1
|
Tsiomita S, Liveri EM, Vardaka P, Vogiatzi A, Skiadaresis A, Saridis G, Tsigkas I, Michaelidis TM, Mavrothalassitis G, Thyphronitis G. ETS2 repressor factor (ERF) is involved in T lymphocyte maturation acting as regulator of thymocyte lineage commitment. J Leukoc Biol 2022; 112:641-657. [PMID: 35258130 DOI: 10.1002/jlb.1a0720-439r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/30/2021] [Indexed: 11/12/2022] Open
Abstract
Thymocyte differentiation and lineage commitment is regulated by an extensive network of transcription factors and signaling molecules among which Erk plays a central role. However, Erk effectors as well as the molecular mechanisms underlying this network are not well understood. Erf is a ubiquitously expressed transcriptional repressor regulated by Erk-dependent phosphorylation. Here, we investigated the role of Erf in T cell maturation and lineage commitment, using a double-fluorescent Erf-floxed mouse to produce thymus-specific Erf knockouts. We observed significant accumulation of thymocytes in the CD4/CD8 DP stage, followed by a significant reduction in CD4SP cells, a trend for lower CD8SP cell frequency, and an elevated percentage of γδ expressing thymocytes in Erf-deficient mice. Also, an elevated number of CD69+ TCRβ+ cells indicates that thymocytes undergoing positive selection accumulate at this stage. The expression of transcription factors Gata3, ThPOK, and Socs1 that promote CD4+ cell commitment was significantly decreased in Erf-deficient mice. These findings suggest that Erf is involved in T cell maturation, acting as a positive regulator during CD4 and eventually CD8 lineage commitment, while negatively regulates the production of γδ T cells. In addition, Erf-deficient mice displayed decreased percentages of CD4+ and CD8+ splenocytes and elevated levels of IL-4 indicating that Erf may have an additional role in the homeostasis, differentiation, and immunologic response of helper and cytotoxic T cells in the periphery. Overall, our results show, for the first time, Erf's involvement in T cell biology suggesting that Erf acts as a potential regulator during thymocyte maturation and thymocyte lineage commitment, in γδ T cell generation, as well as in Th cell differentiation.
Collapse
Affiliation(s)
- Spyridoula Tsiomita
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Effrosyni Maria Liveri
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Panagiota Vardaka
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Angeliki Vogiatzi
- Department of Medicine, Medical School, University of Crete, Heraklion, Greece
| | - Argyris Skiadaresis
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - George Saridis
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Ioannis Tsigkas
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece.,Department of Biomedical Research, Institute of Molecular Biology & Biotechnology, Foundation for Research and Technology-Hellas, Ioannina, Greece
| | - Theologos M Michaelidis
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece.,Department of Biomedical Research, Institute of Molecular Biology & Biotechnology, Foundation for Research and Technology-Hellas, Ioannina, Greece
| | - George Mavrothalassitis
- Department of Medicine, Medical School, University of Crete, Heraklion, Greece.,IMBB, FORTH, Heraklion, Crete, Greece
| | - George Thyphronitis
- Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| |
Collapse
|
2
|
Differential functions of ERK1 and ERK2 in lung metastasis processes in triple-negative breast cancer. Sci Rep 2020; 10:8537. [PMID: 32444778 PMCID: PMC7244517 DOI: 10.1038/s41598-020-65250-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer characterized by metastasis, drug resistance and high rates of recurrence. With a lack or targeted therapies, TNBC is challenging to treat and carries a poor prognosis. Patients with TNBC tumors expressing high levels of ERK2 have a poorer prognosis than those with low ERK2-expressing tumors. The MAPK pathway is often found to be highly activated in TNBC, however the precise functions of the ERK isoforms (ERK1 and ERK2) in cancer progression have not been well defined. We hypothesized that ERK2, but not ERK1, promotes the cancer stem cell (CSC) phenotype and metastasis in TNBC. Stable knockdown clones of the ERK1 and ERK2 isoforms were generated in SUM149 and BT549 TNBC cells using shRNA lentiviral vectors. ERK2 knockdown significantly inhibited anchorage-independent colony formation and mammosphere formation, indicating compromised self-renewal capacity. This effect correlated with a reduction in migration and invasion. SCID-beige mice injected via the tail vein with ERK clones were employed to determine metastatic potential. SUM149 shERK2 cells had a significantly lower lung metastatic burden than control mice or mice injected with SUM149 shERK1 cells. The Affymetrix HGU133plus2 microarray platform was employed to identify gene expression changes in ERK isoform knockdown clones. Comparison of gene expression levels between SUM149 cells with ERK2 or ERK1 knockdown revealed differential and in some cases opposite effects on mRNA expression levels. Those changes associated with ERK2 knockdown predominantly altered regulation of CSCs and metastasis. Our findings indicate that ERK2 promotes metastasis and the CSC phenotype in TNBC.
