1
|
Gu CB, Wang C. GRB14: A prognostic biomarker driving tumor progression in gastric cancer through the PI3K/AKT signaling pathway by interacting with COBLL1. Open Life Sci 2025; 20:20251084. [PMID: 40321159 PMCID: PMC12048902 DOI: 10.1515/biol-2025-1084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/14/2025] [Accepted: 02/22/2025] [Indexed: 05/08/2025] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy with a high incidence rate. Growth factor receptor-bound protein 14 (GRB14) is crucial in cell signal transduction and is associated with tumor growth, invasion, and metastasis. The aim of this study is to investigate the impact of GRB14 on GC growth and metastasis. GRB14 expression and prognosis in GC tissues were analyzed using bioinformatics. The GC cell lines, SGC-7901, MGC-803, BGC-823, and normal gastric epithelial cell line (GES-1) were used in this study. Cell viability, cycle progression, and apoptosis were assessed via CCK-8 and flow cytometry. The colony formation, transwell, and wound-healing assays were conducted to evaluate cell proliferation, invasion, and migration. Protein levels involved in the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway were analyzed by Western blot. GRB14 expression was significantly higher in GC tissues than adjacent healthy tissues, correlating with poor prognosis. GRB14 knockdown promoted apoptosis and inhibited cell growth, invasion, and migration, while its overexpression exhibited opposite effects. GRB14 directly interacted with cordon-bleu WH2 repeat protein like 1, facilitating PI3K/AKT signaling in GC cells. This study highlights GRB14's critical role in GC progression and suggests its potential as a therapeutic target.
Collapse
Affiliation(s)
- Chun-Bin Gu
- Medical College, Soochow University,
Suzhou, 215006, P.R. China
- Department of General Surgery, Sheyang County People’s Hospital, Sheyang, 224300,
P.R. China
| | - Chuang Wang
- Department of General Surgery, Hulunbuir People’s Hospital Affiliated to Soochow University,
Hulunbuir, 021000, P.R. China
| |
Collapse
|
2
|
Many GM, Sanford JA, Sagendorf TJ, Hou Z, Nigro P, Whytock KL, Amar D, Caputo T, Gay NR, Gaul DA, Hirshman MF, Jimenez-Morales D, Lindholm ME, Muehlbauer MJ, Vamvini M, Bergman BC, Fernández FM, Goodyear LJ, Hevener AL, Ortlund EA, Sparks LM, Xia A, Adkins JN, Bodine SC, Newgard CB, Schenk S. Sexual dimorphism and the multi-omic response to exercise training in rat subcutaneous white adipose tissue. Nat Metab 2024; 6:963-979. [PMID: 38693320 PMCID: PMC11132991 DOI: 10.1038/s42255-023-00959-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 12/01/2023] [Indexed: 05/03/2024]
Abstract
Subcutaneous white adipose tissue (scWAT) is a dynamic storage and secretory organ that regulates systemic homeostasis, yet the impact of endurance exercise training (ExT) and sex on its molecular landscape is not fully established. Utilizing an integrative multi-omics approach, and leveraging data generated by the Molecular Transducers of Physical Activity Consortium (MoTrPAC), we show profound sexual dimorphism in the scWAT of sedentary rats and in the dynamic response of this tissue to ExT. Specifically, the scWAT of sedentary females displays -omic signatures related to insulin signaling and adipogenesis, whereas the scWAT of sedentary males is enriched in terms related to aerobic metabolism. These sex-specific -omic signatures are preserved or amplified with ExT. Integration of multi-omic analyses with phenotypic measures identifies molecular hubs predicted to drive sexually distinct responses to training. Overall, this study underscores the powerful impact of sex on adipose tissue biology and provides a rich resource to investigate the scWAT response to ExT.
Collapse
Affiliation(s)
- Gina M Many
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - James A Sanford
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Tyler J Sagendorf
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Zhenxin Hou
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Pasquale Nigro
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - David Amar
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Tiziana Caputo
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nicole R Gay
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - David A Gaul
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - David Jimenez-Morales
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Malene E Lindholm
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michael J Muehlbauer
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA
| | - Maria Vamvini
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Bryan C Bergman
- Division of Endocrinology, Diabetes, and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Facundo M Fernández
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Andrea L Hevener
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, University of California, Los Angeles, CA, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Ashley Xia
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joshua N Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Sue C Bodine
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| | - Christopher B Newgard
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC, USA.
| | - Simon Schenk
- Department of Orthopaedic Surgery, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
3
|
Sun JR, Kong CF, Ye YX, Wang Q, Qu XK, Jia LQ, Wu S. Integrated analysis of single-cell and bulk RNA-sequencing reveals a novel signature based on NK cell marker genes to predict prognosis and immunotherapy response in gastric cancer. Sci Rep 2024; 14:7648. [PMID: 38561388 PMCID: PMC10985121 DOI: 10.1038/s41598-024-57714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Natural killer (NK) cells play essential roles in the tumor development, diagnosis, and prognosis of tumors. In this study, we aimed to establish a reliable signature based on marker genes in NK cells, thus providing a new perspective for assessing immunotherapy and the prognosis of patients with gastric cancer (GC). We analyzed a total of 1560 samples retrieved from the public database. We performed a comprehensive analysis of single-cell RNA-sequencing (scRNA-seq) data of gastric cancer and identified 377 marker genes for NK cells. By performing Cox regression analysis, we established a 12-gene NK cell-associated signature (NKCAS) for the Cancer Genome Atlas (TCGA) cohort, that assigned GC patients into a low-risk group (LRG) or a high-risk group (HRG). In the TCGA cohort, the areas under curve (AUC) value were 0.73, 0.81, and 0.80 at 1, 3, and 5 years. External validation of the predictive ability for the signature was then validated in the Gene Expression Omnibus (GEO) cohorts (GSE84437). The expression levels of signature genes were measured and validated in GC cell lines by real-time PCR. Moreover, NKCAS was identified as an independent prognostic factor by multivariate analysis. We combined this with a variety of clinicopathological characteristics (age, M stage, and tumor grade) to construct a nomogram to predict the survival outcomes of patients. Moreover, the LRG showed higher immune cell infiltration, especially CD8+ T cells and NK cells. The risk score was negatively associated with inflammatory activities. Importantly, analysis of the independent immunotherapy cohort showed that the LRG had a better prognosis and immunotherapy response when compared with the HRG. The identification of NK cell marker genes in this study suggests potential therapeutic targets. Additionally, the developed predictive signatures and nomograms may aid in the clinical management of GC.
