1
|
Zhu R, Liu X, Zhang X, Zhong Z, Qi S, Jin R, Gu Y, Wang Y, Ling C, Chen K, Ye D, Yu FX. Gene therapy for diffuse pleural mesotheliomas in preclinical models by concurrent expression of NF2 and SuperHippo. Cell Rep Med 2024; 5:101763. [PMID: 39368484 PMCID: PMC11513813 DOI: 10.1016/j.xcrm.2024.101763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/03/2024] [Accepted: 09/12/2024] [Indexed: 10/07/2024]
Abstract
Diffuse pleural mesothelioma (DPM) is a lethal cancer with a poor prognosis and limited treatment options. The Hippo signaling pathway genes, such as NF2 and LATS1/2, are frequently mutated in DPM, indicating a tumor suppressor role in the development of DPM. Here, we show that in DPM cell lines lacking NF2 and in mice with a conditional Nf2 knockout, downregulation of WWC proteins, another family of Hippo pathway regulators, accelerates DPM progression. Conversely, the expression of SuperHippo, a WWC-derived minigene, effectively enhances Hippo signaling and suppresses DPM development. Moreover, the adeno-associated virus serotype 6 (AAV6) has been engineered to deliver both NF2 and SuperHippo genes into mesothelial cells, which substantially impedes tumor growth in xenograft and genetic DPM models and prolongs the median survival of mice. These findings serve as a proof of concept for the potential use of gene therapy targeting the Hippo pathway to treat DPM.
Collapse
Affiliation(s)
- Rui Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xincheng Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Sixian Qi
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ruxin Jin
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuan Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dan Ye
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
2
|
Hosur V, Erhardt V, Hartig E, Lorenzo K, Megathlin H, Tarchini B. Large-Scale Genome-Wide Optimization and Prediction of the Cre Recombinase System for Precise Genome Manipulation in Mice. RESEARCH SQUARE 2024:rs.3.rs-4595968. [PMID: 39011108 PMCID: PMC11247941 DOI: 10.21203/rs.3.rs-4595968/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
The Cre-Lox recombination system is a powerful tool in mouse genetics, offering spatial-temporal control over gene expression and facilitating the large-scale generation of conditional knockout mice. Its versatility also extends to other research models, such as rats, pigs, and zebrafish. However, the Cre-Lox technology presents a set of challenges that includes high costs, a time-intensive process, and the occurrence of unpredictable recombination events, which can lead to unexpected phenotypic outcomes. To better understand factors affecting recombination, we embarked on a systematic and genome-wide analysis of Cre-mediated recombination in mice. To ensure uniformity and reproducibility, we generated 11 novel strains with conditional alleles at the ROSA26 locus, utilizing a single inbred mouse strain background, C57BL/6J. We examined several factors influencing Cre-recombination, including the inter-loxP distance, mutant loxP sites, the zygosity of the conditional alleles, chromosomal location, and the age of the breeders. We discovered that the selection of the Cre-driver strain profoundly impacts recombination efficiency. We also found that successful and complete recombination is best achieved when loxP sites are spaced between 1 to 4 kb apart, with mutant loxP sites facilitating recombination at distances of 1 to 3 kb. Furthermore, we demonstrate that complete recombination does not occur at an inter-loxP distance of ≥ 15 kb with wildtype loxP sites, nor at a distance of ≥ 7 kb with mutant lox71/66 sites. Interestingly, the age of the Cre-driver mouse at the time of breeding emerged as a critical factor in recombination efficiency, with best results observed between 8 and 20 weeks old. Moreover, crossing heterozygous floxed alleles with the Cre-driver strain resulted in more efficient recombination than using homozygous floxed alleles. Lastly, maintaining an inter-loxP distance of 4 kb or less ensures efficient recombination of the conditional allele, regardless of the chromosomal location. While CRISPR/Cas has revolutionized genome editing in mice, Cre-Lox technology remains a cornerstone for the generation of sophisticated alleles and for precise control of gene expression in mice. The knowledge gained here will enable investigators to select a Cre-Lox approach that is most efficient for their desired outcome in the generation of both germline and non-germline mouse models of human disease, thereby reducing time and cost of Cre-Lox technology-mediated genome modification.
