1
|
Archetti M. Collapse of Intra-Tumor Cooperation Induced by Engineered Defector Cells. Cancers (Basel) 2021; 13:cancers13153674. [PMID: 34359576 PMCID: PMC8345189 DOI: 10.3390/cancers13153674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 11/16/2022] Open
Abstract
Anti-cancer therapies promote clonal selection of resistant cells that evade treatment. Effective therapy must be stable against the evolution of resistance. A potential strategy based on concepts from evolutionary game theory is to impair intra-tumor cooperation using genetically modified cells in which genes coding for essential growth factors have been knocked out. Such engineered cells would spread by clonal selection, driving the collapse of intra-tumor cooperation and a consequent reduction in tumor growth. Here, I test this idea in vitro in four cancer types (neuroendocrine pancreatic cancer, mesothelioma, lung adenocarcinoma and multiple myeloma). A reduction, or even complete eradication, of the producer clone and the consequent reduction in cell proliferation, is achieved in some but not all cases by introducing a small fraction of non-producer cells in the population. I show that the collapse of intra-tumor cooperation depends on the cost/benefit ratio of growth factor production. When stable cooperation among producer and non-producer cells occurs, its collapse can be induced by increasing the number of growth factors available to the cells. Considerations on nonlinear dynamics in the framework of evolutionary game theory explain this as the result of perturbation of the equilibrium of a system that resembles a public goods game, in which the production of growth factors is a cooperative phenotype. Inducing collapse of intra-tumor cooperation by engineering cancer cells will require the identification of growth factors that are essential for the tumor and that have a high cost of production for the cell.
Collapse
Affiliation(s)
- Marco Archetti
- Department of Biology, Pennsylvania State University, University Park, State College, PA 16802, USA
| |
Collapse
|
2
|
Zhang H, Wang Y, Zhang W, Wu Q, Fan J, Zhan Q. BAALC-AS1/G3BP2/c-Myc feedback loop promotes cell proliferation in esophageal squamous cell carcinoma. Cancer Commun (Lond) 2021; 41:240-257. [PMID: 33476486 PMCID: PMC7968881 DOI: 10.1002/cac2.12127] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 12/16/2020] [Indexed: 12/15/2022] Open
Abstract
Background Long non‐coding RNAs (lncRNAs) have been found to be involved in the development of many cancers. In this study, we aimed to identify the molecular mechanisms of lncRNA BAALC antisense RNA 1 (BAALC‐AS1) in regulating the malignancy of esophageal squamous cell carcinoma (ESCC). Methods The expression of BAALC‐AS1 in cancer patients was analyzed using a tissue microarray. The protein and RNA levels of BAALC‐AS1 were determined by Western blotting analysis and quantitative reverse transcription‐PCR (RT‐qPCR), respectively. The cell proliferation was determined by cell viability assays, bromodeoxyuridine incorporation, and flow cytometry. The relationships among BAALC‐AS1, RasGAPSH3 domain‐binding protein 2 (G3BP2), and c‐Myc were determined using RNA immunoprecipitation, RNA pull‐down assays, and luciferase assays. Results The expression of BAALC‐AS1 was highly up‐regulated and associated with malignant phenotypes in ESCC tissues and cell lines. In vivo and in vitro assays showed that BAALC‐AS1 promoted ESCC cell proliferation, migration, and invasion. BAALC‐AS1 directly interacted with G3BP2, and thereby inhibited the degradation of c‐Myc RNA 3'‐UTR by G3BP2, thus leading to the accumulation of c‐Myc expression. Additionally, c‐Myc acted as a transcription factor that can induce the expression of BAALC‐AS1 by directly binding to its promoter region. Conclusions BAALC‐AS1/G3BP2/c‐Myc feedback loop plays a critical role in the development of ESCC, which might provide a novel therapeutic target and facilitate the development of new therapeutic strategies for the treatment of ESCC.
Collapse
Affiliation(s)
- Hongyue Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Weimin Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Jiawen Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, P. R. China.,Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518132, P. R. China
| |
Collapse
|
3
|
Matrood S, de Prisco N, Wissniowski TT, Wiese D, Jabari S, Griesmann H, Wanzel M, Stiewe T, Neureiter D, Klieser E, Mintziras I, Buchholz M, Bartsch DK, Gennarino VA, Di Fazio P. Modulation of Pancreatic Neuroendocrine Neoplastic Cell Fate by Autophagy-Mediated Death. Neuroendocrinology 2021; 111:965-985. [PMID: 33108790 DOI: 10.1159/000512567] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/02/2020] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Autophagic cell death in cancer cells can be mediated by inhibition of deacetylases. Although extensive studies have focused on the autophagic process in cancer, little is known about the role of autophagy in degrading cytosolic and nuclear components of pancreatic neuroendocrine neoplastic (pNEN) cells leading to cell death, thus improving the therapy of patients affected by pNEN. METHODS 2D and 3D human pNEN and pancreatic stellate cells were treated with panobinostat and bafilomycin. Autophagy markers were detected by RT-qPCR, immunofluorescence, and Western blot. Autophagosomes were detected by electron microscopy and their maturation by real-time fluorescence of LC3B stable transfected cells. ChIP was performed at the cAMP responsive element. Immunofluorescence was performed in murine pancreatic tissue. RESULTS We observed that pan-deacetylase inhibitor panobinostat treatment causes autophagic cell death in pNEN cells. We also found that although AMPK-α phosphorylation is counterbalanced by phosphorylated AKT, it is not capable to inhibiting autophagic cell death. However, the binding activity of the cAMP responsive element is prompted by panobinostat. Although autophagy inhibition prevented autophagosome synthesis, maturation, and cell death, panobinostat treatment induced the accumulation of mature autophagosomes in the cytosol and the nucleus, leading to disruption of the organelles, cellular digestion, and decay. Observation of autophagosome membrane proteins Beclin1 and LC3B aggregation in murine pancreatic islets indicates that autophagy restoration may also lead to autophagosome aggregation in murine insulinoma cells. A basal low expression of autophagy markers was detectable in patients affected by pNEN, and, interestingly, the expression of these markers was significantly lower in metastatic pNEN. DISCUSSION/CONCLUSION Our study highlights that the autophagy functional restoration and prolongation of this catabolic process, mediated by inhibition of deacetylase, is responsible for the reduction of pNEN cells. Prompting of autophagy cell death could be a promising strategy for the therapy of pNEN.
Collapse
Affiliation(s)
- Sami Matrood
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Nicola de Prisco
- Departments of Genetics and Development, Pediatrics and Neurology, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Dominik Wiese
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Samir Jabari
- Institute of Anatomy I, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Heidi Griesmann
- Department of Internal Medicine I, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Michael Wanzel
- Institute for Molecular Oncology, Member of the German Center for Lung Research, Philipps University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Institute for Molecular Oncology, Member of the German Center for Lung Research, Philipps University Marburg, Marburg, Germany
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria
| | - Eckhard Klieser
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg, Austria
| | - Ioannis Mintziras
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Malte Buchholz
- Department of Gastroenterology, Philipps University Marburg, Marburg, Germany
| | - Detlef K Bartsch
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany
| | - Vincenzo A Gennarino
- Departments of Genetics and Development, Pediatrics and Neurology, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, New York, USA
| | - Pietro Di Fazio
- Department of Visceral Thoracic and Vascular Surgery, Philipps University Marburg, Marburg, Germany,
| |
Collapse
|
4
|
The IGF-II-Insulin Receptor Isoform-A Autocrine Signal in Cancer: Actionable Perspectives. Cancers (Basel) 2020; 12:cancers12020366. [PMID: 32033443 PMCID: PMC7072655 DOI: 10.3390/cancers12020366] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 12/18/2022] Open
Abstract
Insulin receptor overexpression is a common event in human cancer. Its overexpression is associated with a relative increase in the expression of its isoform A (IRA), a shorter variant lacking 11 aa in the extracellular domain, conferring high affinity for the binding of IGF-II along with added intracellular signaling specificity for this ligand. Since IGF-II is secreted by the vast majority of malignant solid cancers, where it establishes autocrine stimuli, the co-expression of IGF-II and IRA in cancer provides specific advantages such as apoptosis escape, growth, and proliferation to those cancers bearing such a co-expression pattern. However, little is known about the exact role of this autocrine ligand–receptor system in sustaining cancer malignant features such as angiogenesis, invasion, and metastasis. The recent finding that the overexpression of angiogenic receptor kinase EphB4 along with VEGF-A is tightly dependent on the IGF-II/IRA autocrine system independently of IGFIR provided new perspectives for all malignant IGF2omas (those aggressive solid cancers secreting IGF-II). The present review provides an updated view of the IGF system in cancer, focusing on the biology of the autocrine IGF-II/IRA ligand–receptor axis and supporting its underscored role as a malignant-switch checkpoint target.
Collapse
|
5
|
Duvillié B, Kourdoughli R, Druillennec S, Eychène A, Pouponnot C. Interplay Between Diabetes and Pancreatic Ductal Adenocarcinoma and Insulinoma: The Role of Aging, Genetic Factors, and Obesity. Front Endocrinol (Lausanne) 2020; 11:563267. [PMID: 33101198 PMCID: PMC7556217 DOI: 10.3389/fendo.2020.563267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
Epidemiologic analyses have shed light on an association between type 2 diabetes (T2D) and pancreatic ductal adenocarcinoma (PDAC). Recent data also suggest a potential relationship between T2D and insulinoma. Under rare circumstances, type 1 diabetes (T1D) can also be implicated in tumorigenesis. The biological mechanisms underlying such relationships are extremely complex. Some genetic factors contributing to the development of T2D are shared with pancreatic exocrine and endocrine tumors. Obesity and overweight can also contribute to the initiation and severity of T2D, while aging may influence both endocrine and exocrine tumors. Finally, pharmacological treatments of T2D may have an impact on PDAC. On the other hand, some treatments for insulinoma can trigger diabetes. In the present minireview, we discuss the cellular and molecular mechanisms that could explain these interactions. This analysis may help to define new potential therapeutic strategies.