Collapse
Affiliation(s)
- Jian-Rong Sun
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Chen-Fan Kong
- Department of Urology, The affiliated Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16, Liantangxiantong Road, Shenzhen, 518009, Luohu, People's Republic of China
| | - Yi-Xiang Ye
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Qin Wang
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Xiang-Ke Qu
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Li-Qun Jia
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China.
| | - Song Wu
- Department of Urology, The affiliated Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16, Liantangxiantong Road, Shenzhen, 518009, Luohu, People's Republic of China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, People's Republic of China.
| |
Collapse
|
4
|
Parga-Pazos M, Cusimano N, Rábano M, Akhmatskaya E, Vivanco MDM. A Novel Mathematical Approach for Analysis of Integrated Cell-Patient Data Uncovers a 6-Gene Signature Linked to Endocrine Therapy Resistance. J Transl Med 2024; 104:100286. [PMID: 37951307 DOI: 10.1016/j.labinv.2023.100286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 10/17/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023] Open
Abstract
A significant number of breast cancers develop resistance to hormone therapy. This progression, while posing a major clinical challenge, is difficult to predict. Despite important contributions made by cell models and clinical studies to tackle this problem, both present limitations when taken individually. Experiments with cell models are highly reproducible but do not reflect the indubitable heterogenous landscape of breast cancer. On the other hand, clinical studies account for this complexity but introduce uncontrolled noise due to external factors. Here, we propose a new approach for biomarker discovery that is based on a combined analysis of sequencing data from controlled MCF7 cell experiments and heterogenous clinical samples that include clinical and sequencing information from The Cancer Genome Atlas. Using data from differential gene expression analysis and a Bayesian logistic regression model coupled with an original simulated annealing-type algorithm, we discovered a novel 6-gene signature for stratifying patient response to hormone therapy. The experimental observations and computational analysis built on independent cohorts indicated the superior predictive performance of this gene set over previously known signatures of similar scope. Together, these findings revealed a new gene signature to identify patients with breast cancer with an increased risk of developing resistance to endocrine therapy.
Collapse
Affiliation(s)
- Martin Parga-Pazos
- Modelling and Simulation in Life and Materials Sciences, Basque Center for Applied Mathematics, Spain; Cancer Heterogeneity Lab, CIC bioGUNE, Basque Research and Technology Alliance, Derio, Spain
| | - Nicole Cusimano
- Modelling and Simulation in Life and Materials Sciences, Basque Center for Applied Mathematics, Spain
| | - Miriam Rábano
- Cancer Heterogeneity Lab, CIC bioGUNE, Basque Research and Technology Alliance, Derio, Spain
| | - Elena Akhmatskaya
- Modelling and Simulation in Life and Materials Sciences, Basque Center for Applied Mathematics, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Maria dM Vivanco
- Cancer Heterogeneity Lab, CIC bioGUNE, Basque Research and Technology Alliance, Derio, Spain.
| |
Collapse
|
5
|
Giriyappagoudar M, Vastrad B, Horakeri R, Vastrad C. Identification and Interaction Analysis of Molecular Markers in Pancreatic Ductal Adenocarcinoma by Bioinformatics and Next-Generation Sequencing Data Analysis. Bioinform Biol Insights 2023; 17:11779322231186719. [PMID: 37529485 PMCID: PMC10387711 DOI: 10.1177/11779322231186719] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/18/2023] [Indexed: 08/03/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the most common cancers worldwide. Intense efforts have been made to elucidate the molecular pathogenesis, but the molecular mechanisms of PDAC are still not well understood. The purpose of this study is to further explore the molecular mechanism of PDAC through integrated bioinformatics analysis. Methods To identify the candidate genes in the carcinogenesis and progression of PDAC, next-generation sequencing (NGS) data set GSE133684 was downloaded from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were identified, and Gene Ontology (GO) and pathway enrichment analyses were performed. The protein-protein interaction network (PPI) was constructed and the module analysis was performed using Integrated Interactions Database (IID) interactome database and Cytoscape. Subsequently, miRNA-DEG regulatory network and TF-DEG regulatory network were constructed using miRNet database, NetworkAnalyst database, and Cytoscape software. The expression levels of hub genes were validated based on Kaplan-Meier analysis, expression analysis, stage analysis, mutation analysis, protein expression analysis, immune infiltration analysis, and receiver operating characteristic (ROC) curve analysis. Results A total of 463 DEGs were identified, consisting of 232 upregulated genes and 233 downregulated genes. The enriched GO terms and pathways of the DEGs include vesicle organization, secretory vesicle, protein dimerization activity, lymphocyte activation, cell surface, transferase activity, transferring phosphorus-containing groups, hemostasis, and adaptive immune system. Four hub genes (namely, cathepsin B [CCNB1], four-and-a-half LIM domains 2 (FHL2), major histocompatibility complex, class II, DP alpha 1 (HLA-DPA1) and tubulin beta 1 class VI (TUBB1)) were obtained via taking interaction of different analysis results. Conclusions On the whole, the findings of this investigation enhance our understanding of the potential molecular mechanisms of PDAC and provide potential targets for further investigation.
Collapse
Affiliation(s)
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. Society’s College of Pharmacy, Gadag, India
| | - Rajeshwari Horakeri
- Department of Computer Science, Government First Grade College, Hubballi, India
| | | |
Collapse
|
6
|
Many GM, Sanford JA, Sagendorf TJ, Hou Z, Nigro P, Whytock K, Amar D, Caputo T, Gay NR, Gaul DA, Hirshman M, Jimenez-Morales D, Lindholm ME, Muehlbauer MJ, Vamvini M, Bergman B, Fern Ndez FM, Goodyear LJ, Ortlund EA, Sparks LM, Xia A, Adkins JN, Bodine SC, Newgard CB, Schenk S. Sexual dimorphism and the multi-omic response to exercise training in rat subcutaneous white adipose tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.527012. [PMID: 36778330 PMCID: PMC9915732 DOI: 10.1101/2023.02.03.527012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Subcutaneous white adipose tissue (scWAT) is a dynamic storage and secretory organ that regulates systemic homeostasis, yet the impact of endurance exercise training and sex on its molecular landscape has not been fully established. Utilizing an integrative multi-omics approach with data generated by the Molecular Transducers of Physical Activity Consortium (MoTrPAC), we identified profound sexual dimorphism in the dynamic response of rat scWAT to endurance exercise training. Despite similar cardiorespiratory improvements, only male rats reduced whole-body adiposity, scWAT adipocyte size, and total scWAT triglyceride abundance with training. Multi-omic analyses of adipose tissue integrated with phenotypic measures identified sex-specific training responses including enrichment of mTOR signaling in females, while males displayed enhanced mitochondrial ribosome biogenesis and oxidative metabolism. Overall, this study reinforces our understanding that sex impacts scWAT biology and provides a rich resource to interrogate responses of scWAT to endurance training.