Collapse
Affiliation(s)
| | | | - Elli Hartig
- The Jackson Laboratory for Mammalian Genetics
| | | | | | | |
Collapse
|
3
|
Erhardt V, Hartig E, Lorenzo K, Megathlin HR, Tarchini B, Hosur V. Large-Scale Genome-Wide Optimization and Prediction of the Cre Recombinase System for Precise Genome Manipulation in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599022. [PMID: 38948742 PMCID: PMC11212873 DOI: 10.1101/2024.06.14.599022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Cre-Lox recombination system is a powerful tool in mouse genetics, offering spatial-temporal control over gene expression and facilitating the large-scale generation of conditional knockout mice. Its versatility also extends to other research models, such as rats, pigs, and zebrafish. However, the Cre-Lox technology presents a set of challenges that includes high costs, a time-intensive process, and the occurrence of unpredictable recombination events, which can lead to unexpected phenotypic outcomes. To better understand factors affecting recombination, we embarked on a systematic and genome-wide analysis of Cre-mediated recombination in mice. To ensure uniformity and reproducibility, we generated 11 novel strains with conditional alleles at the ROSA26 locus, utilizing a single inbred mouse strain background, C57BL/6J. We examined several factors influencing Cre-recombination, including the inter-loxP distance, mutant loxP sites, the zygosity of the conditional alleles, chromosomal location, and the age of the breeders. We discovered that the selection of the Cre-driver strain profoundly impacts recombination efficiency. We also found that successful and complete recombination is best achieved when loxP sites are spaced between 1 to 4 kb apart, with mutant loxP sites facilitating recombination at distances of 1 to 3 kb. Furthermore, we demonstrate that complete recombination does not occur at an inter-loxP distance of ≥ 15 kb with wildtype loxP sites, nor at a distance of ≥ 7 kb with mutant lox71/66 sites. Interestingly, the age of the Cre-driver mouse at the time of breeding emerged as a critical factor in recombination efficiency, with best results observed between 8 and 20 weeks old. Moreover, crossing heterozygous floxed alleles with the Cre-driver strain resulted in more efficient recombination than using homozygous floxed alleles. Lastly, maintaining an inter-loxP distance of 4 kb or less ensures efficient recombination of the conditional allele, regardless of the chromosomal location. While CRISPR/Cas has revolutionized genome editing in mice, Cre-Lox technology remains a cornerstone for the generation of sophisticated alleles and for precise control of gene expression in mice. The knowledge gained here will enable investigators to select a Cre-Lox approach that is most efficient for their desired outcome in the generation of both germline and non-germline mouse models of human disease, thereby reducing time and cost of Cre-Lox technology-mediated genome modification.
Collapse
Affiliation(s)
- Valerie Erhardt
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| | - Elli Hartig
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
- Tufts University School of Medicine, Boston, MA
| | - Kristian Lorenzo
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
- The Roux Institute at Northeastern University, Portland, ME
| | - Hannah R Megathlin
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
- Graduate School of Biomedical Sciences and Engineering, UMaine, Orono, ME
| | - Basile Tarchini
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
- Tufts University School of Medicine, Boston, MA
| | - Vishnu Hosur
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME
| |
Collapse
|
4
|
Abstract
All cancers arise from normal cells whose progeny acquire the cancer-initiating mutations and epigenetic modifications leading to frank tumorigenesis. The identity of those "cells-of-origin" has historically been a source of controversy across tumor types, as it has not been possible to witness the dynamic events giving rise to human tumors. Genetically engineered mouse models (GEMMs) of cancer provide an invaluable substitute, enabling researchers to interrogate the competence of various naive cellular compartments to initiate tumors in vivo. Researchers using these models have relied on lineage-specific promoters, knowledge of preneoplastic disease states in humans, and technical advances allowing more precise manipulations of the mouse germline. These approaches have given rise to the emerging view that multiple lineages within a given organ may generate tumors with similar histopathology. Here, we review some of the key studies leading to this conclusion in solid tumors and highlight the biological and clinical ramifications.
Collapse
Affiliation(s)
- Jason R Pitarresi
- Division of Hematology and Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655, USA
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts 01655, USA
| | - Ben Z Stanger
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
5
|
Solta A, Ernhofer B, Boettiger K, Megyesfalvi Z, Heeke S, Hoda MA, Lang C, Aigner C, Hirsch FR, Schelch K, Döme B. Small cells - big issues: biological implications and preclinical advancements in small cell lung cancer. Mol Cancer 2024; 23:41. [PMID: 38395864 PMCID: PMC10893629 DOI: 10.1186/s12943-024-01953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Current treatment guidelines refer to small cell lung cancer (SCLC), one of the deadliest human malignancies, as a homogeneous disease. Accordingly, SCLC therapy comprises chemoradiation with or without immunotherapy. Meanwhile, recent studies have made significant advances in subclassifying SCLC based on the elevated expression of the transcription factors ASCL1, NEUROD1, and POU2F3, as well as on certain inflammatory characteristics. The role of the transcription regulator YAP1 in defining a unique SCLC subset remains to be established. Although preclinical analyses have described numerous subtype-specific characteristics and vulnerabilities, the so far non-existing clinical subtype distinction may be a contributor to negative clinical trial outcomes. This comprehensive review aims to provide a framework for the development of novel personalized therapeutic approaches by compiling the most recent discoveries achieved by preclinical SCLC research. We highlight the challenges faced due to limited access to patient material as well as the advances accomplished by implementing state-of-the-art models and methodologies.