Collapse
Affiliation(s)
- Bertrand Duvillié
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
- *Correspondence: Bertrand Duvillié,
| | - Rayane Kourdoughli
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Sabine Druillennec
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Alain Eychène
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Celio Pouponnot
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| |
Collapse
|
6
|
Wang S, Gribskov M. Transcriptome analysis identifies metallothionein as biomarkers to predict recurrence in hepatocellular cacinoma. Mol Genet Genomic Med 2019; 7:e693. [PMID: 31056863 PMCID: PMC6565558 DOI: 10.1002/mgg3.693] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/17/2019] [Accepted: 03/14/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Liver cancer is the fifth most common cancer, and hepatocellular carcinoma (HCC) is the major liver tumor type seen in adults. HCC is usually caused by chronic liver disease such as hepatitis B virus or hepatitis C virus infection. One of the promising treatments for HCC is liver transplantation, in which a diseased liver is replaced with a healthy liver from another person. However, recurrence of HCC after surgery is a significant problem. Therefore, it is important to discover reliable cellular biomarkers that can predict recurrence in HCC. METHODS We analyzed previously published HCC RNA-Seq data that includes 21 paired tumor and normal samples, in which nine tumors were recurrent after orthotopic liver transplantation and 12 were nonrecurrent tumors with their paired normal samples. We used both the reference genome and de novo transcriptome assembly based analyses to identify differentially expressed genes (DEG) and used RandomForest to discover biomarkers. RESULTS We obtained 398 DEG using the Reference approach and 412 DEG using de novo assembly approach. Among these DEG, 258 genes were identified by both approaches. We further identified 30 biomarkers that could predict the recurrence. We used another independent HCC study that includes 50 patients normal and tumor samples. By using these 30 biomarkers, the prediction accuracy was 100% for normal condition and 98% for tumor condition. A group of Metallothionein was specifically discovered as biomarkers in both reference and de novo assembly approaches. CONCLUSION We identified a group of Metallothionein genes as biomarkers to predict recurrence. The metallothionein genes were all down-regulated in tumor samples, suggesting that low metallothionein expression may be a promoter of tumor growth. In addition, using de novo assembly identified some unique biomarkers, further confirmed the necessity of conducting a de novo assembly in human cancer study.
Collapse
Affiliation(s)
- Sufang Wang
- School of Life SciencesNorthwestern Polytechnical UniversityXi'anShaanxiChina
- Center of Special Environmental Biomechanics & Biomedical EngineeringNorthwestern Polytechnical UniversityXi'anShaanxiChina
| | - Michael Gribskov
- Department of Biological SciencesPurdue UniversityWest LafayetteIndianaUSA
- Department of Computer SciencesPurdue UniversityWest LafayetteIndianaUSA
| |
Collapse
|
7
|
Ma C, Luo H, Liu B, Li F, Tschöpe C, Fa X. Long noncoding RNAs: A new player in the prevention and treatment of diabetic cardiomyopathy? Diabetes Metab Res Rev 2018; 34:e3056. [PMID: 30160026 DOI: 10.1002/dmrr.3056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/12/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022]
Abstract
Diabetic cardiomyopathy (DCM) can cause extensive necrosis of the heart muscle by metabolic disorders and microangiopathy, with subclinical cardiac dysfunction, and eventually progress to heart failure, arrhythmia, and cardiogenic shock; severe patients may even die suddenly. Long noncoding RNAs (lncRNAs) are a class of nonprotein-coding RNAs longer than 200 nucleotides. They have critical roles in various biological processes, including gene expression regulation, genomic imprinting, nuclear-cytoplasmic trafficking, RNA splicing, and translational control. Recent studies indicated that lncRNAs extensively participate in the development of diverse cardiac diseases, such as cardiac ischaemia, hypertrophy, and heart failure. Little is known about lncRNA in DCM. In this review, we summarize the current literature on lncRNAs in DCM studies, aiming to provide new methods for DCM's future prevention and treatment strategies.
Collapse
Affiliation(s)
- Chao Ma
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Cardiology, Campus Virchow, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Huan Luo
- Department of Ophthalmology, Campus Virchow, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bing Liu
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Thoracic Surgery, Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow, Charité-Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Xianen Fa
- Department of Cardiovascular Surgery, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Evans JR, Feng FY, Chinnaiyan AM. The bright side of dark matter: lncRNAs in cancer. J Clin Invest 2016; 126:2775-82. [PMID: 27479746 DOI: 10.1172/jci84421] [Citation(s) in RCA: 341] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The traditional view of genome organization has been upended in the last decade with the discovery of vast amounts of non-protein-coding transcription. After initial concerns that this "dark matter" of the genome was transcriptional noise, it is apparent that a subset of these noncoding RNAs are functional. Long noncoding RNA (lncRNA) genes resemble protein-coding genes in several key aspects, and they have myriad molecular functions across many cellular pathways and processes, including oncogenic signaling. The number of lncRNA genes has recently been greatly expanded by our group to triple the number of protein-coding genes; therefore, lncRNAs are likely to play a role in many biological processes. Based on their large number and expression specificity in a variety of cancers, lncRNAs are likely to serve as the basis for many clinical applications in oncology.
Collapse
|
9
|
Trump BF. Mechanisms of Toxicity and Carcinogenesis. Toxicol Pathol 2016. [DOI: 10.1177/019262339502300616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
10
|
Modi H, Jacovetti C, Tarussio D, Metref S, Madsen OD, Zhang FP, Rantakari P, Poutanen M, Nef S, Gorman T, Regazzi R, Thorens B. Autocrine Action of IGF2 Regulates Adult β-Cell Mass and Function. Diabetes 2015; 64:4148-57. [PMID: 26384384 DOI: 10.2337/db14-1735] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 08/30/2015] [Indexed: 11/13/2022]
Abstract
Insulin-like growth factor 2 (IGF2), produced and secreted by adult β-cells, functions as an autocrine activator of the β-cell insulin-like growth factor 1 receptor signaling pathway. Whether this autocrine activity of IGF2 plays a physiological role in β-cell and whole-body physiology is not known. Here, we studied mice with β-cell-specific inactivation of Igf2 (βIGF2KO mice) and assessed β-cell mass and function in aging, pregnancy, and acute induction of insulin resistance. We showed that glucose-stimulated insulin secretion (GSIS) was markedly reduced in old female βIGF2KO mice; glucose tolerance was, however, normal because of increased insulin sensitivity. While on a high-fat diet, both male and female βIGF2KO mice displayed lower GSIS compared with control mice, but reduced β-cell mass was observed only in female βIGF2KO mice. During pregnancy, there was no increase in β-cell proliferation and mass in βIGF2KO mice. Finally, β-cell mass expansion in response to acute induction of insulin resistance was lower in βIGF2KO mice than in control mice. Thus, the autocrine action of IGF2 regulates adult β-cell mass and function to preserve in vivo GSIS in aging and to adapt β-cell mass in response to metabolic stress, pregnancy hormones, and acute induction of insulin resistance.
Collapse
Affiliation(s)
- Honey Modi
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Cecile Jacovetti
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - David Tarussio
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Salima Metref
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Ole D Madsen
- Hagedorn Research, Diabetes Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Fu-Ping Zhang
- Department of Physiology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pia Rantakari
- Department of Physiology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Department of Physiology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Tracy Gorman
- AstraZeneca, High-Content Biology, Discovery Sciences, Alderley Park, Macclesfield, Cheshire, U.K
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
11
|
Olson OC, Joyce JA. Cysteine cathepsin proteases: regulators of cancer progression and therapeutic response. Nat Rev Cancer 2015; 15:712-29. [PMID: 26597527 DOI: 10.1038/nrc4027] [Citation(s) in RCA: 457] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cysteine cathepsin protease activity is frequently dysregulated in the context of neoplastic transformation. Increased activity and aberrant localization of proteases within the tumour microenvironment have a potent role in driving cancer progression, proliferation, invasion and metastasis. Recent studies have also uncovered functions for cathepsins in the suppression of the response to therapeutic intervention in various malignancies. However, cathepsins can be either tumour promoting or tumour suppressive depending on the context, which emphasizes the importance of rigorous in vivo analyses to ascertain function. Here, we review the basic research and clinical findings that underlie the roles of cathepsins in cancer, and provide a roadmap for the rational integration of cathepsin-targeting agents into clinical treatment.