Collapse
|
7
|
Naudin C, Chevalier C, Roche S. The role of small adaptor proteins in the control of oncogenic signalingr driven by tyrosine kinases in human cancer. Oncotarget 2017; 7:11033-55. [PMID: 26788993 PMCID: PMC4905456 DOI: 10.18632/oncotarget.6929] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/01/2016] [Indexed: 12/15/2022] Open
Abstract
Protein phosphorylation on tyrosine (Tyr) residues has evolved as an important mechanism to coordinate cell communication in multicellular organisms. The importance of this process has been revealed by the discovery of the prominent oncogenic properties of tyrosine kinases (TK) upon deregulation of their physiological activities, often due to protein overexpression and/or somatic mutation. Recent reports suggest that TK oncogenic signaling is also under the control of small adaptor proteins. These cytosolic proteins lack intrinsic catalytic activity and signal by linking two functional members of a catalytic pathway. While most adaptors display positive regulatory functions, a small group of this family exerts negative regulatory functions by targeting several components of the TK signaling cascade. Here, we review how these less studied adaptor proteins negatively control TK activities and how their loss of function induces abnormal TK signaling, promoting tumor formation. We also discuss the therapeutic consequences of this novel regulatory mechanism in human oncology.
Collapse
Affiliation(s)
- Cécile Naudin
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Present address: INSERM U1016, CNRS UMR8104, Institut Cochin, Paris, France
| | - Clément Chevalier
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Present address: SFR Biosit (UMS CNRS 3480/US INSERM 018), MRic Photonics Platform, University Rennes, Rennes, France
| | - Serge Roche
- CNRS UMR5237, University Montpellier, CRBM, Montpellier, France.,Equipe Labellisée LIGUE 2014, Ligue Contre le Cancer, Paris, France
| |
Collapse
|
8
|
Morzyglod L, Caüzac M, Popineau L, Denechaud PD, Fajas L, Ragazzon B, Fauveau V, Planchais J, Vasseur-Cognet M, Fartoux L, Scatton O, Rosmorduc O, Guilmeau S, Postic C, Desdouets C, Desbois-Mouthon C, Burnol AF. Growth factor receptor binding protein 14 inhibition triggers insulin-induced mouse hepatocyte proliferation and is associated with hepatocellular carcinoma. Hepatology 2017; 65:1352-1368. [PMID: 27981611 DOI: 10.1002/hep.28972] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 11/16/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Metabolic diseases such as obesity and type 2 diabetes are recognized as independent risk factors for hepatocellular carcinoma (HCC). Hyperinsulinemia, a hallmark of these pathologies, is suspected to be involved in HCC development. The molecular adapter growth factor receptor binding protein 14 (Grb14) is an inhibitor of insulin receptor catalytic activity, highly expressed in the liver. To study its involvement in hepatocyte proliferation, we specifically inhibited its liver expression using a short hairpin RNA strategy in mice. Enhanced insulin signaling upon Grb14 inhibition was accompanied by a transient induction of S-phase entrance by quiescent hepatocytes, indicating that Grb14 is a potent repressor of cell division. The proliferation of Grb14-deficient hepatocytes was cell-autonomous as it was also observed in primary cell cultures. Combined Grb14 down-regulation and insulin signaling blockade using pharmacological approaches as well as genetic mouse models demonstrated that Grb14 inhibition-mediated hepatocyte division involved insulin receptor activation and was mediated by the mechanistic target of rapamycin complex 1-S6K pathway and the transcription factor E2F1. In order to determine a potential dysregulation in GRB14 gene expression in human pathophysiology, a collection of 85 human HCCs was investigated. This revealed a highly significant and frequent decrease in GRB14 expression in hepatic tumors when compared to adjacent nontumoral parenchyma, with 60% of the tumors exhibiting a reduced Grb14 mRNA level. CONCLUSION Our study establishes Grb14 as a physiological repressor of insulin mitogenic action in the liver and further supports that dysregulation of insulin signaling is associated with HCC. (Hepatology 2017;65:1352-1368).
Collapse
Affiliation(s)
- Lucille Morzyglod
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Michèle Caüzac
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Lucie Popineau
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Pierre-Damien Denechaud
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.,Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Lluis Fajas
- Department of Physiology, University of Lausanne, Lausanne, Switzerland.,Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Bruno Ragazzon
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Véronique Fauveau
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Julien Planchais
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Mireille Vasseur-Cognet
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRA 1392, Paris, and Institut d'Ecologie et des Sciences de l'Environnement de Paris, Bondy, France.,Sorbonne Universités, Paris, France.,Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Laetitia Fartoux
- APHP, Hôpital La Pitié Salpêtrière, Service d'Hépato-Gastroentérologie, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Olivier Scatton
- Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine, Paris, France.,APHP, Hôpital La Pitié-Salpêtrière, Service de Chirurgie Hépatobiliaire et Transplantation, Paris, France
| | - Olivier Rosmorduc
- APHP, Hôpital La Pitié Salpêtrière, Service d'Hépato-Gastroentérologie, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Sandra Guilmeau
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Catherine Postic
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Chantal Desdouets
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Christèle Desbois-Mouthon
- Sorbonne Universités, UPMC Université Paris 06, INSERM, Centre de Recherche Saint-Antoine, Paris, France
| | - Anne-Françoise Burnol
- Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| |
Collapse
|
9
|
García-Palmero I, Pompas-Veganzones N, Villalobo E, Gioria S, Haiech J, Villalobo A. The adaptors Grb10 and Grb14 are calmodulin-binding proteins. FEBS Lett 2017; 591:1176-1186. [PMID: 28295264 DOI: 10.1002/1873-3468.12623] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/06/2017] [Indexed: 01/24/2023]
Abstract
We identified the Grb7 family members, Grb10 and Grb14, as Ca2+ -dependent CaM-binding proteins using Ca2+ -dependent CaM-affinity chromatography as we previously did with Grb7. The potential CaM-binding sites were identified and experimentally tested using fluorescent-labeled peptides corresponding to these sites. The apparent affinity constant of these peptides for CaM, and the minimum number of calcium ions bound to CaM that are required for effective binding to these peptides were also determined. We prepared deletion mutants of the three adaptor proteins lacking the identified sites and determined that they lost or strongly diminished their CaM-binding capacity following the sequence Grb7 > > Grb14 > Grb10. More than one CaM-binding site and/or accessory CaM-binding sites appear to exist in Grb10 and Grb14, as compared to a single one present in Grb7.