Collapse
|
3
|
Inoue K, Fry EA. Tumor suppression by the EGR1, DMP1, ARF, p53, and PTEN Network. Cancer Invest 2018; 36:520-536. [PMID: 30396285 PMCID: PMC6500763 DOI: 10.1080/07357907.2018.1533965] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 02/25/2018] [Accepted: 10/05/2018] [Indexed: 01/08/2023]
Abstract
Recent studies have indicated that EGR1 is a direct regulator of tumor suppressors including TGFβ1, PTEN, and p53. The Myb-like transcription factor Dmp1 is a physiological regulator of the Arf-p53 pathway through transactivation of the Arf promoter and physical interaction of p53. The Dmp1 promoter has binding sites for Egr proteins, and Egr1 is a target for Dmp1. Crosstalks between p53 and PTEN have been reported. The Egr1-Dmp1-Arf-p53-Pten pathway displays multiple modes of interaction with each other, suggesting the existence of a functional network of tumor suppressors that maintain normal cell growth and prevent the emergence of incipient cancer cells.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences,
Medical Center Boulevard, Winston-Salem, NC 27157 USA
| | - Elizabeth A. Fry
- The Department of Pathology, Wake Forest University Health Sciences,
Medical Center Boulevard, Winston-Salem, NC 27157 USA
| |
Collapse
|
4
|
Tu Q, Hao J, Zhou X, Yan L, Dai H, Sun B, Yang D, An S, Lv L, Jiao B, Chen C, Lai R, Shi P, Zhao X. CDKN2B deletion is essential for pancreatic cancer development instead of unmeaningful co-deletion due to juxtaposition to CDKN2A. Oncogene 2017; 37:128-138. [PMID: 28892048 PMCID: PMC5759028 DOI: 10.1038/onc.2017.316] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 06/07/2017] [Accepted: 07/31/2017] [Indexed: 12/28/2022]
Abstract
Pancreatic cancer is among the deadliest malignancies; however, the genetic events that lead to pancreatic carcinogenesis in adults remain unclear. In vivo models in which these genetic alterations occur in adult animals may more accurately reflect the features of human cancer. In this study, we demonstrate that inactivation of Cdkn2b (p15ink4b) is necessary for induction of pancreatic cancer by oncogenic KRASG12D expression and inactivation of Tp53 and Cdkn2a in adult mouse pancreatic ductal cells (P60 or older). KRASG12D overexpression in these cells activated transforming growth factor-β signaling and expression of CDKN2B, which, along with CDKN2A, led to cellular senescence and protected cells from KRAS-mediated transformation via inhibition of retinoblastoma phosphorylation. These results show a critical role of CDKN2B inactivation in pancreatic carcinogenesis, and provide a useful adult animal model by genetic engineering via lentiviral delivery.