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Büsra Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Simon Heeke
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Thoracic Oncology, Mount Sinai Health System, Tisch Cancer Institute, New York, NY, USA.
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Balazs Döme
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
6
|
Liu F, Wang F, He J, Zhou S, Luo M. Correlation between KRAS mutation subtypes and prognosis in Chinese advanced non-squamous non-small cell lung cancer patients. Cancer Med 2023. [PMID: 37140194 DOI: 10.1002/cam4.5995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023] Open
Abstract
PURPOSE The relationship between mutant KRAS and the risk of disease progression and death in advanced non-squamous non-small cell lung cancer (NSCLC) is still controversial among current studies, and the effects of distinct KRAS mutations on prognosis may be different. This study aimed to further investigate the association between them. PATIENTS AND METHODS Of the 184 patients eventually included in the study, 108 had KRAS wild type (WT) and 76 had KRAS mutant type (MT). Kaplan-Meier curves were plotted to describe the survival for patients among groups, while log-rank tests were conducted to evaluate the survival differences. The univariate and multivariate Cox regression were performed to identify predictors, and subgroup analysis was used to verify the interaction effect. RESULTS Similar efficacy of first-line therapy was observed for KRAS MT and WT patients (p = 0.830). The association between KRAS mutation and progression-free survival (PFS) was not significant in univariate analysis (hazard ratio [HR] = 0.94; 95% CI, 0.66-1.35), and no KRAS mutation subtype significantly affected PFS. However, KRAS mutation and KRAS non-G12C were associated with increased risk of death compared to KRAS WT in univariate and multivariate analysis. Univariate and multivariate analysis also confirmed that chemotherapy combined with antiangiogenesis or immunotherapy in the KRAS mutation group was associated with decreased risk of disease progression. However, the overall survival (OS) among KRAS mutant patients received different first-line treatments did not significantly differ. CONCLUSION KRAS mutations and their subtypes are not independent negative predictors of PFS, while KRAS mutation and KRAS non-G12C were independent prognostic factors for OS. Chemotherapy combined with antiangiogenesis or immunotherapy conferred decreased risk of disease progression to KRAS mutation patients compared to single chemotherapy.
Collapse
Affiliation(s)
- Feiwen Liu
- The Third Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Zhuang Autonomous Region, China
| | - Fang Wang
- Guangxi Qianhai Life Hospital, Nanning City, Guangxi Zhuang Autonomous Region, China
| | - Jianbo He
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning City, China
| | - Shaozhang Zhou
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Guangxi Zhuang Autonomous Region, Nanning City, China
| | - Min Luo
- The Third Affiliated Hospital of Guangxi Medical University, Nanning City, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
7
|
Balážová K, Clevers H, Dost AFM. The role of macrophages in non-small cell lung cancer and advancements in 3D co-cultures. eLife 2023; 12:82998. [PMID: 36809334 PMCID: PMC9943070 DOI: 10.7554/elife.82998] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/09/2023] [Indexed: 02/23/2023] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths worldwide. Traditional therapeutic approaches such as chemotherapy or radiotherapy have provided only a marginal improvement in the treatment of lung carcinomas. Inhibitors targeting specific genetic aberrations present in non-small cell lung cancer (NSCLC), the most common subtype (85%), have improved the prognostic outlook, but due to the complexity of the LC mutational spectrum, only a fraction of patients benefit from these targeted molecular therapies. More recently, the realization that the immune infiltrate surrounding solid tumors can foster tumor-promoting inflammation has led to the development and implementation of anticancer immunotherapies in the clinic. In NSCLC, one of the most abundant leukocyte infiltrates is macrophages. These highly plastic phagocytes, which are part of the cellular repertoire of the innate immunity, can have a pivotal role in early NSCLC establishment, malignant progression, and tumor invasion. Emerging macrophage-targeting therapies have been focused on the re-differentiation of the macrophages toward an antitumorigenic phenotype, depletion of tumor-promoting macrophage subtypes, or combination therapies combining traditional cytotoxic treatments with immunotherapeutic agents. The most extensively used models employed for the exploration of NSCLC biology and therapy have been 2D cell lines and murine models. However, studying cancer immunology requires appropriately complex models. 3D platforms, including organoid models, are quickly advancing powerful tools to study immune cell-epithelial cell interactions within the tumor microenvironment. Co-cultures of immune cells along with NSCLC organoids allow for an in vitro observation of the tumor microenvironment dynamics closely resembling in vivo settings. Ultimately, the implementation of 3D organoid technology into tumor microenvironment-modeling platforms might facilitate the exploration of macrophage-targeted therapies in NSCLC immunotherapeutic research, thus establishing a new frontier in NSCLC treatment.