Collapse
Affiliation(s)
- Irene García-Palmero
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain
| | - Noemí Pompas-Veganzones
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain
| | - Eduardo Villalobo
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Spain
| | - Sophie Gioria
- Plate-forme de Chimie Biologique Intégrative de Strasbourg (PCBIS), UMS 3286 CNRS-Université de Strasbourg, France
| | - Jacques Haiech
- Laboratoire d'Excellence Medalis, Université de Strasbourg, CNRS, LIT UMR 7200, France
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Spain
| |
Collapse
|
10
|
Perdereau D, Cailliau K, Browaeys-Poly E, Lescuyer A, Carré N, Benhamed F, Goenaga D, Burnol AF. Insulin-induced cell division is controlled by the adaptor Grb14 in a Chfr-dependent manner. Cell Signal 2015; 27:798-806. [PMID: 25578860 DOI: 10.1016/j.cellsig.2015.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/03/2015] [Indexed: 01/02/2023]
Abstract
Beyond its key role in the control of energy metabolism, insulin is also an important regulator of cell division and neoplasia. However, the molecular events involved in insulin-driven cell proliferation are not fully elucidated. Here, we show that the ubiquitin ligase Chfr, a checkpoint protein involved in G2/M transition, is a new effector involved in the control of insulin-induced cell proliferation. Chfr is identified as a partner of the molecular adapter Grb14, an inhibitor of insulin signalling. Using mammalian cell lines and the Xenopus oocyte as a model of G2/M transition, we demonstrate that Chfr potentiates the inhibitory effect of Grb14 on insulin-induced cell division. Insulin stimulates Chfr binding to the T220 residue of Grb14. Both Chfr binding site and Grb14 C-ter BPS-SH2 domain, mediating IR binding and inhibition, are required to prevent insulin-induced cell division. Targeted mutagenesis revealed that Chfr ligase activity and phosphorylation of its T39 residue, a target of Akt, are required to potentiate Grb14 inhibitory activity. In the presence of insulin, the binding of Chfr to Grb14 activates its ligase activity, leading to Aurora A and Polo-like kinase degradation and blocking cell division. Collectively, our results show that Chfr and Grb14 collaborate in a negative feedback loop controlling insulin-stimulated cell division.
Collapse
Affiliation(s)
- Dominique Perdereau
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Katia Cailliau
- Laboratoire de Régulation des Signaux de Division, Université de Lille 1, UE 4479, IFR 147, Villeneuve d'Ascq 59655, France
| | - Edith Browaeys-Poly
- Laboratoire de Régulation des Signaux de Division, Université de Lille 1, UE 4479, IFR 147, Villeneuve d'Ascq 59655, France
| | - Arlette Lescuyer
- Laboratoire de Régulation des Signaux de Division, Université de Lille 1, UE 4479, IFR 147, Villeneuve d'Ascq 59655, France
| | - Nadège Carré
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Fadila Benhamed
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Diana Goenaga
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France
| | - Anne-Françoise Burnol
- INSERM, U1016, Institut Cochin, Paris, France; CNRS, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité; 24, Rue du Faubourg Saint Jacques, Paris 75014, France.
| |
Collapse
|
11
|
Huang O, Jiang M, Zhang X, Xie Z, Chen X, Wu J, Liu H, Shen K. Grb14 as an independent good prognosis factor for breast cancer patients treated with neoadjuvant chemotherapy. Jpn J Clin Oncol 2013; 43:1064-72. [PMID: 24031083 DOI: 10.1093/jjco/hyt130] [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] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Growth factor receptor-binding protein 14, a new member of noncatalytic adaptor proteins family, has been shown to be upregulated in breast cancer. We investigated the prognostic value of growth factor receptor-binding protein 14 expression in breast cancer patients treated with neoadjuvant chemotherapy. METHODS Primary breast cancer specimens were taken from locally advanced breast cancer patients in a Phase II clinical trial of neoadjuvant chemotherapy and the expression pattern of growth factor receptor-binding protein 14 was determined by immunohistochemistry. Kaplan-Meier analysis and Cox regression model were used to assess disease-free and overall survival, according to the expression of growth factor receptor-binding protein 14 in tumor cells. RESULTS Our result showed that growth factor receptor-binding protein 14 was highly expressed in 23.1% of breast cancer sections, and high expression of growth factor receptor-binding protein 14 was significantly associated with better disease-free (P = 0.016, hazard ratio 0.07, 95% confidence interval 0.06-0.08) and overall survival (P = 0.004, hazard ratio 0.02, 95% confidence interval 0.02-0.03), compared with the low-expression group. Multivariate analysis indicated that high expression of growth factor receptor-binding protein 14 was an independent good prognostic factor for both disease-free (P = 0.04, hazard ratio 0.37, 95% confidence interval 0.14-0.98) and overall survival (P = 0.03, hazard ratio 0.11, 95% confidence interval 0.10-0.82). CONCLUSIONS High expression of growth factor receptor-binding protein 14 in breast cancer cells may help to identify low-risk patients for additional therapies after neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Ou Huang
- *Department of Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, No.197, 2nd Ruijin Road, Huangpu District, Shanghai 200025, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Growth factor receptor-bound protein 14: a potential new gene associated with oocyte competence. ZYGOTE 2013; 22:103-9. [DOI: 10.1017/s0967199413000221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SummaryThe Grb14 protein is a member of the Grb7 protein family. This protein family acts by binding to tyrosine kinase receptors, promoting cell proliferation and differentiation. There is evidence of the involvement of tyrosine kinase factors in the bovine oocyte maturation process. However, Grb14 has not been studied for bovine cumulus–oocyte complexes (COCs). The aim of the present study was to characterize Grb14 mRNA expression in bovine COCs during follicular development. Furthermore, we demonstrated that the expression of Grb14 mRNA is not regulated by estradiol. mRNA expression of Grb14 was assessed in 480 COCs from follicles of different sizes (1–3, 4–6, 6–8 or >8 mm) by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Grb14 mRNA expression decreased in COCs throughout follicular growth (P < 0.05). The role of estradiol in the expression of Grb14 mRNA in COCs was studied. Grb14 mRNA abundance did not differ in COCs cultured in the presence or absence of 17β-estradiol or fulvestrant. In conclusion, we showed that Grb14 mRNA is downregulated in COCs during antral follicle development, a finding that suggests a role for Grb14 in oocyte competence.