Collapse
Affiliation(s)
- Oakley C Olson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center
- Gerstner Sloan Kettering Graduate School of Biomedical Science, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center
- Department of Oncology, University of Lausanne
- Ludwig Institute for Cancer Research, University of Lausanne, CH-1066 Lausanne, Switzerland
| |
Collapse
|
12
|
Dasari A, Phan A, Gupta S, Rashid A, Yeung SCJ, Hess K, Chen H, Tarco E, Chen H, Wei C, Anh-Do K, Halperin D, Meric-Bernstam F, Yao J. Phase I study of the anti-IGF1R antibody cixutumumab with everolimus and octreotide in advanced well-differentiated neuroendocrine tumors. Endocr Relat Cancer 2015; 22:431-41. [PMID: 25900182 PMCID: PMC4566955 DOI: 10.1530/erc-15-0002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2015] [Indexed: 12/22/2022]
Abstract
Preclinical data suggest multiple roles for the IGF1 receptor (IGF1R) in neuroendocrine tumors (NETs), including mediating resistance to mammalian target of rapamycin (mTOR) inhibitors. Everolimus, an oral mTOR inhibitor, and octreotide long-acting repeatable (LAR) are approved for subgroups of well-differentiated NET. The primary objective of the present study was to establish the safety and recommended phase II dose (RP2D) of cixutumumab, a monoclonal antibody (MAB) against IGF1R, with everolimus and octreotide LAR. Patients with well-differentiated NET were treated with 10 mg everolimus p.o. daily, 20 mg octreotide LAR i.m. every 21 days, and escalating doses of cixutumumab. An expansion cohort was enrolled at RP2D. Correlative studies included the evaluation of mTOR pathway inhibition in paired tumor biopsies and the effects of this combination on metabolism via indirect calorimetry. Nineteen patients with progressive disease were enrolled, including nine to the expansion portion. Two patients had dose-limiting toxicities of grade 3 mucositis at 15 mg/kg cixutumumab. Long-term tolerance at RP2D was problematic, and the most common ≥grade 3 adverse event was fatigue. One patient with metastatic insulinoma had a confirmed partial response, whereas 17 had stable disease. The median progression-free survival was 43.6 weeks, and the median overall survival was 25.5 months. The RP2D of this combination per the predefined study protocol of 10 mg/kg cixutumumab i.v., 20 mg octreotide LAR i.m. every 21 days plus 10 mg everolimus p.o. daily is associated with non-dose-limiting toxicities that limit long-term tolerance. Although a signal of activity was noted in the present study, this will need to be reconciled with limited tolerance of the combination and data from larger studies of anti-IGF1R MABs in NET that have been disappointing.
Collapse
Affiliation(s)
- Arvind Dasari
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Alexandria Phan
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Sanjay Gupta
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Asif Rashid
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Sai-Ching Jim Yeung
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Kenneth Hess
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Helen Chen
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Emily Tarco
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Huiqin Chen
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Caimiao Wei
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Kim Anh-Do
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Daniel Halperin
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - Funda Meric-Bernstam
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| | - James Yao
- Department of Gastrointestinal Medical OncologyThe University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Street, Unit 426, Houston, Texas 77030, USANational Cancer InstituteRockville, Maryland, USA
| |
Collapse
|
13
|
Li J, He J, Lin G, Lu G. Inducing human parthenogenetic embryonic stem cells into islet‑like clusters. Mol Med Rep 2014; 10:2882-90. [PMID: 25241773 PMCID: PMC4227434 DOI: 10.3892/mmr.2014.2588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 07/09/2014] [Indexed: 11/05/2022] Open
Abstract
In order to determine whether human parthenogenetic embryonic stem (hpES) cells have the potential to differentiate into functional cells, a modified four-step protocol was used to induce the hpES cells into islet-like clusters (ILCs) in vitro. Growth factors activin A, retinoic acid, nicotinamide, Exendin-4 and betacellulin were added sequentially to the hpES cells at each step. The terminally differentiated cells were shown to gather into ILCs. Immunohistochemistry and semi quantitative polymerase chain reaction analyses demonstrated that the ILCs expressed islet specific hormones and functional markers. Furthermore, an insulin release test indicated that the clusters had the same physiological function as islets. The ILCs derived from hpES cells shared similar characteristics with islets. These results indicate that hpES cell-derived ILCs may be used as reliable material for the treatment of type I diabetes mellitus.
Collapse
Affiliation(s)
- Jin Li
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Jingjing He
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| | - Guangxiu Lu
- Institute of Reproductive and Stem Cell Engineering, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
14
|
Larsson C. Epigenetic aspects on therapy development for gastroenteropancreatic neuroendocrine tumors. Neuroendocrinology 2013; 97:19-25. [PMID: 22456267 DOI: 10.1159/000336087] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/10/2011] [Indexed: 01/16/2023]
Abstract
The understanding of epigenetic modifications in gastroenteropancreatic neuroendocrine tumors is a novel and still small field. Activation of the insulin-like growth factor 2 gene locus by loss of imprinting is a classical epigenetic alteration frequently observed in insulinoma. Inactivation of the MEN1 gene, commonly involved in endocrine pancreatic tumors, impairs the association with mixed lineage leukemia involved in histone H3K4me3 methylation. In addition, promising effects on tumor phenotypes such as growth, apoptosis, cell cycle arrest, and expression of neuroendocrine markers have been obtained in vitro for inhibitors of DNA methyltransferase (azacytidine) and histone deacetylation (butyrate, valproic acid, trichostatin A and MS-275). The frequent need for complementary treatments in addition to surgery in this tumor entity supports further efforts in the development and application of drugs acting at general as well as more specific epigenetic alterations.
Collapse
Affiliation(s)
- Catharina Larsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
15
|
Zhou L, Pelengaris S, Abouna S, Young J, Epstein D, Herold J, Nattkemper TW, Nakhai H, Khan M. Re-expression of IGF-II is important for beta cell regeneration in adult mice. PLoS One 2012; 7:e43623. [PMID: 22970135 PMCID: PMC3436856 DOI: 10.1371/journal.pone.0043623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 07/25/2012] [Indexed: 11/25/2022] Open
Abstract
Background The key factors which support re-expansion of beta cell numbers after injury are largely unknown. Insulin-like growth factor II (IGF-II) plays a critical role in supporting cell division and differentiation during ontogeny but its role in the adult is not known. In this study we investigated the effect of IGF-II on beta cell regeneration. Methodology/Principal Findings We employed an in vivo model of ‘switchable’ c-Myc-induced beta cell ablation, pIns-c-MycERTAM, in which 90% of beta cells are lost following 11 days of c-Myc (Myc) activation in vivo. Importantly, such ablation is normally followed by beta cell regeneration once Myc is deactivated, enabling functional studies of beta cell regeneration in vivo. IGF-II was shown to be re-expressed in the adult pancreas of pIns-c-MycERTAM/IGF-II+/+ (MIG) mice, following beta cell injury. As expected in the presence of IGF-II beta cell mass and numbers recover rapidly after ablation. In contrast, in pIns-c-MycERTAM/IGF-II+/− (MIGKO) mice, which express no IGF-II, recovery of beta cell mass and numbers were delayed and impaired. Despite failure of beta cell number increase, MIGKO mice recovered from hyperglycaemia, although this was delayed. Conclusions/Significance Our results demonstrate that beta cell regeneration in adult mice depends on re-expression of IGF-II, and supports the utility of using such ablation-recovery models for identifying other potential factors critical for underpinning successful beta cell regeneration in vivo. The potential therapeutic benefits of manipulating the IGF-II signaling systems merit further exploration.
Collapse
Affiliation(s)
- Luxian Zhou
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Stella Pelengaris
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Sylvie Abouna
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - James Young
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - David Epstein
- Department of Mathematics, University of Warwick, Coventry, United Kingdom
| | - Julia Herold
- Biodata Mining Group, Bielefeld University, Bielefeld, Germany
| | | | - Hassan Nakhai
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Michael Khan
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
- * E-mail:
| |
Collapse
|
16
|
Haley VL, Barnes DJ, Sandovici I, Constancia M, Graham CF, Pezzella F, Bühnemann C, Carter EJ, Hassan AB. Igf2 pathway dependency of the Trp53 developmental and tumour phenotypes. EMBO Mol Med 2012; 4:705-18. [PMID: 22674894 PMCID: PMC3494071 DOI: 10.1002/emmm.201101105] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 04/24/2012] [Accepted: 04/27/2012] [Indexed: 01/22/2023] Open
Abstract
Insulin-like growth factor 2 (IGF2) and the transformation related protein 53 (Trp53) are potent regulators of cell growth and metabolism in development and cancer. In vitro evidence suggests several mechanistic pathway interactions. Here, we tested whether loss of function of p53 leads to IGF2 ligand pathway dependency in vivo. Developmental lethality occurred in p53 homozygote null mice that lacked the paternal expressed allele of imprinted Igf2. Further lethality due to post-natal lung haemorrhage occurred in female progeny with Igf2 paternal null allele only if derived from double heterozygote null fathers, and was associated with a specific gene expression signature. Conditional deletion of Igf2(fl/fl) attenuated the rapid tumour onset promoted by homozygous deletion of p53(fl/fl) . Accelerated carcinoma and sarcoma tumour formation in p53(+/-) females with bi-allelic Igf2 expression was associated with reductions in p53 loss of heterozygosity and apoptosis. Igf2 genetic dependency of the p53 null phenotype during development and tumour formation suggests that targeting the IGF2 pathway may be useful in the prevention and treatment of human tumours with a disrupted Trp53 pathway.
