer cell migration. J Biol Chem 2013; 288:28656-67. [PMID: 23966000 DOI: 10.1074/jbc.m113.509505] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Metastasis is the primary cause of death in cancer patients. CXCR4/CXCL12 chemokine axis provides directional cues for breast cancer cells to metastasize to specific organs. Despite their potential clinical importance, how CXCR4 expression in breast cancer cells is regulated at the molecular level is not well understood. We identified an isoform of C/EBPβ, liver-enriched inhibitory protein (LIP), as a previously unrecognized transcriptional regulator of CXCR4 in breast cancer cells. LIP up-regulated the transcription of CXCR4 through direct interaction with the CXCR4 promoter. The increase in CXCR4 mRNA was paralleled by an increased cell surface expression of the CXCR4, which in turn promoted CXCR4-mediated breast cancer cell migration. A significant positive correlation between LIP and CXCR4 expression was observed in stage III and IV human breast carcinoma specimens. Neuregulin 1 (or NRG1, hereafter referred to as heregulin) increased CXCR4 expression in breast cancer cells, and this coincided with increased LIP binding on the CXCR4 promoter. These findings may have important implications for understanding the molecular basis of CXCR4-mediated breast cancer cell metastasis and could potentially allow us to develop novel strategies to reduce morbidity and mortality in patients with metastatic breast cancer.
Collapse
Affiliation(s)
- Bae-Hang Park
- From the University of Pittsburgh Cancer Institute, Department of Medicine, Division of Hematology and Oncology, Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | | | | | | | | | | |
Collapse
|
24
|
Johansson J, Berg T, Kurzejamska E, Pang MF, Tabor V, Jansson M, Roswall P, Pietras K, Sund M, Religa P, Fuxe J. MiR-155-mediated loss of C/EBPβ shifts the TGF-β response from growth inhibition to epithelial-mesenchymal transition, invasion and metastasis in breast cancer. Oncogene 2013; 32:5614-24. [PMID: 23955085 PMCID: PMC3898103 DOI: 10.1038/onc.2013.322] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 06/01/2013] [Accepted: 06/24/2013] [Indexed: 12/19/2022]
Abstract
During breast cancer progression, transforming growth factor-beta (TGF-β) switches from acting as a growth inhibitor to become a major promoter of epithelial-mesenchymal transition (EMT), invasion and metastasis. However, the mechanisms involved in this switch are not clear. We found that loss of CCAAT-enhancer binding protein beta (C/EBPβ), a differentiation factor for the mammary epithelium, was associated with signs of EMT in triple-negative human breast cancer, and in invasive areas of mammary tumors in MMTV-PyMT mice. Using an established model of TGF-β-induced EMT in mouse mammary gland epithelial cells, we discovered that C/EBPβ was repressed during EMT by miR-155, an oncomiR in breast cancer. Depletion of C/EBPβ potentiated the TGF-β response towards EMT, and contributed to evasion of the growth inhibitory response to TGF-β. Furthermore, loss of C/EBPβ enhanced invasion and metastatic dissemination of the mouse mammary tumor cells to the lungs after subcutaneous injection into mice. The mechanism by which loss of C/EBPβ promoted the TGF-β response towards EMT, invasion and metastasis, was traced to a previously uncharacterized role of C/EBPβ as a transcriptional activator of genes encoding the epithelial junction proteins E-cadherin and coxsackie virus and adenovirus receptor. The results identify miR-155-mediated loss of C/EBPβ as a mechanism, which promotes breast cancer progression by shifting the TGF-β response from growth inhibition to EMT, invasion and metastasis.