Collapse
|
13
|
Burnol AF, Morzyglod L, Popineau L. [Cross-talk between insulin signaling and cell proliferation pathways]. ANNALES D'ENDOCRINOLOGIE 2013; 74:74-8. [PMID: 23582850 DOI: 10.1016/j.ando.2013.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epidemiological studies provide evidence for a close relationship between diabetes and cancer. Insulin is in fact a growth factor, and its binding to its membrane receptor activates intracellular signaling pathways involved in the regulation of both metabolism and cell proliferation. The balance between mitogenic and metabolic actions of insulin can be modulated by various mechanisms, including the way the ligand binds to its receptor or to the closely related insulin-like growth factor-1 (IGF-1) receptor. Cross-talks with other signaling pathways implicated in cell proliferation have also been described, like the Wnt/β catenin pathway, and involve the activation of common downstream effectors such as insulin receptor substrate-1 (IRS-1). Finally, the identification of new proteins activated by insulin and involved in intracellular signaling would allow a better understanding of the complex connections linking metabolic and proliferative regulatory pathways. As an example, the molecular adaptor Grb14, which is a specific inhibitor of insulin receptor catalytic activity, also controls insulin-induced metabolic and mitogenic signaling pathways through post-receptor mechanisms that remain to be fully elucidated.
Collapse
|
14
|
Desbuquois B, Carré N, Burnol AF. Regulation of insulin and type 1 insulin-like growth factor signaling and action by the Grb10/14 and SH2B1/B2 adaptor proteins. FEBS J 2013. [PMID: 23190452 DOI: 10.1111/febs.12080] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The effects of insulin and type 1 insulin-like growth factor (IGF-1) on metabolism, growth and survival are mediated by their association with specific receptor tyrosine kinases, which results in both receptor and substrate phosphorylation. Phosphotyrosine residues on receptors and substrates provide docking sites for signaling proteins containing SH2 (Src homology 2) domains, including molecular adaptors. This review focuses on the regulation of insulin/IGF-1 signaling and action by two adaptor families with a similar domain organization: the growth factor receptor-bound proteins Grb7/10/14 and the SH2B proteins. Both Grb10/14 and SH2B1/B2 associate with the activation loop of insulin/IGF-1 receptors through their SH2 domains, but association of Grb10/14 also involves their unique BPS domain. Consistent with Grb14 binding as a pseudosubstrate to the kinase active site, insulin/IGF-induced activation of receptors and downstream signaling pathways in cultured cells is inhibited by Grb10/14 adaptors, but is potentiated by SH2B1/B2 adaptors. Accordingly, Grb10 and Grb14 knockout mice show improved insulin/IGF sensitivity in vivo, and, for Grb10, overgrowth and increased skeketal muscle and pancreatic β-cell mass. Conversely, SH2B1-depleted mice display insulin and IGF-1 resistance, with peripheral depletion leading to reduced adiposity and neuronal depletion leading to obesity through associated leptin resistance. Grb10/14 and SH2B1 adaptors also modulate insulin/IGF-1 action by interacting with signaling components downstream of receptors and exert several tissue-specific effects. The identification of Grb10/14 and SH2B1 as physiological regulators of insulin signaling and action, together with observations that variants at their gene loci are associated with obesity and/or insulin resistance, highlight them as potential therapeutic targets for these conditions.
Collapse
Affiliation(s)
- Bernard Desbuquois
- Institut Cochin, Départment d'Endocrinologie, Métabolisme et Cancer, Université Paris-Descartes, Institut National de la Santé et de la Recherche Médicale, Unité 1016, et Centre National de la Recherche Scientifique, Unité Mixte de Recherche, Paris, France
| | | | | |
Collapse
|
15
|
Bohrer RC, Rosa PRA, Ferreira R, Bordignon V, Oliveira JFC, Gonçalves PBD. Grb14 mRNA Levels During Follicular Deviation in Cattle are Higher in Granulosa Cells of Subordinate Compared to Dominant Follicles. Reprod Domest Anim 2012; 48:396-401. [DOI: 10.1111/rda.12086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 08/19/2012] [Indexed: 11/26/2022]
Affiliation(s)
| | - PRA Rosa
- Laboratory of Biotechnology and Animal Reproduction, BioRep; Federal University of Santa Maria; Santa Maria; Rio Grande do Sul; Brazil
| | - R Ferreira
- Department of Animal Science; Santa Catarina State University; Chapecó; SC; Brazil
| | - V Bordignon
- Department of Animal Science; McGill University; Ste-Anne-de-Bellevue; QC; Canada
| | - JFC Oliveira
- Laboratory of Biotechnology and Animal Reproduction, BioRep; Federal University of Santa Maria; Santa Maria; Rio Grande do Sul; Brazil
| | - PBD Gonçalves
- Laboratory of Biotechnology and Animal Reproduction, BioRep; Federal University of Santa Maria; Santa Maria; Rio Grande do Sul; Brazil
| |
Collapse
|
16
|
Graae L, Karlsson R, Paddock S. Significant association of estrogen receptor binding site variation with bipolar disorder in females. PLoS One 2012; 7:e32304. [PMID: 22389694 PMCID: PMC3289647 DOI: 10.1371/journal.pone.0032304] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 01/26/2012] [Indexed: 12/22/2022] Open
Abstract
Major depression is nearly twice as prevalent in women compared to men. In bipolar disorder, depressive episodes have been reported to be more common amongst female patients. Furthermore, periods of depression often correlate with periods of hormonal fluctuations. A link between hormone signaling and these mood disorders has, therefore, been suggested to exist in many studies. Estrogen, one of the primary female sex hormones, mediates its effect mostly by binding to estrogen receptors (ERs). Nuclear ERs function as transcription factors and regulate gene transcription by binding to specific DNA sequences. A nucleotide change in the binding sequence might alter the binding efficiency, which could affect transcription levels of nearby genes. In order to investigate if variation in ER DNA-binding sequences may be involved in mood disorders, we conducted a genome-wide study of ER DNA-binding in patients diagnosed with major depression or bipolar disorder. Association studies were performed within each gender separately and the results were corrected for multiple testing by the Bonferroni method. In the female bipolar disorder material a significant association result was found for rs6023059 (corrected p-value = 0.023; odds ratio (OR) 0.681, 95% confidence interval (CI) 0.570–0.814), a single nucleotide polymorphism (SNP) placed downstream of the gene coding for transglutaminase 2 (TGM2). Thus, females with a specific genotype at this SNP may be more vulnerable to fluctuating estrogen levels, which may then act as a triggering factor for bipolar disorder.