Collapse
Affiliation(s)
- Victoria L Haley
- CR-UK Tumour Growth Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of OxfordOxford, UK
| | - David J Barnes
- CR-UK Tumour Growth Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of OxfordOxford, UK
| | - Ionel Sandovici
- University of Cambridge Metabolic Research Laboratories, Department of Obstetrics & Gynaecology, The Rosie HospitalRobinson Way, Cambridge, UK
- Centre for Trophoblast Research, University of CambridgeCambridge, UK
- National Institute of Health Research, Cambridge Biomedical Research CentreCambridge, UK
| | - Miguel Constancia
- University of Cambridge Metabolic Research Laboratories, Department of Obstetrics & Gynaecology, The Rosie HospitalRobinson Way, Cambridge, UK
- Centre for Trophoblast Research, University of CambridgeCambridge, UK
- National Institute of Health Research, Cambridge Biomedical Research CentreCambridge, UK
| | | | - Francesco Pezzella
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe HospitalOxford, UK
| | - Claudia Bühnemann
- CR-UK Tumour Growth Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of OxfordOxford, UK
| | - Emma J Carter
- CR-UK Tumour Growth Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of OxfordOxford, UK
| | - A Bassim Hassan
- CR-UK Tumour Growth Group, Oxford Molecular Pathology Institute, Sir William Dunn School of Pathology, University of OxfordOxford, UK
| |
Collapse
|
17
|
Affiliation(s)
- Frederic Clermont
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB-KULeuven, Leuven, Belgium
| | | | | |
Collapse
|
18
|
Zumsteg A, Caviezel C, Pisarsky L, Strittmatter K, García-Echeverría C, Hofmann F, Christofori G. Repression of malignant tumor progression upon pharmacologic IGF1R blockade in a mouse model of insulinoma. Mol Cancer Res 2012; 10:800-9. [PMID: 22562956 DOI: 10.1158/1541-7786.mcr-11-0522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
NVP-AEW541, a specific ATP-competitive inhibitor of the insulin-like growth factor-1 receptor (IGF1R) tyrosine kinase, has been reported to interfere with tumor growth in various tumor transplantation models. We have assessed the efficacy of NVP-AEW541 in repressing tumor growth and tumor progression in the Rip1Tag2 transgenic mouse model of pancreatic β-cell carcinogenesis. In addition, we have tested NVP-AEW541 in Rip1Tag2;RipIGF1R double-transgenic mice which show accelerated tumor growth and increased tumor malignancy compared with Rip1Tag2 single-transgenic mice. Previously, we have shown that high levels of IGF-2, a high-affinity ligand for IGF1R, are required for Rip1Tag2 tumor cell survival and tumor growth. Unexpectedly, treatment of Rip1Tag2 mice with NVP-AEW541 in prevention and intervention trials neither did affect tumor growth nor tumor cell proliferation and apoptosis. Yet, it significantly repressed progression to tumor malignancy, that is, the rate of the transition from differentiated adenoma to invasive carcinoma. Treatment of Rip1Tag2;RipIGF1R double-transgenic mice resulted in moderately reduced tumor volumes and increased rates of tumor cell apoptosis. Sustained expression of IGF-2 and of the IGF-2-binding form of insulin receptor (IR-A) in tumor cells suggests a compensatory role of IR-A upon IGF1R blockade. The results indicate that inhibition of IGF1R alone is not sufficient to efficiently block insulinoma growth and imply an overlapping role of IGF1R and insulin receptor in executing mitogenic and survival stimuli elicited by IGF-2. The reduction of tumor invasion upon IGF1R blockade on the other hand indicates a critical function of IGF1R signaling for the acquisition of a malignant phenotype.
Collapse
Affiliation(s)
- Adrian Zumsteg
- Institute of Biochemistry and Genetics, Department of Biomedicine, University of Basel, Mattenstrasse 28, Basel 4058, Switzerland
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Insulin-like growth factor-II (IGF-II) affects many aspects of cellular function through its ability to activate several different receptors and, consequently, numerous intracellular signalling molecules. Thus, IGF-II is a key regulator of normal foetal development and growth. However, abnormalities in IGF-II function are associated with cardiovascular disease and cancer. Here, we review the cellular mechanisms by which IGF-II's physiological and pathophysiological actions are exerted by discussing the involvement of the type 1 and type 2 IGF receptors (IGF1R and IGF2R), the insulin receptor and the downstream MAP kinase, PI-3 kinase and G-protein-coupled signalling pathways in mediating IGF-II stimulated cellular proliferation, survival, differentiation and migration.
Collapse
Affiliation(s)
- Lynda K Harris
- Maternal and Fetal Health Research Centre, University of Manchester, UK
| | | |
Collapse
|
20
|
Hu Q, Gao F, Tian W, Ruteshouser EC, Wang Y, Lazar A, Stewart J, Strong LC, Behringer RR, Huff V. Wt1 ablation and Igf2 upregulation in mice result in Wilms tumors with elevated ERK1/2 phosphorylation. J Clin Invest 2011; 121:174-83. [PMID: 21123950 PMCID: PMC3007149 DOI: 10.1172/jci43772] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 10/06/2010] [Indexed: 01/13/2023] Open
Abstract
Wilms tumor (WT) is a genetically heterogeneous childhood kidney tumor. Several genetic alterations have been identified in WT patients, including inactivating mutations in WT1 and loss of heterozygosity or loss of imprinting at 11p15, which results in biallelic expression of IGF2. However, the mechanisms by which one or a combination of genetic alterations results in tumorigenesis has remained challenging to determine, given the lack of a mouse model of WT. Here, we engineered mice to sustain mosaic, somatic ablation of Wt1 and constitutional Igf2 upregulation, mimicking a subset of human tumors. Mice with this combination of genetic alterations developed tumors at an early age. Mechanistically, Wt1 ablation blocked mesenchyme differentiation, and increased Igf2 expression upregulated ERK1/2 phosphorylation. Importantly, a subset of human tumors similarly displayed upregulation of ERK1/2 phosphorylation, which suggests ERK signaling might contribute to WT development. Thus, we have generated a biologically relevant mouse model of WT and defined one combination of driver alterations for WT. This mouse model will provide a powerful tool to study the biology of WT initiation and progression and to investigate therapeutic strategies for cancers with IGF pathway dysregulation.
Collapse
Affiliation(s)
- Qianghua Hu
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Fei Gao
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Weihua Tian
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - E. Cristy Ruteshouser
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Yaqing Wang
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Alexander Lazar
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - John Stewart
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Louise C. Strong
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Richard R. Behringer
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Vicki Huff
- Department of Genetics,
Department of Experimental Therapeutics, and
Department of Pathology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA.
Graduate Program in Genes and Development and
Graduate Program in Human Molecular Genetics, UT-Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
21
|
Clayton PE, Banerjee I, Murray PG, Renehan AG. Growth hormone, the insulin-like growth factor axis, insulin and cancer risk. Nat Rev Endocrinol 2011; 7:11-24. [PMID: 20956999 DOI: 10.1038/nrendo.2010.171] [Citation(s) in RCA: 249] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Growth hormone (GH), insulin-like growth factor (IGF)-I and insulin have potent growth-promoting and anabolic actions. Their potential involvement in tumor promotion and progression has been of concern for several decades. The evidence that GH, IGF-I and insulin can promote and contribute to cancer progression comes from various sources, including transgenic and knockout mouse models and animal and human cell lines derived from cancers. Assessments of the GH-IGF axis in healthy individuals followed up to assess cancer incidence provide direct evidence of this risk; raised IGF-I levels in blood are associated with a slightly increased risk of some cancers. Studies of human diseases characterized by excess growth factor secretion or treated with growth factors have produced reassuring data, with no notable increases in de novo cancers in children treated with GH. Although follow-up for the vast majority of these children does not yet extend beyond young adulthood, a slight increase in cancers in those with long-standing excess GH secretion (as seen in patients with acromegaly) and no overall increase in cancer with insulin treatment, have been observed. Nevertheless, long-term surveillance for cancer incidence in all populations exposed to increased levels of GH is vitally important.
Collapse
Affiliation(s)
- Peter E Clayton
- Manchester Academic Health Sciences Centre, University of Manchester, Paediatric Endocrinology, Royal Manchester Children's Hospital, Oxford Road, Manchester, UK.
| | | | | | | |
Collapse
|
22
|
Maki RG. Small is beautiful: insulin-like growth factors and their role in growth, development, and cancer. J Clin Oncol 2010; 28:4985-95. [PMID: 20975071 PMCID: PMC3039924 DOI: 10.1200/jco.2009.27.5040] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 08/23/2010] [Indexed: 12/17/2022] Open
Abstract
Insulin-like growth factors were discovered more than 50 years ago as mediators of growth hormone that effect growth and differentiation of bone and skeletal muscle. Interest of the role of insulin-like growth factors in cancer reached a peak in the 1990s, and then waned until the availability in the past 5 years of monoclonal antibodies and small molecules that block the insulin-like growth factor 1 receptor. In this article, we review the history of insulin-like growth factors and their role in growth, development, organism survival, and in cancer, both epithelial cancers and sarcomas. Recent developments regarding phase I to II clinical trials of such agents are discussed, as well as potential studies to consider in the future, given the lack of efficacy of one such monoclonal antibody in combination with cytotoxic chemotherapy in a first-line study in metastatic non-small-cell lung adenocarcinoma. Greater success with these agents clinically is expected when combining the agents with inhibitors of other cell signaling pathways in which cross-resistance has been observed.
Collapse
Affiliation(s)
- Robert G Maki
- Memorial Sloan-Kettering Cancer Center, New York, NY 10065-6007, USA.
| |
Collapse
|
23
|
Abstract
Under physiological conditions, cells receive fate-determining signals from their tissue surroundings, primarily in the form of polypeptide growth factors. Integration of these extracellular signals underlies tissue homeostasis. Although departure from homeostasis and tumor initiation are instigated by oncogenic mutations rather than by growth factors, the latter are the major regulators of all subsequent steps of tumor progression, namely clonal expansion, invasion across tissue barriers, angiogenesis, and colonization of distant niches. Here, we discuss the relevant growth factor families, their roles in tumor biology, as well as the respective downstream signaling pathways. Importantly, cancer-associated activating mutations that impinge on these pathways often relieve, in part, the reliance of tumors on growth factors. On the other hand, growth factors are frequently involved in evolvement of resistance to therapeutic regimens, which extends the roles for polypeptide factors to very late phases of tumor progression and offers opportunities for cancer therapy.