Collapse
Affiliation(s)
- Q Tu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - J Hao
- State Key Laboratory of Genetic Resources and Evolution, Laboratory of Evolutionary and Functional Genomics, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - X Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - L Yan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming Primate Research Center, Chinese Academy of Sciences, Kunming, China
| | - H Dai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - B Sun
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - D Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - S An
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China
| | - L Lv
- Kunming Primate Research Center, Chinese Academy of Sciences, Kunming, China
| | - B Jiao
- State Key Laboratory of Genetic Resources and Evolution, Laboratory of Evolutionary and Functional Genomics, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - C Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - R Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - P Shi
- State Key Laboratory of Genetic Resources and Evolution, Laboratory of Evolutionary and Functional Genomics, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - X Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming Primate Research Center, Chinese Academy of Sciences, Kunming, China.,KIZ-SU Joint Laboratory of Animal Model and Drug Development, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
5
|
LL-37 stimulates the functions of adipose-derived stromal/stem cells via early growth response 1 and the MAPK pathway. Stem Cell Res Ther 2016; 7:58. [PMID: 27095351 PMCID: PMC4837546 DOI: 10.1186/s13287-016-0313-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 03/21/2016] [Accepted: 04/04/2016] [Indexed: 01/03/2023] Open
Abstract
Background LL-37 is a naturally occurring antimicrobial peptide found in the wound bed and assists wound repair. No published study has characterized the role of LL-37 in the function(s) of human mesenchymal stem cells (MSCs). This study investigated the functions of adipose-derived stromal/stem cells (ASCs) activated by LL-37 by performing both in vitro assays with cultured cells and in vivo assays with C57BL/6 mice with hair loss. Methods Human ASCs were isolated from healthy donors with written informed consent. To examine the effects of LL-37 on ASC function, cell proliferation and migration were measured by a cell counting kit (CCK-8) and a Transwell migration assay. Early growth response 1 (EGR1) mRNA expression was determined by microarray and real-time PCR analyses. The protein levels of EGR1 and regenerative factors were analyzed by specific enzyme-linked immunosorbent assays and western blotting. Results LL-37 treatment enhanced the proliferation and migration of human ASCs expressing formyl peptide receptor like-1. Microarray and real-time PCR data showed that EGR1 expression was rapidly and significantly increased by LL-37 treatment. LL-37 treatment also enhanced the production of EGR1. Moreover, small interfering RNA-mediated knockdown of EGR1 inhibited LL-37-enhanced ASC proliferation and migration. Activation of mitogen-activated protein kinases (MAPKs) was essential not only for LL-37-enhanced ASC proliferation and migration but also EGR1 expression; treatment with a specific inhibitor of extracellular signal-regulated kinase, p38, or c-Jun N-terminal kinase blocked the stimulatory effect of LL-37. EGR1 has a strong paracrine capability and can influence angiogenic factors in ASCs; therefore, we evaluated the secretion levels of vascular endothelial growth factor, thymosin beta-4, monocyte chemoattractant protein-1, and stromal cell-derived factor-1. LL-37 treatment increased the secretion of these regenerative factors. Moreover, treatment with the conditioned medium of ASCs pre-activated with LL-37 strongly promoted hair growth in vivo. Conclusions These findings show that LL-37 increases EGR1 expression and MAPK activation, and that preconditioning of ASCs with LL-37 has a strong potential to promote hair growth in vivo. This study correlates LL-37 with MSC functions (specifically those of ASCs), including cell expansion, cell migration, and paracrine actions, which may be useful in terms of implantation for tissue regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0313-4) contains supplementary material, which is available to authorized users.