Collapse
Affiliation(s)
- Lisette Graae
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | |
Collapse
|
17
|
Giricz O, Calvo V, Pero SC, Krag DN, Sparano JA, Kenny PA. GRB7 is required for triple-negative breast cancer cell invasion and survival. Breast Cancer Res Treat 2011; 133:607-15. [PMID: 22005836 DOI: 10.1007/s10549-011-1822-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 10/06/2011] [Indexed: 11/28/2022]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease that is usually associated with poor prognosis, and frequently associated with the basal-like breast cancer gene expression profile. There are no targeted therapeutic modalities for this disease, and no useful biomarkers. High GRB7 RNA expression levels are associated with an elevated risk of recurrence in patients with operable TNBC treated with standard adjuvant anthracycline and taxane therapy. To determine whether GRB7 is involved in the pathobiology of TNBC, we evaluated the biological effects of GRB7 inhibition in a panel of triple-negative cell lines-MDA-MB-468, MDA-MB-231, HCC70, and T4-2. We found GRB7 inhibition reduced cell motility and invasion of these cell lines and promoted cell death by apoptosis in 3D culture. These data suggest that GRB7 itself, or GRB7-dependent pathways, may prove to be important therapeutic targets in this disease.
Collapse
Affiliation(s)
- Orsi Giricz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | | | |
Collapse
|
18
|
Lucas-Fernández E, García-Palmero I, Villalobo A. Genomic organization and control of the grb7 gene family. Curr Genomics 2011; 9:60-8. [PMID: 19424485 PMCID: PMC2674303 DOI: 10.2174/138920208783884847] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 02/20/2008] [Accepted: 02/21/2008] [Indexed: 11/22/2022] Open
Abstract
Grb7 and their related family members Grb10 and Grb14 are adaptor proteins, which participate in the functionality of multiple signal transduction pathways under the control of a variety of activated tyrosine kinase receptors and other tyrosine-phosphorylated proteins. They are involved in the modulation of important cellular and organismal functions such as cell migration, cell proliferation, apoptosis, gene expression, protein degradation, protein phosphorylation, angiogenesis, embryonic development and metabolic control. In this short review we shall describe the organization of the genes encoding the Grb7 protein family, their transcriptional products and the regulatory mechanisms implicated in the control of their expression. Finally, the alterations found in these genes and the mechanisms affecting their expression under pathological conditions such as cancer, diabetes and some congenital disorders will be highlighted.
Collapse
Affiliation(s)
- E Lucas-Fernández
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid. Arturo Duperier 4, E-28029 Madrid, Spain
| | | | | |
Collapse
|
19
|
Heinonen M, Hemmes A, Salmenkivi K, Abdelmohsen K, Vilén ST, Laakso M, Leidenius M, Salo T, Hautaniemi S, Gorospe M, Heikkilä P, Haglund C, Ristimäki A. Role of RNA binding protein HuR in ductal carcinoma in situ of the breast. J Pathol 2011; 224:529-39. [PMID: 21480233 DOI: 10.1002/path.2889] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/21/2011] [Accepted: 02/28/2011] [Indexed: 01/26/2023]
Abstract
HuR is a ubiquitously expressed RNA-binding protein that modulates gene expression at the post-transcriptional level. It is predominantly nuclear, but can shuttle between the nucleus and the cytoplasm. While in the cytoplasm HuR can stabilize its target transcripts, many of which encode proteins involved in carcinogenesis. While cytoplasmic HuR expression is a marker of reduced survival in breast cancer, its role in precursor lesions of malignant diseases is unclear. To address this we explored HuR expression in atypical ductal hyperplasia (ADH) and in ductal in situ carcinomas (DCIS). We show that cytoplasmic HuR expression is elevated in both ADH and DCIS when compared to normal controls, and that this expression associated with high grade, progesterone receptor negativity and microinvasion and/or tumour-positive sentinel nodes of the DCIS. To study the mechanisms of HuR in breast carcinogenesis, HuR expression was silenced in an immortalized breast epithelial cell line (184B5Me), which led to reduction in anchorage-independent growth, increased programmed cell death and inhibition of invasion. In addition, we identified two novel target transcripts (CTGF and RAB31) that are regulated by HuR and that bind HuR protein in this cell line. Our results show that HuR is aberrantly expressed at early stages of breast carcinogenesis and that its inhibition can lead to suppression of this process. ArrayExpress Accession No. E-MEXP-3035.
Collapse
Affiliation(s)
- Mira Heinonen
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Finland; Genome-Scale Biology, Research Program Unit, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Russo J, Russo IH. The role of the basal stem cell of the human breast in normal development and cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 720:121-34. [PMID: 21901623 DOI: 10.1007/978-1-4614-0254-1_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
MCF-10F, an ERα negative human breast epithelial cell line derived from normal breast tissue, is able to form ductal structures in a tridimensional collagen matrix system. MCF-10F cells that are estrogen transformed (trMCF cells) progressively express phenotypes of in vitro cell transformation, including colony formation in agar methocel and loss of the ductulogenic capacity. Selection of these trMCF cells for invasiveness identified cells (bcMCF) that formed tumors in severe combined immunodeficient mice. The cell lines derived from those tumors (caMCF) were poorly differentiated ER, PR, and ERBB2 negative adenocarcinomas. These characteristics are similar to the human basal cell-like carcinomas. This in vitro-in vivo model demonstrates the importance of the basal cell type as a stem cell that reconstitutes the branching pattern of the breast and that is also target of a carcinogenic insult leading to transformation and cancer.