Collapse
Affiliation(s)
- J Johansson
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - T Berg
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - E Kurzejamska
- 1] Department of Medicine, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden [2] Department of Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - M-F Pang
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - V Tabor
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - M Jansson
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - P Roswall
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - K Pietras
- 1] Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden [2] Department of Laboratory Medicine Malmö, Lund University Cancer Center, Lund University, Malmö, Sweden
| | - M Sund
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umeå, Sweden
| | - P Religa
- Department of Medicine, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - J Fuxe
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
25
|
Jiao W, Zhao J, Wang M, Wang Y, Luo Y, Zhao Y, Tang D, Shen Y. CUG-binding protein 1 (CUGBP1) expression and prognosis of non-small cell lung cancer. Clin Transl Oncol 2013; 15:789-95. [PMID: 23359188 DOI: 10.1007/s12094-013-1005-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 01/13/2013] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Non-small cell lung cancer (NSCLC) is the leading cause of cancer mortality worldwide. As CUGBP1 may also play a great role in tumor genesis and deterioration, the purpose of this study was to detect the expression of CUGBP1 mRNA and CUGBP1 and assess the prognostic significance of CUGBP1 in NSCLC. METHODS Expression of CUGBP1 mRNA and CUGBP was detected by Semi-quantitative PCR and Immunohistochemistry, respectively, from 57 NSCLC patients. The percentage of CUGBP1 mRNA and CUGBP1 expression was correlated with clinical characteristics using χ (2) test. The prognostic significance was assessed by univariate and multivariate analyses in the Cox hazard model. RESULTS The expression of CUGBP1 mRNA and CUGBP1 was over-expressed in cancer group and was correlated with TNM stage and Differentiation. By both univariate and multivariate survival analyses, CUGBP1 expression (P = 0.0074, HR = 3.701, 95 % CI 1.420-9.648), TNM-stage (HR = 4.043, 95 % CI 2.098-7.794) and age (HR = 3.207, 95 % CI 1.544-6.664) were noted to be independent indicators of a shorter postsurgical survival. CONCLUSIONS The expression of CUGBP1 independently predicted a shorter postsurgical survival in NSCLC.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/mortality
- Adenocarcinoma/surgery
- Adult
- Aged
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- CELF1 Protein
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/surgery
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/surgery
- Cell Differentiation
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Immunoenzyme Techniques
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/surgery
- Male
- Middle Aged
- Neoplasm Staging
- Prognosis
- RNA, Messenger/genetics
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
Collapse
Affiliation(s)
- W Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Medical College, Qingdao University, 16 Jiangsu Road, Qingdao, 266003, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Scott JM, Lakoski S, Mackey JR, Douglas PS, Haykowsky MJ, Jones LW. The potential role of aerobic exercise to modulate cardiotoxicity of molecularly targeted cancer therapeutics. Oncologist 2013; 18:221-31. [PMID: 23335619 PMCID: PMC3579607 DOI: 10.1634/theoncologist.2012-0226] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 09/05/2012] [Indexed: 01/03/2023] Open
Abstract
Molecularly targeted therapeutics (MTT) are the future of cancer systemic therapy. They have already moved from palliative therapy for advanced solid malignancies into the setting of curative-intent treatment for early-stage disease. Cardiotoxicity is a frequent and potentially serious adverse complication of some targeted therapies, leading to a broad range of potentially life-threatening complications, therapy discontinuation, and poor quality of life. Low-cost pleiotropic interventions are therefore urgently required to effectively prevent and/or treat MTT-induced cardiotoxicity. Aerobic exercise therapy has the unique capacity to modulate, without toxicity, multiple gene expression pathways in several organ systems, including a plethora of cardiac-specific molecular and cell-signaling pathways implicated in MTT-induced cardiac toxicity. In this review, we examine the molecular signaling of antiangiogenic and HER2-directed therapies that may underpin cardiac toxicity and the hypothesized molecular mechanisms underlying the cardioprotective properties of aerobic exercise. It is hoped that this knowledge can be used to maximize the benefits of small molecule inhibitors, while minimizing cardiac damage in patients with solid malignancies.