Collapse
|
6
|
Sakakini N, Turchi L, Bergon A, Holota H, Rekima S, Lopez F, Paquis P, Almairac F, Fontaine D, Baeza-Kallee N, Van Obberghen-Schilling E, Junier MP, Chneiweiss H, Figarella-Branger D, Burel-Vandenbos F, Imbert J, Virolle T. A Positive Feed-forward Loop Associating EGR1 and PDGFA Promotes Proliferation and Self-renewal in Glioblastoma Stem Cells. J Biol Chem 2016; 291:10684-99. [PMID: 27002148 DOI: 10.1074/jbc.m116.720698] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Indexed: 01/06/2023] Open
Abstract
Glioblastomas are the most common primary brain tumors, highly vascularized, infiltrating, and resistant to current therapies. This cancer leads to a fatal outcome in less than 18 months. The aggressive behavior of glioblastomas, including resistance to current treatments and tumor recurrence, has been attributed to glioma stemlike/progenitor cells. The transcription factor EGR1 (early growth response 1), a member of a zinc finger transcription factor family, has been described as tumor suppressor in gliomas when ectopically overexpressed. Although EGR1 expression in human glioblastomas has been associated with patient survival, its precise location in tumor territories as well as its contribution to glioblastoma progression remain elusive. In the present study, we show that EGR1-expressing cells are more frequent in high grade gliomas where the nuclear expression of EGR1 is restricted to proliferating/progenitor cells. We show in primary cultures of glioma stemlike cells that EGR1 contributes to stemness marker expression and proliferation by orchestrating a PDGFA-dependent growth-stimulatory loop. In addition, we demonstrate that EGR1 acts as a positive regulator of several important genes, including SHH, GLI1, GLI2, and PDGFA, previously linked to the maintenance and proliferation of glioma stemlike cells.
Collapse
Affiliation(s)
- Nathalie Sakakini
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France, INSERM, U1090, Transcriptomic and Genomic Marseille-Luminy/Technical Advances for Genomics and Clinics (TGML/TAGC), Marseille F-13009, France, UMR_S 1090, TGML/TAGC, Aix-Marseille Université, Marseille F-13009, France
| | - Laurent Turchi
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France
| | - Aurélie Bergon
- INSERM, U1090, Transcriptomic and Genomic Marseille-Luminy/Technical Advances for Genomics and Clinics (TGML/TAGC), Marseille F-13009, France, UMR_S 1090, TGML/TAGC, Aix-Marseille Université, Marseille F-13009, France
| | - Hélène Holota
- INSERM, U1090, Transcriptomic and Genomic Marseille-Luminy/Technical Advances for Genomics and Clinics (TGML/TAGC), Marseille F-13009, France, UMR_S 1090, TGML/TAGC, Aix-Marseille Université, Marseille F-13009, France
| | - Samah Rekima
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France
| | - Fabrice Lopez
- INSERM, U1090, Transcriptomic and Genomic Marseille-Luminy/Technical Advances for Genomics and Clinics (TGML/TAGC), Marseille F-13009, France, UMR_S 1090, TGML/TAGC, Aix-Marseille Université, Marseille F-13009, France
| | - Philipe Paquis
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France, the Service de Neurchirurgie, Hôpital Pasteur, CHU de Nice, Nice 06107, France
| | - Fabien Almairac
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France, the Service de Neurchirurgie, Hôpital Pasteur, CHU de Nice, Nice 06107, France
| | - Denys Fontaine
- the Service de Neurchirurgie, Hôpital Pasteur, CHU de Nice, Nice 06107, France
| | - Nathalie Baeza-Kallee
- Aix Marseille Université, Faculté de Médecine de la Timone, 13284 Marseille, France, CRO2, INSERM UMR 911, 13284 Marseille Cedex, France
| | | | - Marie-Pierre Junier
- CNRS UMR8246 Neuroscience Paris Seine-IBPS, Team Glial Plasticity, 7 Quai Saint-Bernard, Paris 75005, France, INSERM U1130, Neuroscience Paris Seine-IBPS, Team Glial Plasticity, 7 Quai Saint-Bernard, Paris 75005, France, and University Pierre and Marie Curie UMCR18, Neuroscience Paris Seine-IBPS, Team Glial Plasticity, 7 Quai Saint-Bernard, Paris 75005, France
| | - Hervé Chneiweiss