Collapse
Affiliation(s)
- Jose Russo
- Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | |
Collapse
|
21
|
Spink BC, Bennett JA, Pentecost BT, Lostritto N, Englert NA, Benn GK, Goodenough AK, Turesky RJ, Spink DC. Long-term estrogen exposure promotes carcinogen bioactivation, induces persistent changes in gene expression, and enhances the tumorigenicity of MCF-7 human breast cancer cells. Toxicol Appl Pharmacol 2009; 240:355-66. [PMID: 19619570 DOI: 10.1016/j.taap.2009.07.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Revised: 07/14/2009] [Accepted: 07/15/2009] [Indexed: 12/21/2022]
Abstract
The cumulative exposure to estrogens is an important determinant in the risk of breast cancer, yet the full range of mechanisms involving estrogens in the genesis and progression of breast cancer remains a subject of debate. Interactions of estrogens and environmental toxicants have received attention as putative factors contributing to carcinogenesis. Mechanistic studies have demonstrated interactions between estrogen receptor alpha (ERalpha) and the aryl hydrocarbon receptor (AhR), with consequences on the genes that they regulate. Many studies of ERalpha and AhR-mediated effects and crosstalk between them have focused on the initial molecular events. In this study, we investigated ERalpha- and AhR-mediated effects in long-term estrogen exposed (LTEE) MCF-7 human breast cancer cells, which were obtained by continuous culturing for at least 12 weeks in medium supplemented with 1 nM of 17beta-estradiol (E(2)). With these LTEE cells and with parallel control cells cultured without E(2) supplementation, we performed an extensive study of cytochrome P450 (CYP) induction, carcinogen bioactivation, global gene expression, and tumorigenicity in immunocompromised mice. We found that LTEE cells, in comparison with control cells, had higher levels of AhR mRNA and protein, greater responsiveness for AhR-regulated CYP1A1 and CYP1B1 induction, a 6-fold higher initial level of benzo(a)pyrene-DNA adducts as determined by liquid chromatography tandem mass spectrometry, marked differences in the expression of numerous genes, and a higher rate of E(2)-dependent tumor growth as xenografts. These studies indicate that LTEE causes adaptive responses in MCF-7 cells, which may reflect processes that contribute to the overall carcinogenic effect of E(2).
Collapse
Affiliation(s)
- Barbara C Spink
- Laboratory of Molecular Toxicology, Wadsworth Center, New York State Department of Health, Albany, NY 12201-0509, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Goenaga D, Hampe C, Carré N, Cailliau K, Browaeys-Poly E, Perdereau D, Holt LJ, Daly RJ, Girard J, Broutin I, Issad T, Burnol AF. Molecular determinants of Grb14-mediated inhibition of insulin signaling. Mol Endocrinol 2009; 23:1043-51. [PMID: 19359342 DOI: 10.1210/me.2008-0360] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Grb14 belongs to the Grb7 family of molecular adapters and was identified as an inhibitor of insulin signaling. Grb14 binds to activated insulin receptors (IR) and inhibits their catalytic activity. To gain more insight into the Grb14 molecular mechanism of action, we generated various mutants and studied the Grb14-IR interaction using coimmunoprecipitation and bioluminescence resonance energy transfer (BRET) experiments. Biological activity was further analyzed using the Xenopus oocyte model and a functional complementation assay measuring cellular proliferation rate in Grb14 knockout mouse embryonic fibroblasts. These studies identified two important interaction sites, Grb14 L404-IR L1038 and Grb14 R385-IR K1168, involving the IR alphaC-helix and activation loop, respectively. Interestingly, the former involves residues that are likely to be crucial for the specificity of IR binding with regard to other members of the Grb7 family. In addition, mutation of the Grb14-S370 residue suggested that its phosphorylation status controlled the biological activity of the protein. We further demonstrated that insulin-induced Grb14-PDK1 interaction is required in addition to Grb14-IR binding to mediate maximal inhibition of insulin signaling. This study provides important insights into the molecular determinants of Grb14 action by demonstrating that Grb14 regulates insulin action at two levels, through IR binding and by interfering with downstream pathways. Indeed, a precise knowledge of the molecular mechanism of insulin signaling inhibition by Grb14 is a prerequisite for the development of insulin-sensitizing molecules to treat pathophysiological states such as obesity or type 2 diabetes.
Collapse
Affiliation(s)
- Diana Goenaga
- Institut Cochin, Université Paris Descartes, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8104, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
van Agthoven T, Veldscholte J, Smid M, van Agthoven TLA, Vreede L, Broertjes M, de Vries I, de Jong D, Sarwari R, Dorssers LCJ. Functional identification of genes causing estrogen independence of human breast cancer cells. Breast Cancer Res Treat 2008; 114:23-30. [PMID: 18351453 DOI: 10.1007/s10549-008-9969-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2008] [Accepted: 03/05/2008] [Indexed: 01/24/2023]
Abstract
Endocrine treatment of breast cancer is widely applied and effective. However, in advanced disease cases, the tumors will eventually progress into an estrogen-independent and therapy-resistant phenotype. To elucidate the molecular mechanisms underlying this endocrine therapy failure, we applied retroviral insertion mutagenesis to identify the main genes conferring estrogen independence to human breast cancer cells. Estrogen-dependent ZR-75-1 cells were infected with replication-defective retroviruses followed by selection with the anti-estrogen 4-hydroxy-tamoxifen. In the resulting panel of 79 tamoxifen-resistant cell lines, the viral integrations were mapped within the human genome. Genes located in the immediate proximity of the retroviral integration sites were characterized for altered expression and their capacity to confer anti-estrogen resistance when transfected into breast cancer cells. Out of 15 candidate BCAR (breast cancer anti-estrogen resistance) genes, seven (AKT1, AKT2, BCAR1, BCAR3, EGFR, GRB7, and TRERF1/BCAR2) were shown to directly underlie estrogen independence. Our results show that insertion mutagenesis is a powerful tool to identify BCAR loci, which may provide insights into the molecular and cellular mechanisms of breast tumor progression and therapy resistance thereby offering novel targets for the development of tailor-made therapeutical and prevention strategies.
Collapse
Affiliation(s)
- Ton van Agthoven
- Department of Pathology, Josephine Nefkens Institute, Be 432, Erasmus MC-University Medical Center Rotterdam, P.O. Box 2040, Rotterdam, CA, 3000, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Woodmansee WW, Kerr JM, Tucker EA, Mitchell JR, Haakinson DJ, Gordon DF, Ridgway EC, Wood WM. The proliferative status of thyrotropes is dependent on modulation of specific cell cycle regulators by thyroid hormone. Endocrinology 2006; 147:272-82. [PMID: 16223861 DOI: 10.1210/en.2005-1013] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In this report we have examined changes in cell growth parameters, cell cycle effectors, and signaling pathways that accompany thyrotrope growth arrest by thyroid hormone (TH) and growth resumption after its withdrawal. Flow cytometry and immunohistochemistry of proliferation markers demonstrated that TH treatment of thyrotrope tumors resulted in a reduction in the fraction of cells in S-phase that is restored upon TH withdrawal. This is accompanied by dephosphorylation and rephosphorylation of retinoblastoma (Rb) protein. The expression levels of cyclin-dependent kinase 2 and cyclin A, as well as cyclin-dependent kinase 1 and cyclin B, were decreased by TH, and after withdrawal not only did these regulators of Rb phosphorylation and mitosis increase in their expression but so too did the D1 and D3 cyclins. We also noted a rapid induction and subsequent disappearance of the type 5 receptor for the growth inhibitor somatostatin with TH treatment and withdrawal, respectively. Because somatostatin can arrest growth by activating MAPK pathways, we examined these pathways in TtT-97 tumors and found that the ERK pathway and several of its upstream and downstream effectors, including cAMP response element binding protein, were activated with TH treatment and deactivated after its withdrawal. This led to the hypothesis that TH, acting through increased type 5 somatostatin receptor, could activate the ERK pathway leading to cAMP response element binding protein-dependent decreased expression of critical cell cycle proteins, specifically cyclin A, resulting in hypophosphorylation of Rb and its subsequent arrest of S-phase progression. These processes are reversed when TH is withdrawn, resulting in an increase in the fraction of S-phase cells.