Collapse
Affiliation(s)
- Jessica M Scott
- Exercise Physiology and Countermeasures, NASA Johnson Space Center, Universities Space Research Association, 2101 NASA Parkway, Houston, TX 77058, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Vlasova-St Louis I, Dickson AM, Bohjanen PR, Wilusz CJ. CELFish ways to modulate mRNA decay. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:695-707. [PMID: 23328451 DOI: 10.1016/j.bbagrm.2013.01.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 01/03/2013] [Accepted: 01/05/2013] [Indexed: 12/14/2022]
Abstract
The CELF family of RNA-binding proteins regulates many steps of mRNA metabolism. Although their best characterized function is in pre-mRNA splice site choice, CELF family members are also powerful modulators of mRNA decay. In this review we focus on the different modes of regulation that CELF proteins employ to mediate mRNA decay by binding to GU-rich elements. After starting with an overview of the importance of CELF proteins during development and disease pathogenesis, we then review the mRNA networks and cellular pathways these proteins regulate and the mechanisms by which they influence mRNA decay. Finally, we discuss how CELF protein activity is modulated during development and in response to cellular signals. We conclude by highlighting the priorities for new experiments in this field. This article is part of a Special Issue entitled: RNA Decay mechanisms.
Collapse
|
28
|
Abstract
The basic elements of the transforming growth factor-β (TGFβ) pathway were revealed more than a decade ago. Since then, the concept of how the TGFβ signal travels from the membrane to the nucleus has been enriched with additional findings, and its multifunctional nature and medical relevance have relentlessly come to light. However, an old mystery has endured: how does the context determine the cellular response to TGFβ? Solving this question is key to understanding TGFβ biology and its many malfunctions. Recent progress is pointing at answers.
Collapse
Affiliation(s)
- Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.
| |
Collapse
|
29
|
Cells expressing the C/EBPbeta isoform, LIP, engulf their neighbors. PLoS One 2012; 7:e41807. [PMID: 22860016 PMCID: PMC3409234 DOI: 10.1371/journal.pone.0041807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 06/29/2012] [Indexed: 01/05/2023] Open
Abstract
Descriptions of various processes that lead to cell-in-cell structures have been reported for decades. The exact molecular mechanism(s) of their formation and the physiological significance of cell-in-cell structures remain poorly understood. We had previously shown that an isoform of the CCAAT/enhancer-binding protein beta (C/EBPbeta) transcription factor, liver-enriched inhibitory protein (LIP), induces cell death in human breast cancer cells and stimulates autophagy. Here we describe a non-apoptotic cell death process where LIP mediates the engulfment of neighboring cells. We provide evidence of LIP-mediated engulfment via DNA profiling, fluorescent imaging and cell sorting studies, as well as ultrastructure analysis of LIP-expressing MDA-MB-468 breast cancer cells. Our work illustrates that expression of a specific transcription factor, LIP, can mediate cell engulfment.
Collapse
|
30
|
Nahta R. Pharmacological strategies to overcome HER2 cross-talk and Trastuzumab resistance. Curr Med Chem 2012; 19:1065-75. [PMID: 22229414 DOI: 10.2174/092986712799320691] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/28/2011] [Accepted: 12/29/2011] [Indexed: 01/23/2023]
Abstract
Approximately 20-30% of breast cancers show increased expression of the HER2 receptor tyrosine kinase. Trastuzumab (Herceptin) is a clinically approved anti-HER2 monoclonal antibody. Many patients with HER2-overexpressing metastatic breast cancer respond to trastuzumab; however, a subset display primary drug resistance. In addition, many patients who initially respond to trastuzumab ultimately develop disease progression. Multiple molecular mechanisms contributing to trastuzumab resistance have been proposed in the literature. These mechanisms include cross-signaling from related HER/erbB receptors and compensatory signaling from receptors outside of the HER/erbB family, including receptors for insulin-like growth factor-I, vascular endothelial growth factor, and transforming growth factor beta. The major downstream signaling pathway activated by HER2 cross-talk is PI3K/mTOR, and a potential integrator of receptor cross-talk is Src-focal adhesion kinase (FAK) signaling. PI3K, Src, and FAK have independently been implicated in trastuzumab resistance. In this review, we will discuss pharmacological inhibition of HER2 cross-talk as a strategy to treat trastuzumab-refractory HER2-overexpresssing breast cancer.