- CNRS UMR8246 Neuroscience Paris Seine-IBPS, Team Glial Plasticity, 7 Quai Saint-Bernard, Paris 75005, France, INSERM U1130, Neuroscience Paris Seine-IBPS, Team Glial Plasticity, 7 Quai Saint-Bernard, Paris 75005, France, and University Pierre and Marie Curie UMCR18, Neuroscience Paris Seine-IBPS, Team Glial Plasticity, 7 Quai Saint-Bernard, Paris 75005, France
| | - Dominique Figarella-Branger
- Aix Marseille Université, Faculté de Médecine de la Timone, 13284 Marseille, France, CRO2, INSERM UMR 911, 13284 Marseille Cedex, France, the Departement de Pathology, CHU de la Timone, 13385 Marseille Cedex 5, France
| | - Fanny Burel-Vandenbos
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France, the Service d'Anatomopathologie, Hôpital Pasteur, CHU de Nice, Nice 06107, France
| | - Jean Imbert
- INSERM, U1090, Transcriptomic and Genomic Marseille-Luminy/Technical Advances for Genomics and Clinics (TGML/TAGC), Marseille F-13009, France, UMR_S 1090, TGML/TAGC, Aix-Marseille Université, Marseille F-13009, France,
| | - Thierry Virolle
- From the Université Nice Sophia Antipolis, CNRS, INSERM, iBV, 06108 Nice, France,
| |
Collapse
|
7
|
Jamnikar U, Nikolic P, Belic A, Blas M, Gaser D, Francky A, Laux H, Blejec A, Baebler S, Gruden K. Transcriptome study and identification of potential marker genes related to the stable expression of recombinant proteins in CHO clones. BMC Biotechnol 2015; 15:98. [PMID: 26499110 PMCID: PMC4812793 DOI: 10.1186/s12896-015-0218-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 10/19/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chinese hamster ovary (CHO) cells have become the host of choice for the production of recombinant proteins, due to their capacity for correct protein folding, assembly, and posttranslational modifications. The most widely used system for recombinant proteins is the gene amplification procedure that uses the CHO-Dhfr expression system. However, CHO cells are known to have a very unstable karyotype. This is due to chromosome rearrangements that can arise from translocations and homologous recombination, especially when cells with the CHO-Dhfr expression system are treated with methotrexate hydrate. The present method used in the industry for testing clones for their long-term stability of recombinant protein production is empirical, and it involves their cultivation over extended periods of time prior to the selection of the most suitable clone for further bioprocess development. The aim of the present study was the identification of marker genes that can predict stable expression of recombinant genes in particular clones early in the development stage. RESULTS The transcriptome profiles of CHO clones with stable and unstable recombinant protein production were investigated over 10-weeks of cultivation, using a DNA microarray. We identified 14 genes that were differentially expressed between the stable and unstable clones already at 2 weeks from the beginning of the cultivation. Their expression was validated by reverse-transcription quantitative real-time PCR (RT-qPCR). Furthermore, the k-nearest neighbour algorithm approach shows that the combination of the gene expression patterns of only five of these 14 genes is sufficient to predict stable recombinant protein production in clones in the early phases of cell-line development. CONCLUSIONS The exact molecular mechanisms that cause unstable recombinant protein production are not fully understood. However, the expression profiles of some genes in clones with stable and unstable recombinant protein production allow prediction of such instability early in the cell-line development stage. We have thus developed a proof-of-concept for a novel approach to eliminate unstable clones in the CHO-Dhfr expression system, which saves time and labour-intensive work in cell-line development.
Collapse
Affiliation(s)
- Uros Jamnikar
- Sandoz Biopharmaceuticals, Kolodvorska 27, SI-1234, Menges, Slovenia.
| | - Petra Nikolic
- Jozef Stefan Institute, Jamova cesta 39, SI-1000, Ljubljana, Slovenia.
| | - Ales Belic
- Sandoz Biopharmaceuticals, Kolodvorska 27, SI-1234, Menges, Slovenia.
| | - Marjanca Blas
- Sandoz Biopharmaceuticals, Kolodvorska 27, SI-1234, Menges, Slovenia.