Collapse
Affiliation(s)
- Whitney W Woodmansee
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado at Denver and Health Sciences Center, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kasukabe T, Okabe-Kado J, Kato N, Sassa T, Honma Y. Effects of combined treatment with rapamycin and cotylenin A, a novel differentiation-inducing agent, on human breast carcinoma MCF-7 cells and xenografts. Breast Cancer Res 2005; 7:R1097-110. [PMID: 16457690 PMCID: PMC1410757 DOI: 10.1186/bcr1344] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 09/22/2005] [Accepted: 10/06/2005] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Rapamycin, an inhibitor of the serine/threonine kinase target of rapamycin, induces G1 arrest and/or apoptosis. Although rapamycin and its analogues are attractive candidates for cancer therapy, their sensitivities with respect to growth inhibition differ markedly among various cancer cells. Using human breast carcinoma cell line MCF-7 as an experimental model system, we examined the growth-inhibitory effects of combinations of various agents and rapamycin to find the agent that most potently enhances the growth-inhibitory effect of rapamycin. METHOD We evaluated the growth-inhibitory effect of rapamycin plus various agents, including cotylenin A (a novel inducer of differentiation of myeloid leukaemia cells) to MCF-7 cells, using either MTT assay or trypan blue dye exclusion test. The cell cycle was analyzed using propidium iodide-stained nuclei. Expressions of several genes in MCF-7 cells with rapamycin plus cotylenin A were studied using cDNA microarray analysis and RT-PCR. The in vitro results of MCF-7 cells treated with rapamycin plus cotylenin A were further confirmed in vivo in a mouse xenograft model. RESULTS We found that the sensitivity of rapamycin to MCF-7 cells was markedly affected by cotylenin A. This treatment induced growth arrest of the cells at the G1 phase, rather than apoptosis, and induced senescence-associated beta-galactosidase activity. We examined the gene expression profiles associated with exposure to rapamycin and cotylenin A using cDNA microarrays. We found that expressions of cyclin G2, transforming growth factor-beta-induced 68 kDa protein, BCL2-interacting killer, and growth factor receptor-bound 7 were markedly induced in MCF-7 cells treated with rapamycin plus cotylenin A. Furthermore, combined treatment with rapamycin and cotylenin A significantly inhibited the growth of MCF-7 cells as xenografts, without apparent adverse effects. CONCLUSION Rapamycin and cotylenin A cooperatively induced growth arrest in breast carcinoma MCF-7 cells in vitro, and treatment with rapamycin and cotylenin A combined more strongly inhibited the growth of MCF-7 cells as xenografts in vivo than treatment with rapamycin or cotylenin A alone, suggesting that this combination may have therapeutic value in treating breast cancer. We also identified several genes that were markedly modulated in MCF-7 cells treated with rapamycin plus cotylenin A.
Collapse
Affiliation(s)
- Takashi Kasukabe
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Junko Okabe-Kado
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
| | - Nobuo Kato
- The Institute of Scientific and Industrial Research, Osaka University, Osaka, Japan
| | - Takeshi Sassa
- Department of Bioresource Engineering, Yamagata University, Tsuruoka, Japan
| | - Yoshio Honma
- Research Institute for Clinical Oncology, Saitama Cancer Center, Saitama, Japan
- School of Medicine, Shimane University, Izumo, Japan
| |
Collapse
|
26
|
Abstract
The Grb proteins (growth factor receptor-bound proteins) Grb7, Grb10 and Grb14 constitute a family of structurally related multidomain adapters with diverse cellular functions. Grb10 and Grb14, in particular, have been implicated in the regulation of insulin receptor signalling, whereas Grb7 appears predominantly to be involved in focal adhesion kinase-mediated cell migration. However, at least in vitro, these adapters can bind to a variety of growth factor receptors. The highest identity within the Grb7/10/14 family occurs in the C-terminal SH2 (Src homology 2) domain, which mediates binding to activated receptors. A second well-conserved binding domain, BPS [between the PH (pleckstrin homology) and SH2 domains], can act to enhance binding to the IR (insulin receptor). Consistent with a putative adapter function, some non-receptor-binding partners, including protein kinases, have also been identified. Grb10 and Grb14 are widely, but not uniformly, expressed in mammalian tissues, and there are various isoforms of Grb10. Binding of Grb10 or Grb14 to autophosphorylated IR in vitro inhibits tyrosine kinase activity towards other substrates, but studies on cultured cell lines have been conflicting as to whether Grb10 plays a positive or negative role in insulin signalling. Recent gene knockouts in mice have established that Grb10 and Grb14 act as inhibitors of intracellular signalling pathways regulating growth and metabolism, although the phenotypes of the two knockouts are distinct. Ablation of Grb14 enhances insulin action in liver and skeletal muscle and improves whole-body tolerance, with little effect on embryonic growth. Ablation of Grb10 results in disproportionate overgrowth of the embryo and placenta involving unidentified pathways, and also impacts on hepatic glycogen synthesis, and probably on glucose homoeostasis. This review discusses the extent to which previous studies in vitro can account for the observed phenotype of knockout animals, and considers evidence that aberrant function of Grb10 or Grb14 may contribute to disorders of growth and metabolism in humans.
Collapse
Affiliation(s)
- Lowenna J Holt
- University of Cambridge, Department of Clinical Biochemistry, Addenbrooke's Hospital, Cambridge CB2 2QR, UK.
| | | |
Collapse
|
27
|
Affiliation(s)
- Lowenna J Holt
- Garvan Institute of Medical Research, St Vincent's Hospital, Cancer Research Program, Sydney, NSW, Australia
| | | |
Collapse
|