Collapse
|
24
|
Anastassiadis K, Rostovskaya M, Lubitz S, Weidlich S, Stewart AF. Precise conditional immortalization of mouse cells using tetracycline-regulated SV40 large T-antigen. Genesis 2010; 48:220-32. [PMID: 20146354 DOI: 10.1002/dvg.20605] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cellular immortalization provides a way for expansion and subsequent molecular characterization of rare cell types. Ideally, immortalization can be achieved by the reversible expression of immortalizing proteins. Here, we describe the use of conditional immortalization based on a modified tetracycline-regulated system for the expression of SV40 large T-antigen in embryonic stem (ES) cells and mice. The modified system relies on a codon improved reverse tetracycline transactivator (irtTA) fused to the ligand-binding domain (LBD) of the androgen receptor (irtTA-ABD) or of a mutated glucocorticoid receptor (irtTA-GBD*). Induction of T-antigen is conferred only after addition of two ligands, one to activate the LBD (mibolerone for irtTA-ABD or dexamethasone for irtTA-GBD*) and one to activate the tetracycline transactivator (doxycycline). In ES cells, changes in gene expression upon large T induction were limited and reversible upon deinduction. Similarly, expression of T-antigen was very tightly regulated in mice. We have isolated and expanded bone marrow mesenchymal stem cells that could be genetically manipulated and maintained their differentiation properties after several passages of expansion under conditions that induce the expression of large T-antigen.
Collapse
Affiliation(s)
- Konstantinos Anastassiadis
- Center for Regenerative Therapies Dresden, BioInnovationsZentrum, Technische Universität Dresden, Dresden, Germany.
| | | | | | | | | |
Collapse
|
25
|
Rosenzweig SA, Atreya HS. Defining the pathway to insulin-like growth factor system targeting in cancer. Biochem Pharmacol 2010; 80:1115-24. [PMID: 20599789 DOI: 10.1016/j.bcp.2010.06.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2010] [Revised: 06/12/2010] [Accepted: 06/15/2010] [Indexed: 12/28/2022]
Abstract
The insulin-like growth factors (IGFs; IGF-1 and IGF-2) play central roles in cell growth, differentiation, survival, transformation and metastasis. The biologic effects of the IGFs are mediated by the IGF-1 receptor (IGF-1R), a receptor tyrosine kinase with homology to the insulin receptor (IR). Dysregulation of the IGF system is well recognized as a key contributor to the progression of multiple cancers, with IGF-1R activation increasing the tumorigenic potential of breast, prostate, lung, colon and head and neck squamous cell carcinoma (HNSCC). Despite this relationship, targeting the IGF-1R has only recently undergone development as a molecular cancer therapeutic. As it has taken hold, we are witnessing a robust increase and interest in targeting the inhibition of IGF-1R signaling. This is accentuated by the list of over 30 drugs, including monoclonal antibodies (mAbs) and tyrosine kinase inhibitors (TKIs) that are under evaluation as single agents or in combination therapies. The IGF-binding proteins (IGFBPs) represent the third component of the IGF system consisting of a class of six soluble secretory proteins. They represent a unique class of naturally occurring IGF-antagonists that bind to and sequester IGF-1 and IGF-2, inhibiting their access to the IGF-1R. Due to their dual targeting of the IGFs without affecting insulin action, the IGFBPs are an untapped "third" class of IGF-1R inhibitors. In this commentary, we highlight some of the significant aspects of and prospects for targeting the IGF-1R and describe what the future may hold.
Collapse
Affiliation(s)
- Steven A Rosenzweig
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, PO Box 250505, Charleston, SC 29425-5050, USA.
| | | |
Collapse
|
26
|
Cornu M, Modi H, Kawamori D, Kulkarni RN, Joffraud M, Thorens B. Glucagon-like peptide-1 increases beta-cell glucose competence and proliferation by translational induction of insulin-like growth factor-1 receptor expression. J Biol Chem 2010; 285:10538-45. [PMID: 20145256 DOI: 10.1074/jbc.m109.091116] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) protects beta-cells against apoptosis, increases their glucose competence, and induces their proliferation. We previously demonstrated that the anti-apoptotic effect was mediated by an increase in insulin-like growth factor-1 receptor (IGF-1R) expression and signaling, which was dependent on autocrine secretion of insulin-like growth factor 2 (IGF-2). Here, we further investigated how GLP-1 induces IGF-1R expression and whether the IGF-2/IGF-1R autocrine loop is also involved in mediating GLP-1-increase in glucose competence and proliferation. We show that GLP-1 up-regulated IGF-1R expression by a protein kinase A-dependent translational control mechanism, whereas isobutylmethylxanthine, which led to higher intracellular accumulation of cAMP than GLP-1, increased both IGF-1R transcription and translation. We then demonstrated, using MIN6 cells and primary islets, that the glucose competence of these cells was dependent on the level of IGF-1R expression and on IGF-2 secretion. We showed that GLP-1-induced primary beta-cell proliferation was suppressed by Igf-1r gene inactivation and by IGF-2 immunoneutralization or knockdown. Together our data show that regulation of beta-cell number and function by GLP-1 depends on the cAMP/protein kinase A mediated-induction of IGF-1R expression and the increased activity of an IGF-2/IGF-1R autocrine loop.
Collapse
Affiliation(s)
- Marion Cornu
- Department of Physiology and Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
27
|
Zhang J, Zhang F, Didelot X, Bruce KD, Cagampang FR, Vatish M, Hanson M, Lehnert H, Ceriello A, Byrne CD. Maternal high fat diet during pregnancy and lactation alters hepatic expression of insulin like growth factor-2 and key microRNAs in the adult offspring. BMC Genomics 2009; 10:478. [PMID: 19835573 PMCID: PMC2770530 DOI: 10.1186/1471-2164-10-478] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2009] [Accepted: 10/16/2009] [Indexed: 12/14/2022] Open
Abstract
Background miRNAs play important roles in the regulation of gene functions. Maternal dietary modifications during pregnancy and gestation have long-term effects on the offspring, but it is not known whether a maternal high fat (HF) diet during pregnancy and lactation alters expression of key miRNAs in the offspring. Results We studied the effects of maternal HF diet on the adult offspring by feeding mice with either a HF or a chow diet prior to conception, during pregnancy and lactation, and all offspring were weaned onto the same chow diet until adulthood. Maternal HF fed offspring had markedly increased hepatic mRNA levels of peroxisome proliferator activated receptor-alpha (ppar-alpha) and carnitine palmitoyl transferase-1a (cpt-1a) as well as insulin like growth factor-2 (Igf2). A HF diet induced up-regulation of ppar-alpha and cpt-1a expression in the wild type but not in Igf2 knock out mice. Furthermore, hepatic expression of let-7c was also reduced in maternal HF fed offspring. Among 579 miRNAs measured with microarray, ~23 miRNA levels were reduced by ~1.5-4.9-fold. Reduced expression of miR-709 (a highly expressed miRNA), miR-122, miR-192, miR-194, miR-26a, let-7a, let7b and let-7c, miR-494 and miR-483* (reduced by ~4.9 fold) was validated by qPCR. We found that methyl-CpG binding protein 2 was the common predicted target for miR-709, miR-let7s, miR-122, miR-194 and miR-26a using our own purpose-built computer program. Conclusion Maternal HF feeding during pregnancy and lactation induced co-ordinated and long-lasting changes in expression of Igf2, fat metabolic genes and several important miRNAs in the offspring.
Collapse
Affiliation(s)
- Junlong Zhang
- Clinical Science Research Institute, Warwick Medical School, Clinical Sciences Building, University Hospital, Walsgrave Campus, Coventry, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gu M, Pritlove DC, Boyd CAR, Vatish M. Placental expression of 2,3 bisphosphoglycerate mutase in IGF-II knock out mouse: correlation of circulating maternal 2,3 bisphosphoglycerate and fetal growth. Placenta 2009; 30:919-22. [PMID: 19733906 DOI: 10.1016/j.placenta.2009.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 08/14/2009] [Accepted: 08/18/2009] [Indexed: 11/16/2022]
Abstract
Bisphosphoglycerate mutase (BPGM) catalyses the formation of 2,3 bisphosphoglycerate (BPG) a ligand of haemoglobin. BPG facilitates liberation of oxygen from haemoglobin at low oxygen tension enabling efficient delivery of oxygen to tissues. We describe expression of BPGM in mouse labyrinthine trophoblasts, located at the maternal-placental interface. Expression is lower in placentae of igf2(+/-) knockout mice, a widely used model of growth restriction, compared to wild type placentae. Circulating maternal BPG increased throughout gestation but this increase was less in wt mothers carrying igf2(+/-) pups than in those carrying exclusively wt pups. This reduction was observed well before term and may contribute to the low birth weight of igf2(+/-) pups. Strikingly, we also measured reductions of fetal and placental weight in wt littermates of igf2(+/-) pups compared to pups developing in an exclusively wt environment. These data suggest that placental expression of BPGM can influence maternal BPG concentrations and supports a hypothesis under which BPG synthesized in the placenta may act on maternal haemoglobin to enhance delivery of oxygen to the developing fetus.
Collapse
Affiliation(s)
- M Gu
- Clinical Sciences Research Institute, University of Warwick Medical School, Clifford Bridge Road, Coventry, UK
| | | | | | | |
Collapse
|
29
|
Sachdev D. Regulation of breast cancer metastasis by IGF signaling. J Mammary Gland Biol Neoplasia 2008; 13:431-41. [PMID: 19030970 DOI: 10.1007/s10911-008-9105-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 11/06/2008] [Indexed: 02/06/2023] Open
Abstract
The insulin-like growth factors (IGFs) signaling via the type I insulin-like growth factor receptor (IGF-1R) regulate multiple aspects of malignancy. The importance of IGF-1R in regulating the malignant phenotype is currently being validated in numerous clinical trials for cancer including breast cancer. This review discusses the regulation of breast cancer metastasis by IGF-1R. IGF-1R stimulates invasion and survival in anchorage independent conditions. The regulation of metastasis independently of tumor growth by IGF-1R is also discussed. Finally, the impact of this on clinical trial design and outcomes, and the need for biomarkers, other than reduction in tumor size, are discussed in light of the fact that inhibition of metastasis is not measured in conventional clinical trial design.