Collapse
Affiliation(s)
- R Nahta
- Departments of Pharmacology, Emory University School of Medicine, USA.
| |
Collapse
|
31
|
Regulation of C/EBPβ and resulting functions in cells of the monocytic lineage. Cell Signal 2012; 24:1287-96. [DOI: 10.1016/j.cellsig.2012.02.007] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 02/14/2012] [Indexed: 01/10/2023]
|
32
|
Tarragona M, Pavlovic M, Arnal-Estapé A, Urosevic J, Morales M, Guiu M, Planet E, González-Suárez E, Gomis RR. Identification of NOG as a specific breast cancer bone metastasis-supporting gene. J Biol Chem 2012; 287:21346-55. [PMID: 22547073 DOI: 10.1074/jbc.m112.355834] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Metastasis requires numerous biological functions that jointly provide tumor cells from a primary site to seed and colonize a distant organ. Some of these activities are selected for in the primary site, whereas others are acquired at the metastatic niche. We provide molecular evidence showing that the BMP inhibitor, NOG, provides metastatic breast cancer cells with the ability to colonize the bone. NOG expression is acquired during the late events of metastasis, once cells have departed from the primary site, because it is not enriched in primary tumors with high risk of bone relapse. On the contrary, breast cancer bone metastatic lesions do select for high levels of NOG expression when compared with metastasis to the lung, liver, and brain. Pivotal to the bone colonization functions is the contribution of NOG to metastatic autonomous and nonautonomous cell functions. Using genetic approaches, we show that when NOG is expressed in human breast cancer cells, it facilitates bone colonization by fostering osteoclast differentiation and bone degradation and also contributes to metastatic lesions reinitiation. These findings reveal how aggressive cancer cell autonomous and nonautonomous functions can be mechanistically coupled to greater bone metastatic potential.
Collapse
Affiliation(s)
- Maria Tarragona
- Oncology Programme, Institute for Research in Biomedicine, IRB-Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sundqvist A, Ten Dijke P, van Dam H. Key signaling nodes in mammary gland development and cancer: Smad signal integration in epithelial cell plasticity. Breast Cancer Res 2012; 14:204. [PMID: 22315972 PMCID: PMC3496114 DOI: 10.1186/bcr3066] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Smad proteins are the key intermediates of transforming growth factor-beta (TGF-β) signaling during development and in tissue homeostasis. Pertubations in TGF-β/Smad signaling have been implicated in cancer and other diseases. In the cell nucleus, Smad complexes trigger cell type- and context-specific transcriptional programs, thereby transmitting and integrating signals from a variety of ligands of the TGF-β superfamily and other stimuli in the cell microenvironment. The actual transcriptional and biological outcome of Smad activation critically depends on the genomic integrity and the modification state of genome and chromatin of the cell. The cytoplasmic and nuclear Smads can also modulate the activity of other signal transducers and enzymes such as microRNA-processing factors. In the case of breast cancer, the role of Smads in epithelial plasticity, tumor-stroma interactions, invasion, and metastasis seems of particular importance.
Collapse
Affiliation(s)
- Anders Sundqvist
- Ludwig Institute for Cancer Research, Uppsala University, Box 595, 75124, Uppsala, Sweden
| | | | | |
Collapse
|
34
|
Reddy JP, Li Y. Oncogene-induced senescence and its role in tumor suppression. J Mammary Gland Biol Neoplasia 2011; 16:247-56. [PMID: 21681694 DOI: 10.1007/s10911-011-9221-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/07/2011] [Indexed: 01/14/2023] Open
Abstract
While senescence has been known for some time as an inevitable result of repeated DNA replication, oncogene-induced senescence (OIS) represents a relatively new phenomenon. OIS, like apoptosis, has emerged to represent a putative barrier to tumorigenesis in many tissues, including the breast. Here we discuss signals that initiate OIS, evidence for its role in tumor suppression, and mechanisms for its evasion in tumorigenesis.