| | - Dominik Gaser
- Sandoz Biopharmaceuticals, Kolodvorska 27, SI-1234, Menges, Slovenia.
| | - Andrej Francky
- Sandoz Biopharmaceuticals, Kolodvorska 27, SI-1234, Menges, Slovenia.
| | - Holger Laux
- Novartis Pharma AG, WKL-681.1.08, 4002, Basel, Switzerland.
| | - Andrej Blejec
- National Institute of Biology, Vecna pot 111, SI-1000, Ljubljana, Slovenia.
| | - Spela Baebler
- National Institute of Biology, Vecna pot 111, SI-1000, Ljubljana, Slovenia.
| | - Kristina Gruden
- National Institute of Biology, Vecna pot 111, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
8
|
McNeal AS, Liu K, Nakhate V, Natale CA, Duperret EK, Capell BC, Dentchev T, Berger SL, Herlyn M, Seykora JT, Ridky TW. CDKN2B Loss Promotes Progression from Benign Melanocytic Nevus to Melanoma. Cancer Discov 2015; 5:1072-85. [PMID: 26183406 DOI: 10.1158/2159-8290.cd-15-0196] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 07/09/2015] [Indexed: 12/21/2022]
Abstract
UNLABELLED Deletion of the entire CDKN2B-CDKN2A gene cluster is among the most common genetic events in cancer. The tumor-promoting effects are generally attributed to loss of CDKN2A-encoded p16 and p14ARF tumor suppressors. The degree to which the associated CDKN2B-encoded p15 loss contributes to human tumorigenesis is unclear. Here, we show that CDKN2B is highly upregulated in benign melanocytic nevi, contributes to maintaining nevus melanocytes in a growth-arrested premalignant state, and is commonly lost in melanoma. Using primary melanocytes isolated directly from freshly excised human nevi naturally expressing the common BRAF(V600E)-activating mutation, nevi progressing to melanoma, and normal melanocytes engineered to inducibly express BRAF(V600E), we show that BRAF activation results in reversible, TGFβ-dependent, p15 induction that halts proliferation. Furthermore, we engineer human skin grafts containing nevus-derived melanocytes to establish a new, architecturally faithful, in vivo melanoma model, and demonstrate that p15 loss promotes the transition from benign nevus to melanoma. SIGNIFICANCE Although BRAF(V600E) mutations cause melanocytes to initially proliferate into benign moles, mechanisms responsible for their eventual growth arrest are unknown. Using melanocytes from human moles, we show that BRAF activation leads to a CDKN2B induction that is critical for restraining BRAF oncogenic effects, and when lost, contributes to melanoma.
Collapse
Affiliation(s)
- Andrew S McNeal
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kevin Liu
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vihang Nakhate
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher A Natale
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth K Duperret
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian C Capell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania. Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tzvete Dentchev
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shelley L Berger
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - John T Seykora
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Todd W Ridky
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Feldman ME, Yarden Y. Steering tumor progression through the transcriptional response to growth factors and stroma. FEBS Lett 2014; 588:2407-14. [PMID: 24873881 DOI: 10.1016/j.febslet.2014.05.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 02/04/2023]
Abstract
Tumor progression can be understood as a collaborative effort of mutations and growth factors, which propels cell proliferation and matrix invasion, and also enables evasion of drug-induced apoptosis. Concentrating on EGFR, we discuss downstream signaling and the initiation of transcriptional events in response to growth factors. Specifically, we portray a wave-like program, which initiates by rapid disappearance of two-dozen microRNAs, followed by an abrupt rise of immediate early genes (IEGs), relatively short transcripts encoding transcriptional regulators. Concurrent with the fall of IEGs, some 30-60 min after stimulation, a larger group, the delayed early genes, is up-regulated and its own fall overlaps the rise of the final wave of late response genes. This late wave persists and determines long-term phenotype acquisition, such as invasiveness. Key regulatory steps in the orderly response to growth factors provide a trove of potential oncogenes and tumor suppressors.