Collapse
Affiliation(s)
- Deepali Sachdev
- Department of Medicine and Masonic Cancer Center, University of Minnesota, MMC 806, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
30
|
Brown KW, Power F, Moore B, Charles AK, Malik KTA. Frequency and timing of loss of imprinting at 11p13 and 11p15 in Wilms' tumor development. Mol Cancer Res 2008; 6:1114-23. [PMID: 18644976 DOI: 10.1158/1541-7786.mcr-08-0002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Epigenetic changes occur frequently in Wilms' tumor (WT), especially loss of imprinting (LOI) of IGF2/H19 at 11p15. Our previous results have identified imprinted transcripts (WT1-AS and AWT1) from the WT1 locus at 11p13 and showed LOI of these in some WTs. In this article, we set out to test the relationship between LOI at 11p13 and 11p15 and their timing in WT progression relative to other genetic changes. We found a higher level (83%) of 11p13 LOI in WT than of 11p15 LOI (71%). There was no correlation between methylation levels at the 11p13 and 11p15 differentially methylated regions or between allelic expression of WT1-AS/AWT1 and IGF2. Interestingly, retention of normal imprinting at 11p13 was associated with a small group of relatively late-onset, high-stage WTs. An examination of genetic and epigenetic alterations in nephrogenic rests, which are premalignant WT precursors, showed that LOI at both 11p13 and 11p15 occurred before either 16q loss of heterozygosity (LOH) or 7p LOH. This suggests that these LOH events are very unlikely to be a cause of LOI but that LOH may act by potentiating the effects of overexpression of IGF2 and/or WT1-AS/AWT1 that result from LOI.
Collapse
Affiliation(s)
- Keith W Brown
- Department of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom.
| | | | | | | | | |
Collapse
|
31
|
Prince SN, Foulstone EJ, Zaccheo OJ, Williams C, Hassan AB. Functional evaluation of novel soluble insulin-like growth factor (IGF)-II–specific ligand traps based on modified domain 11 of the human IGF2 receptor. Mol Cancer Ther 2007; 6:607-17. [PMID: 17308058 DOI: 10.1158/1535-7163.mct-06-0509] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ligands transported by the mannose 6-phosphate/insulin-like growth factor (IGF)-II receptor (IGF2R) include IGF-II- and mannose 6-phosphate-modified proteins. Increased extracellular supply of IGF-II, either secondary to loss of the clearance function of IGF2R, loss of IGF binding protein function, or increased IGF2 gene expression, can lead to embryonic overgrowth and cancer promotion. Reduced supply of IGF-II is detrimental to tumor growth, and this suggests that gain of function of IGF-II is a molecular target for human cancer therapy. Domain 11 of IGF2R binds IGF-II with high specificity and affinity. Mutagenesis studies have shown that substitution of glutamic acid for lysine at residue 1554 results in a 6-fold higher affinity for IGF-II (20.5 nmol/L) than native domain 11 (119 nmol/L). Here, we generate a novel high-affinity IGF-II ligand trap by fusion of mutated human 11(E1554K) to a COOH-terminal human IgG1 Fc domain (11(E1554K)-Fc). The resulting homodimer has a significantly increased affinity for IGF-II (1.79 nmol/L) when measured by surface plasmon resonance. IGF-II signaling via the IGF-I receptor and the proliferative effect of IGF-II were specifically inhibited by 11(E1554K)-Fc in both HaCaT and Igf2(-/-) mouse embryonic fibroblast cells. These data confirm that a novel engineered and soluble IGF2R-11(E1554K)-Fc protein functions as an IGF-II-specific and high-affinity ligand trap in vitro and that this protein has potential application as an IGF-II antagonist for cancer therapy following in vivo experimental evaluation.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Proliferation
- Drosophila melanogaster/growth & development
- Drosophila melanogaster/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Embryo, Nonmammalian
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Genetic Vectors
- Humans
- Insulin-Like Growth Factor II/genetics
- Insulin-Like Growth Factor II/physiology
- Keratinocytes/metabolism
- Ligands
- Mice
- Mice, Knockout
- Pichia/chemistry
- Pichia/metabolism
- Protein Binding
- Receptor, IGF Type 1/genetics
- Receptor, IGF Type 1/metabolism
- Receptor, IGF Type 2/genetics
- Receptor, IGF Type 2/metabolism
- Receptors, Fc/genetics
- Receptors, Fc/metabolism
- Surface Plasmon Resonance
- Thymidine/metabolism
Collapse
Affiliation(s)
- Stuart N Prince
- Weatherall Institute for Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom
| | | | | | | | | |
Collapse
|
32
|
Samani AA, Yakar S, LeRoith D, Brodt P. The role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocr Rev 2007; 28:20-47. [PMID: 16931767 DOI: 10.1210/er.2006-0001] [Citation(s) in RCA: 730] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IGF-I receptor (IGF-IR) signaling and functions are mediated through the activities of a complex molecular network of positive (e.g., type I IGF) and negative (e.g., the type II IGF receptor, IGF-IIR) effectors. Under normal physiological conditions, the balance between the expression and activities of these molecules is tightly controlled. Changes in this delicate balance (e.g., overexpression of one effector) may trigger a cascade of molecular events that can ultimately lead to malignancy. In recent years, evidence has been mounting that the IGF axis may be involved in human cancer progression and can be targeted for therapeutic intervention. Here we review old and more recent evidence on the role the IGF system in malignancy and highlight experimental and clinical studies that provide novel insights into the complex mechanisms that contribute to its oncogenic potential. Controversies arising from conflicting evidence on the relevance of IGF-IR and its ligands to human cancer are discussed. Our review highlights the importance of viewing the IGF axis as a complex multifactorial system and shows that changes in the expression levels of any one component of the axis, in a given malignancy, should be interpreted with caution and viewed in a wider context that takes into account the expression levels, state of activation, accessibility, and functionality of other interacting components. Because IGF targeting for anticancer therapy is rapidly becoming a clinical reality, an understanding of this complexity is timely because it is likely to have an impact on the design, mode of action, and clinical outcomes of newly developed drugs.
Collapse
Affiliation(s)
- Amir Abbas Samani
- Department of Medicine, McGill University Health Center, Royal Victoria Hospital, Room H6.25687, Pine Avenue West, Montreal, Québec, Canada H3A 1A1
| | | | | | | |
Collapse
|
33
|
Chen HL, Li T, Qiu XW, Wu J, Ling JQ, Sun ZH, Wang W, Chen W, Hou A, Vu TH, Hoffman AR, Hu JF. Correction of aberrant imprinting of IGF2 in human tumors by nuclear transfer-induced epigenetic reprogramming. EMBO J 2006; 25:5329-38. [PMID: 17082775 PMCID: PMC1636609 DOI: 10.1038/sj.emboj.7601399] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Accepted: 09/27/2006] [Indexed: 01/20/2023] Open
Abstract
Loss of genomic imprinting of insulin-like growth factor II (IGF2) is a hallmark of many human neoplasms. We attempted to correct this aberrant epigenotype by transferring nuclei from human tumor cells that showed loss of IGF2 imprinting into enucleated mouse and human fibroblasts that had maintained normal IGF2 imprinting. After nuclear transfer, the abnormal biallelic expression of IGF2 in tumor nuclei transiently converted to normal monoallelic imprinted expression in the reconstructed diploid cells. In tetraploid hybrid cells, however, normal IGF2 imprinting was permanently restored in the tumor genome. Inhibition of the synthesis of putative trans imprinting factors with cycloheximide led to loss of IGF2 imprinting in normal cultured fibroblasts, suggesting that normal cells produce proteins that act in trans to induce or maintain genomic imprinting. These data demonstrate that an abnormal tumor epigenotype can be corrected by in vitro reprogramming, and suggest that loss of imprinting is associated with the loss of activity of non-CTCF trans imprinting factor(s) that are either inactivated or mutated in tumors.
Collapse
Affiliation(s)
- Hui Ling Chen
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
- These authors contributed equally to this work
| | - Tao Li
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University Medical School, Palo Alto, CA, USA
- These authors contributed equally to this work
| | - Xin Wen Qiu
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University Medical School, Palo Alto, CA, USA
| | - Jie Wu
- GMR Epigenetics Corporation, Palo Alto, CA, USA
| | - Jian Qun Ling
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University Medical School, Palo Alto, CA, USA
| | | | - Weibo Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Wei Chen
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Aiju Hou
- Department of Medicine, Stanford University Medical School, Palo Alto, CA, USA
| | - Thanh H Vu
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University Medical School, Palo Alto, CA, USA
| | - Andrew R Hoffman
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University Medical School, Palo Alto, CA, USA
- These authors are senior authors of this report
- Department of Medicine, Stanford University Medical School, Palo Alto, CA 94304, USA. Tel.: +1 650 858 3930; Fax: +1 650 856 8024; E-mail:
| | - Ji-Fan Hu
- Medical Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
- GMR Epigenetics Corporation, Palo Alto, CA, USA
- These authors are senior authors of this report
- Department of Medicine, PAIRE, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA. Tel.: +1 650 493 5000 x 63175; Fax: +1 650 856 8024; E-mail:
| |
Collapse
|
34
|
Harper J, Burns JL, Foulstone EJ, Pignatelli M, Zaina S, Hassan AB. Soluble IGF2 receptor rescues Apc(Min/+) intestinal adenoma progression induced by Igf2 loss of imprinting. Cancer Res 2006; 66:1940-8. [PMID: 16488992 DOI: 10.1158/0008-5472.can-05-2036] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The potent growth-promoting activity of insulin-like growth factor-II (IGF-II) is highly regulated during development but frequently up-regulated in tumors. Increased expression of the normally monoallelic (paternally expressed) mouse (Igf2) and human (IGF2) genes modify progression of intestinal adenoma in the Apc(Min/+) mouse and correlate with a high relative risk of human colorectal cancer susceptibility, respectively. We examined the functional consequence of Igf2 allelic dosage (null, monoallelic, and biallelic) on intestinal adenoma development in the Apc(Min/+) by breeding with mice with either disruption of Igf2 paternal allele or H19 maternal allele and used these models to evaluate an IGF-II-specific therapeutic intervention. Increased allelic Igf2 expression led to elongation of intestinal crypts, increased adenoma growth independent of systemic growth, and increased adenoma nuclear beta-catenin staining. By introducing a transgene expressing a soluble form of the full-length IGF-II/mannose 6-phosphate receptor (sIGF2R) in the intestine, which acts as a specific inhibitor of IGF-II ligand bioavailability (ligand trap), we show rescue of the Igf2-dependent intestinal and adenoma phenotype. This evidence shows the functional potency of allelic dosage of an epigenetically regulated gene in cancer and supports the application of an IGF-II ligand-specific therapeutic intervention in colorectal cancer.