Collapse
Affiliation(s)
- Jay P Reddy
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
35
|
Li H, Baldwin BR, Zahnow CA. LIP expression is regulated by IGF-1R signaling and participates in suppression of anoikis. Mol Cancer 2011; 10:100. [PMID: 21854628 PMCID: PMC3176234 DOI: 10.1186/1476-4598-10-100] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 08/19/2011] [Indexed: 12/22/2022] Open
Abstract
Background The transcription factor, CCAAT enhancer binding protein-β (C/EBPβ), is expressed as several distinct protein isoforms (LAP1, LAP2 and LIP) that have opposing actions in cellular proliferation and differentiation. Increases in the ratio of LIP/LAP are associated with aggressive, metastatic breast cancer; however, little is known regarding the molecular mechanisms that regulate LIP expression or the biological actions of an increase in the LIP/LAP ratio. Metastasis is highly dependent upon the suppression of anoikis and the role of C/EBPβ and LIP in this anchorage-independent, survival process is currently not known in mammary epithelial cells. IGF-1R signaling is important for the survival of breast cancer cells and crosstalk between IGF-1R and EGFR signaling pathways have been implicated in the development of more aggressive disease. We therefore evaluated in mammary epithelial cells whether IGF-1R signaling regulates the LIP/LAP ratio, analyzed the potential interplay between EGFR and IGF-1R signaling and addressed the biological significance of increased LIP expression in cellular survival and suppression of anoikis. Results Our data provide the first evidence that IGF-1R signaling regulates LIP expression in an EGFR independent manner to increase the LIP/LAP ratio in mammary epithelial cells. Although crosstalk between IGF-1R signaling and EGFR signaling is detectable in MCF10A cells, this crosstalk is not required for the IGF-1 mediated regulation of LIP expression. Rather, the critical regulator of IGF-1 induced LIP expression appears to be EGFR-independent, Akt activity. Our data also demonstrate that increases in LIP expression promote cell survival via suppression of anoikis. Likewise, knockdown of total C/EBPβ leads to increased cell death and suggest that C/EBPβ expression is important for survival and resistance to anoikis. IGF-1 treatment can partially rescue vector control cells from anoikis; however, cells with reduced C/EBPβ expression do not survive anoikis. Conclusions Taken together, our data demonstrate that IGF-1R signaling regulates LIP expression in an EGFR independent manner to increase the LIP/LAP ratio in mammary epithelial cells. C/EBPβ expression and elevations in LIP play an important role in regulating cellular survival via suppression of anoikis, in an IGF-1R mediated context or in a manner independent of IGF-1R signaling.