Collapse
Affiliation(s)
- Morris E Feldman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
10
|
Burgess AW, Henis YI, Hynes NE, Jovin T, Levitzki A, Pinkas-Kramarski R, Yarden Y. EGF receptor family: twisting targets for improved cancer therapies. Growth Factors 2014; 32:74-81. [PMID: 24641597 DOI: 10.3109/08977194.2014.896355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The epidermal growth factor receptor (EGFR) undergoes a conformational change in response to ligand binding. The ligand-induced changes in cell surface aggregation and mobility have a profound effect on the function of all the family members. Ligand also activates the EGFR intracellular kinase, stimulating proliferation and cell survival. The EGFR family are often activated, overexpressed or mutated in cancer cells and therapeutic drugs (including antibodies) can slow the progress of some cancers. This article provides a brief, annotated summary of the presentations and discussion which occurred at the Epidermal Growth Factor Receptor - Future Directions Conference held in Jerusalem in November 2013.
Collapse
Affiliation(s)
- Antony W Burgess
- The Walter & Eliza Hall Institute of Medical Research, Burgess Lab Structural Biology , Parkville , Australia
| | | | | | | | | | | | | |
Collapse
|
11
|
Xie J, Chen M, Zhou J, Mo MS, Zhu LH, Liu YP, Gui QJ, Zhang L, Li GQ. miR-7 inhibits the invasion and metastasis of gastric cancer cells by suppressing epidermal growth factor receptor expression. Oncol Rep 2014; 31:1715-22. [PMID: 24573489 DOI: 10.3892/or.2014.3052] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/11/2014] [Indexed: 01/19/2023] Open
Abstract
The present study profiled differentially expressed microRNAs (miRs) in gastric cancer cell lines and then investigated miR-7 expression in gastric cancer tissue specimens and the effects of miR-7 on the growth, invasion and metastasis of gastric cancer cells and the underlying molecular events. A microRNA microarray was used to profile differentially expressed miRNAs in human gastric cancer cell lines relative to a normal stomach mucosal epithelial cell line. The miRNA miR-7 was selected for further investigation, which included real-time reverse-transcription PCR (qRT-PCR) analysis of miR-7 levels in different gastric cancer cell lines and tissues and distant non-tumor tissues from patient resections. Cell counting kit-8 (CCK-8), Transwell migration and invasion, and western blot assays were performed to assess tumor cell viability, invasion and gene expression, respectively, after miR-7 transfection. The miRNA microarray profiling revealed 14 upregulated miRNAs (including miR-21, miR-26b and miR-30b) and 19 downregulated miRNAs (including let-7i, miR-7 and miR-622) between gastric cancer and normal cell lines. The qRT-PCR analysis confirmed that reduced miR-7 expression occurred more frequently in poorly and moderately differentiated gastric cancer MGC-803, MKN-45 and SGC-7901 cell lines than in the well-differentiated gastric cancer NCI-N87 cell line, which was consistent with the results for gastric cancer tissues. Expression of miR-7 was downregulated in 86.9% (20/23) of the gastric cancer tissues compared with that in the distant non-tumor tissues. Restoration of miR-7 expression significantly inhibited tumor cell viability, invasiveness and migration when compared with the control cells. Luciferase assay confirmed the epidermal growth factor receptor (EGFR) as a target gene of mR-7, and expression of miR-7 significantly suppressed EGFR expression at both the mRNA and protein levels. The data from the present study demonstrated that reduced miR-7 expression contributes to gastric cancer development and progression. Further study will investigate miR-7 in the regulation of EGFR expression in vitro and in vivo.
Collapse
Affiliation(s)
- Juan Xie
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ming Chen
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jing Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ming-Shu Mo
- Department of Diagnostics, Medical College of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li-Hui Zhu
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yan-Ping Liu
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qing-Jun Gui
- Department of Diagnostics, Medical College of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| | - Guo-Qing Li
- Department of Gastroenterology, The Second Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|