Collapse
Affiliation(s)
- James Harper
- Department of Cellular and Molecular Medicine, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Epigenetic alterations in cancer occur at least as commonly as genetic mutations, but epigenetic alterations could occur secondarily to the tumor process itself. To establish a causal role of epigenetic changes, investigators have turned to genetically engineered mouse models. Here, we review a recent study showing that a mouse model of loss of imprinting (LOI) of the insulin-like growth factor II gene (Igf2), which shows aberrant activation of the normally silent maternal allele, modifies the risk of intestinal neoplasia caused by mutations of the adenomatous polyposis coli (Apc) gene. This increased risk corresponds to the apparent increased risk of colorectal cancer in patients with LOI of IGF2. The model suggests that preexisting epigenetic alterations in normal cells increase tumor risk by expanding the target cell population and/or modulating the effect of subsequent genetic alterations on these cells, providing a novel idea for cancer risk management.
Collapse
Affiliation(s)
- Atsushi Kaneda
- Division of Molecular Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
36
|
Abstract
Epigenetic dysregulation is central to cancer development and progression. This dysregulation includes hypomethylation leading to oncogene activation and chromosomal instability, hypermethylation and tumor suppressor gene silencing, and chromatin modification acting directly, and cooperatively with methylation changes, to modify gene expression. In addition, disrupted genomic imprinting appears to contribute to colorectal cancer risk, and serves as a gatekeeper in Wilms tumor. A cancer predisposing disorder, Beckwith-Wiedemann syndrome, usually arises from epigenetic errors, solidifying the causal role of epigenetics in cancer. While cancer epigenetics has been reviewed extensively elsewhere, the main focus of this review will be to present the view that epigenetics and genetics are complementary in the area of cancer etiology, the focus of this volume. I propose a hypothesis in which epigenetic alterations contribute to tumor progression, but they also increase the probability that genetic changes, when they occur, will lead to cancer initiation. This hypothesis could contribute to a new understanding of the role of environmental carcinogens that may not be fully explained through a purely genetic view or by tests, such as bacterial mutation frequency, that ignore epigenetic factors.
Collapse
Affiliation(s)
- Andrew P Feinberg
- Epigenetics Unit, Departments of Medicine, Oncology, and Molecular Biology and Genetics, Johns Hopkins University School of Medicine, 1064 Ross, Johns Hopkins Medical School, 720 Rutland Ave., Baltimore, MD 21205, USA.
| |
Collapse
|
37
|
Foulstone E, Prince S, Zaccheo O, Burns JL, Harper J, Jacobs C, Church D, Hassan AB. Insulin-like growth factor ligands, receptors, and binding proteins in cancer. J Pathol 2005; 205:145-53. [PMID: 15641016 DOI: 10.1002/path.1712] [Citation(s) in RCA: 189] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
This review aims to summarize experimental evidence supporting the role of the insulin-like growth factor (IGF) signalling system in the progression, maintenance, and treatment of cancer. These data implicate the IGF system as an important modifier of cancer cell proliferation, survival, growth, and treatment sensitivity. The role of the IGF system in cancer should be examined in the context of the extra-cellular and intra-cellular signalling networks, in particular: phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt/PKB), mammalian target of rapamycin (mTOR), and forkhead transcription factors (FOXO). This review highlights evidence derived from molecular structure and functional genetics with respect to how the extra-cellular components of the IGF system function normally, and their subsequent modifications in cancer. The therapeutic relevance of the research evidence described is also addressed, as the challenge is to apply this knowledge to human health.
Collapse
Affiliation(s)
- E Foulstone
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol BS8 1TD, UK
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Sakatani T, Kaneda A, Iacobuzio-Donahue CA, Carter MG, de Boom Witzel S, Okano H, Ko MSH, Ohlsson R, Longo DL, Feinberg AP. Loss of imprinting of Igf2 alters intestinal maturation and tumorigenesis in mice. Science 2005; 307:1976-8. [PMID: 15731405 DOI: 10.1126/science.1108080] [Citation(s) in RCA: 228] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Loss of imprinting (LOI) of the insulin-like growth factor II gene (IGF2) is an epigenetic alteration that results in a modest increase in IGF2 expression, and it is present in the normal colonic mucosa of about 30% of patients with colorectal cancer. To investigate its role in intestinal tumorigenesis, we created a mouse model of Igf2 LOI by crossing female H19+/- mice with male Apc+/Min mice. Mice with LOI developed twice as many intestinal tumors as did control littermates. Notably, these mice also showed a shift toward a less differentiated normal intestinal epithelium, reflected by an increase in crypt length and increased staining with progenitor cell markers. A similar shift in differentiation was seen in the normal colonic mucosa of humans with LOI. Thus, altered maturation of nonneoplastic tissue may be one mechanism by which epigenetic changes affect cancer risk.
Collapse
Affiliation(s)
- Takashi Sakatani
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Affiliation(s)
- Andrew P Feinberg
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA.
| | | |
Collapse
|
40
|
Abstract
NICTH is a fairly uncommon disorder but has been well characterized and the molecular mechanisms involved have given insights into the IGF system, both normal and abnormal. NICTH has brought together the molecular biologists and endocrinologists in a classic cross cultural coordination to study the disorder from all angles and to further understand the disorder.
Collapse
|
41
|
Radovanovic I, D'Angelo MG, Aguzzi A. Angiogenesis in transgenic models of multistep angiogenesis. Cancer Treat Res 2004; 117:97-114. [PMID: 15015554 DOI: 10.1007/978-1-4419-8871-3_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The histopathology and the epidemiology of human cancers, as well as studies of animal models of tumorigenesis, have led to a widely accepted notion that multiple genetic and epigenetic changes have to accumulate for progression to malignancy. Formation of new blood vessels (tumor angiogenesis) has been recognized, in addition to proliferative capabilities and ability to down-modulate cell death (apoptosis), as essential for the progressive growth and expansion of solid tumors. Mice overexpressing activated forms of oncogenes or carrying targeted mutations in tumor suppressor genes have proven extremely useful for linking the function of these genes with specific tumor features such as continuous proliferation, escape from apoptosis, invasion and neo-angiogenesis. The interbreeding of these mice allows for studying the extent of cooperativity between different genetic lesions in disease progression, leading to a greater understanding of multi-stage nature of tumorigenesis.
Collapse
Affiliation(s)
- Ivan Radovanovic
- Institute of Neuropathology, University Hospital of Zürich, Zürich, Switzerland
| | | | | |
Collapse
|
42
|
Homo-Delarche F. Neuroendocrine Immuno-ontogeny of the Pathogenesis of Autoimmune Diabetes in the Nonobese Diabetic (NOD) Mouse. ILAR J 2004; 45:237-58. [PMID: 15229372 DOI: 10.1093/ilar.45.3.237] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease in which insulin-producing beta cells of the pancreatic islets of Langerhans are destroyed. The nonobese diabetic (NOD) mouse is one of the rare spontaneous models that enable the study of prediabetic pancreatic events. The etiology of the autoimmune attack in human and animal T1D is still unknown, but genetic and environmental factors are involved in both cases. Although several autoantigens have been identified and defective immune-system regulation is implicated, this information does not satisfactorily explain the generally accepted beta-cell specificity of the disease or how so many and diverse environmental factors intervene in its pathogenesis. Based on data obtained from evaluating glucose homeostasis in a variety of situations, particularly stress and cytokine administration, in young prediabetic NOD mice, the author hypothesizes that the islet of Langerhans is a major actor, and its altered regulation through environmentally induced insulin resistance might reveal latent T1D. It is also postulated that T1D pathogenesis might be linked to abnormal pancreas development, probably due to disturbances of glutamic acid decarboxylase (GAD)+ innervation phagocytosis by defective macrophages during the early postnatal period. Also discussed is the role of defective presentation of pancreatic hormones and GAD in the thymus, and its potential repercussion on T-cell tolerance. Observations have demonstrated that the diabetogenic process in the NOD mouse is extremely complex, involving neuroendocrine immune interaction from fetal life onward.