Collapse
Affiliation(s)
- Huili Li
- Department of Oncology, the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231, USA
| | | | | |
Collapse
|
36
|
Chow A, Arteaga CL, Wang SE. When tumor suppressor TGFβ meets the HER2 (ERBB2) oncogene. J Mammary Gland Biol Neoplasia 2011; 16:81-8. [PMID: 21590373 PMCID: PMC3398103 DOI: 10.1007/s10911-011-9206-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 03/07/2011] [Indexed: 12/28/2022] Open
Abstract
Despite its tumor suppressive role in normal mammary epithelial cells, TGFβ has been reported to promote the migration, invasion and survival in breast cancer cells overexpressing the HER2 (ERBB2; neu) oncogene, and to accelerate the metastasis of neu-induced mammary tumors in mice. A clearer understanding of the molecular mechanisms underlying the crosstalk between TGFβ and HER2 has started to emerge. In recent studies reviewed here, the synergistic effect of TGFβ and HER2 on tumor progression has been shown to likely be a combined result of two distinct features: (1) loss of TGFβ's tumor suppressive effect through functional alterations in the anti-mitogenic effect of Smad-mediated transcription, and (2) gain of pro-survival and pro-migratory function through HER2-dependent mechanisms. In HER2-overexpressing breast cancer, this crosstalk results in increased cancer cell proliferation, survival and invasion, accelerated metastasis in animal models, and resistance to chemotherapy and HER2-targeted therapy. Thus, the transformed cellular context imparted by constitutively active HER2 signaling, as a consequence of HER2 gene amplification or overexpression, aborts the tumor suppressive role of TGFβ and facilitated the oncogenic role of this pathway. In turn, TGFβ potentiates oncogenic HER2 signaling by inducing shedding of the ERBB ligands and clustering of HER2 with integrins. Here we discuss recent studies examining Smad-dependent and -independent mechanisms of crosstalk between TGFβ and HER2. Therefore, blockade of TGFβ:HER2 crosstalk may suppress breast cancer progression and metastasis, and enhance the efficiency of conventional therapies in patients with HER2-overexpressing breast cancer.
Collapse
Affiliation(s)
- Amy Chow
- Division of Tumor Cell Biology, Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | | | | |
Collapse
|
37
|
Vazquez-Martin A, López-Bonetc E, Cufí S, Oliveras-Ferraros C, Del Barco S, Martin-Castillo B, Menendez JA. Repositioning chloroquine and metformin to eliminate cancer stem cell traits in pre-malignant lesions. Drug Resist Updat 2011; 14:212-23. [PMID: 21600837 DOI: 10.1016/j.drup.2011.04.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 01/07/2023]
Abstract
Ideal oncology drugs would be curative after a short treatment course if they could eliminate epithelium-originated carcinomas at their non-invasive, pre-malignant stages. Such ideal molecules, which are expected to molecularly abrogate all the instrumental mechanisms acquired by migrating cancer stem cells (CSCs) to by-pass tumour suppressor barriers, might already exist. We here illustrate how system biology strategies for repositioning existing FDA-approved drugs may accelerate our therapeutic capacity to eliminate CSC traits in pre-invasive intraepithelial neoplasias. First, we describe a signalling network signature that overrides bioenergetics stress- and oncogene-induced senescence (OIS) phenomena in CSCs residing at pre-invasive lesions. Second, we functionally map the anti-malarial chloroquine and the anti-diabetic metformin ("old drugs") to their recently recognized CSC targets ("new uses") within the network. By discussing the preclinical efficacy of chloroquine and metformin to inhibiting the genesis and self-renewal of CSCs we finally underscore the expected translational impact of the "old drugs-new uses" repurposing strategy to open a new CSC-targeted chemoprevention era.
Collapse
Affiliation(s)
- Alejandro Vazquez-Martin
- Unit of Translational Research, Catalan Institute of Oncology-Girona, Avenida de Francia s/n, E-17007 Girona, Catalonia, Spain
| | | | | | | | | | | | | |
Collapse
|
38
|
Mangos JA, Boyd RL, Loughlin GM, Cockrell A, Fucci R. Transductal fluxes of water and monovalent ions in ferret salivary glands. J Dent Res 1981; 130:231-246. [PMID: 6934197 DOI: 10.1172/jci126390] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 09/25/2019] [Indexed: 12/19/2022] Open
Abstract
The net transductal fluxes of water and monovalent ions were measured in the parotid and submandibular salivary glands of the ferret, Mustela putorius furo, during stimulation of secretion with pilocarpine. The duct systems of these glands were found to be impermeable to water using the split-oil droplet method for stationary microperfusion of lobular and main ducts. The net transductal fluxes of Na+, K+, Cl-, and HCO3- were characterized by analysis of ductal fluid samples obtained simultaneously from the intercalated, lobular, and main ducts of these glands.
Collapse
|