Collapse
Affiliation(s)
- Françoise Homo-Delarche
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Université Paris 7/D.Diderot, Paris, France
| |
Collapse
|
43
|
Hernandez L, Kozlov S, Piras G, Stewart CL. Paternal and maternal genomes confer opposite effects on proliferation, cell-cycle length, senescence, and tumor formation. Proc Natl Acad Sci U S A 2003; 100:13344-9. [PMID: 14581617 PMCID: PMC263813 DOI: 10.1073/pnas.2234026100] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Loss of imprinting is the silencing of active imprinted genes or the activation of silent imprinted genes, and it is one of the most common epigenetic changes associated with the development of a wide variety of tumors. Here, we have analyzed the effects that global imprinted gene expression has on cell proliferation and transformation. Primary mouse embryonic fibroblasts (MEFs), whose entire genome is either exclusively paternal (androgenetic) or maternal (parthenogenetic), exhibit dramatically contrasting patterns of growth. In comparison with biparental MEFs, andro-genetic proliferation is characterized by a shorter cell cycle, increased saturation density, spontaneous transformation, and formation of tumors at low passage number. Parthenogenetic MEFs reach a lower saturation density, senesce, and die. The maternally expressed imprinted genes p57kip2 and M6P/Igf2r retard proliferation and reduce the long-term growth of MEFs. In contrast, the paternally expressed growth factor Igf2 is essential for the long-term proliferation of all genotypes. Increased Igf2 expression in primary MEFs not only stimulates proliferation, but also results in their rapid conversion to malignancy with tumor formation of short latency. Our results reveal that paternally expressed imprinted genes, in the absence of maternal imprinted genes, predispose fibroblasts to rapid transformation. A potent factor in their transformation is IGF2, which on increased expression results in the rapid conversion of primary cells to malignancy. These results reveal a route by which malignant choriocarcinoma may arise from molar pregnancies. They also suggest that the derivation of stem cells from parthenogenetic embryos, for the purposes of therapeutic cloning, may be ineffective.
Collapse
Affiliation(s)
- Lidia Hernandez
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
44
|
Yao X, Hu JF, Daniels M, Shiran H, Zhou X, Yan H, Lu H, Zeng Z, Wang Q, Li T, Hoffman AR. A methylated oligonucleotide inhibits IGF2 expression and enhances survival in a model of hepatocellular carcinoma. J Clin Invest 2003. [PMID: 12531883 DOI: 10.1172/jci200315109] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IGF-II is a mitogenic peptide that has been implicated in hepatocellular oncogenesis. Since the silencing of gene expression is frequently associated with cytosine methylation at cytosine-guanine (CpG) dinucleotides, we designed a methylated oligonucleotide (MON1) complementary to a region encompassing IGF2 promoter P4 in an attempt to induce DNA methylation at that locus and diminish IGF2 mRNA levels. MON1 specifically inhibited IGF2 mRNA accumulation in vitro, whereas an oligonucleotide (ON1) with the same sequence but with nonmethylated cytosines had no effect on IGF2 mRNA abundance. MON1 treatment led to the specific induction of de novo DNA methylation in the region of IGF2 promoter hP4. Cells from a human hepatocellular carcinoma (HCC) cell line, Hep 3B, were implanted into the livers of nude mice, resulting in the growth of large tumors. Animals treated with MON1 had markedly prolonged survival as compared with those animals treated with saline or a truncated methylated oligonucleotide that did not alter IGF2 mRNA levels in vitro. This study demonstrates that a methylated sense oligonucleotide can be used to induce epigenetic changes in the IGF2 gene and that inhibition of IGF2 mRNA accumulation may lead to enhanced survival in a model of HCC.
Collapse
Affiliation(s)
- Xiaoming Yao
- Medical Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Yao X, Hu JF, Daniels M, Shiran H, Zhou X, Yan H, Lu H, Zeng Z, Wang Q, Li T, Hoffman AR. A methylated oligonucleotide inhibits IGF2 expression and enhances survival in a model of hepatocellular carcinoma. J Clin Invest 2003; 111:265-73. [PMID: 12531883 PMCID: PMC151856 DOI: 10.1172/jci15109] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IGF-II is a mitogenic peptide that has been implicated in hepatocellular oncogenesis. Since the silencing of gene expression is frequently associated with cytosine methylation at cytosine-guanine (CpG) dinucleotides, we designed a methylated oligonucleotide (MON1) complementary to a region encompassing IGF2 promoter P4 in an attempt to induce DNA methylation at that locus and diminish IGF2 mRNA levels. MON1 specifically inhibited IGF2 mRNA accumulation in vitro, whereas an oligonucleotide (ON1) with the same sequence but with nonmethylated cytosines had no effect on IGF2 mRNA abundance. MON1 treatment led to the specific induction of de novo DNA methylation in the region of IGF2 promoter hP4. Cells from a human hepatocellular carcinoma (HCC) cell line, Hep 3B, were implanted into the livers of nude mice, resulting in the growth of large tumors. Animals treated with MON1 had markedly prolonged survival as compared with those animals treated with saline or a truncated methylated oligonucleotide that did not alter IGF2 mRNA levels in vitro. This study demonstrates that a methylated sense oligonucleotide can be used to induce epigenetic changes in the IGF2 gene and that inhibition of IGF2 mRNA accumulation may lead to enhanced survival in a model of HCC.
Collapse
Affiliation(s)
- Xiaoming Yao
- Medical Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California 94304, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lopez T, Hanahan D. Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 2002; 1:339-53. [PMID: 12086849 DOI: 10.1016/s1535-6108(02)00055-7] [Citation(s) in RCA: 220] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In a prototypical model of multistage tumorigenesis involving pancreatic islets in RIP1-Tag2 transgenic mice, activation of insulin-like growth factor II (IGF-II) was previously shown to serve as a survival factor that inhibited apoptosis. Now IGF-1R, the receptor tyrosine kinase for IGF-II, has been found to be variably upregulated, first uniformly in dysplastic and angiogenic progenitors and then focally at the margins and in invasive regions of carcinomas. When the levels of IGF-1R were forcibly elevated throughout islet tumorigenesis, progression was accelerated at all stages in the pathway, although apoptosis was not differentially suppressed. Notably, encapsulated tumors were absent; instead, invasive carcinomas with downregulated E-cadherin were prevalent, and the majority of mice had local lymph node metastasis.
Collapse
Affiliation(s)
- Theresa Lopez
- Department of Biochemistry & Biophysics and the UCSF Diabetes and Comprehensive Cancer Centers, University of California, San Francisco, 94143, USA
| | | |
Collapse
|
47
|
Hassan AB, Macaulay VM. The insulin-like growth factor system as a therapeutic target in colorectal cancer. Ann Oncol 2002; 13:349-56. [PMID: 11996463 DOI: 10.1093/annonc/mdf096] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The purpose of this review is to examine recent evidence that investigates the role of the insulin-like growth factor (IGF) system in colorectal cancer. We concentrate on the evidence that makes the case for the investigation of strategies that might be used to disrupt the IGF system in prevention and treatment. Even though the weight of evidence suggests that components of the IGF system may be appropriate targets, there are a lack of studies that make a systematic characterisation of all the system components in human colorectal cancer. It is anticipated that this information, and the new therapeutic molecules which follow, will impact on the prevention and treatment of patients with this disease.
Collapse
Affiliation(s)
- A B Hassan
- Department of Zoology, University of Oxford, UK.
| | | |
Collapse
|
48
|
Abstract
Lack of insulin production or abnormalities affecting insulin secretion are key to the development of almost all forms of diabetes, including the common type 1 (insulin-dependent) and type 2 (non-insulin-dependent) diabetes and the more rare forms of maturity-onset diabetes of the young (MODY). Because insulin has such a central role in the pathogenesis of both forms of diabetes, the insulin gene (INS) has always been considered a candidate susceptibility gene. A number of studies have shown that the allelic variation and parent-of-origin effects affect the transmission and expression of the insulin gene in pancreatic beta-cells and extra-pancreatic tissues. These observations have led to the formulation of new hypotheses to explain the biological mechanisms by which functional differences in the expression of the insulin gene may contribute to diabetes susceptibility.
Collapse
Affiliation(s)
- Alberto Pugliese
- Immunogenetics, Diabetes Research Institute, University of Miami School of Medicine, Miami, Florida, FL 33136, USA.
| | | |
Collapse
|
49
|
Burns JL, Hassan AB. Cell survival and proliferation are modified by insulin-like growth factor 2 between days 9 and 10 of mouse gestation. Development 2001; 128:3819-30. [PMID: 11585807 DOI: 10.1242/dev.128.19.3819] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The size of mammalian species involves the interaction of multiple genetic modifiers that control the timing and extent of growth mechanisms. Disruption of the paternal allele of the imprinted embryonic gene coding for insulin-like growth factor 2 (IGF2, Igf2+m/−p), results in viable mice that are 60% the weight of wild-type littermates. Differences in weight are first detected at embryonic day (E) 11, and the growth deficit is maintained throughout life. We report the mechanisms that account for this unusual phenotype. In order to quantify growth, we used novel methods to generate single cell suspensions of post-implantation mouse embryos. We were then able to quantify cell number, cell proliferation and cell death between E8.5 and E11.5 using flow cytometry. Determination of total embryo cell number also allowed us to time litters by a method other than by plugging. Wild-type and Igf2+m/−p embryos accumulated similar total cell numbers up to E9.25, but cell number began to diverge by around E9.5, with significant differences by E11 (75% of wild type). A relative increase in pyknotic nuclei, sub-GI cytometry counts and caspase activity, all indicative of cell death, occurred in Igf2+m/−p embryos at E9.25, reverting to wild-type levels by E9.75. This was followed at E9.75 by a significant reduction in the proportion of cells in S phase, quantified by S-phase cytometry counts and BrdU labelling. No significant differences in cell size were detected. We conclude that the majority of the cell number differences between wild-type and Igf2+m/−p mice can be accounted for by modification of cell survival and proliferation during the period (E9 to E10) of post-implantation development.
Collapse
Affiliation(s)
- J L Burns
- Department of Zoology, University of Oxford, South Parks Road, Oxford OX1 3PS, UK
| | | |
Collapse
|
50
|
Affiliation(s)
- A P Feinberg
- Institute of Genetic Medicine, Departments of Medicine, Molecular Biology and Genetics, and Oncology, Johns Hopkins University School of Medicine, 1064 Ross, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|