Collapse
Affiliation(s)
- Katarína Balážová
- Hubrecht Institute for Developmental Biology and Stem Cell Research-KNAW & University Medical Centre UtrechtUtrechtNetherlands,Oncode Institute, Hubrecht Institute-KNAWUtrechtNetherlands
| | - Hans Clevers
- Roche Pharma Research and early DevelopmentBaselSwitzerland
| | - Antonella FM Dost
- Hubrecht Institute for Developmental Biology and Stem Cell Research-KNAW & University Medical Centre UtrechtUtrechtNetherlands,Oncode Institute, Hubrecht Institute-KNAWUtrechtNetherlands
| |
Collapse
|
8
|
Zitnay RG, Herron MR, Carney KR, Potter S, Emerson LL, Weiss JA, Mendoza MC. Mechanics of lung cancer: A finite element model shows strain amplification during early tumorigenesis. PLoS Comput Biol 2022; 18:e1010153. [PMID: 36279309 PMCID: PMC9632844 DOI: 10.1371/journal.pcbi.1010153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 11/03/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Early lung cancer lesions develop within a unique microenvironment that undergoes constant cyclic stretch from respiration. While tumor stiffening is an established driver of tumor progression, the contribution of stress and strain to lung cancer is unknown. We developed tissue scale finite element models of lung tissue to test how early lesions alter respiration-induced strain. We found that an early tumor, represented as alveolar filling, amplified the strain experienced in the adjacent alveolar walls. Tumor stiffening further increased the amplitude of the strain in the adjacent alveolar walls and extended the strain amplification deeper into the normal lung. In contrast, the strain experienced in the tumor proper was less than the applied strain, although regions of amplification appeared at the tumor edge. Measurements of the alveolar wall thickness in clinical and mouse model samples of lung adenocarcinoma (LUAD) showed wall thickening adjacent to the tumors, consistent with cellular response to strain. Modeling alveolar wall thickening by encircling the tumor with thickened walls moved the strain amplification radially outward, to the next adjacent alveolus. Simulating iterative thickening in response to amplified strain produced tracks of thickened walls. We observed such tracks in early-stage clinical samples. The tracks were populated with invading tumor cells, suggesting that strain amplification in very early lung lesions could guide pro-invasive remodeling of the tumor microenvironment. The simulation results and tumor measurements suggest that cells at the edge of a lung tumor and in surrounding alveolar walls experience increased strain during respiration that could promote tumor progression.
Collapse
Affiliation(s)
- Rebecca G. Zitnay
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
| | - Michael R. Herron
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Keith R. Carney
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, United States of America
| | - Scott Potter
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Lyska L. Emerson
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Jeffrey A. Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, Salt Lake City, Utah, United States of America
| | - Michelle C. Mendoza
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, Salt Lake City, Utah, United States of America
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
9
|
Rashbrook VS, Brash JT, Ruhrberg C. Cre toxicity in mouse models of cardiovascular physiology and disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:806-816. [PMID: 37692772 PMCID: PMC7615056 DOI: 10.1038/s44161-022-00125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 09/12/2023]
Abstract
The Cre-LoxP system provides a widely used method for studying gene requirements in the mouse as the main mammalian genetic model organism. To define the molecular and cellular mechanisms that underlie cardiovascular development, function and disease, various mouse strains have been engineered that allow Cre-LoxP-mediated gene targeting within specific cell types of the cardiovascular system. Despite the usefulness of this system, evidence is accumulating that Cre activity can have toxic effects in cells, independently of its ability to recombine pairs of engineered LoxP sites in target genes. Here, we have gathered published evidence for Cre toxicity in cells and tissues relevant to cardiovascular biology and provide an overview of mechanisms proposed to underlie Cre toxicity. Based on this knowledge, we propose that each study utilising the Cre-LoxP system to investigate gene function in the cardiovascular system should incorporate appropriate controls to account for Cre toxicity.
Collapse
Affiliation(s)
- Victoria S. Rashbrook
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T. Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
10
|
Thege FI, Rupani DN, Barathi BB, Manning SL, Maitra A, Rhim AD, Wörmann SM. A Programmable In Vivo CRISPR Activation Model Elucidates the Oncogenic and Immunosuppressive Functions of MYC in Lung Adenocarcinoma. Cancer Res 2022; 82:2761-2776. [PMID: 35666804 PMCID: PMC9357118 DOI: 10.1158/0008-5472.can-21-4009] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 04/18/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023]
Abstract
Conventional genetically engineered mouse models (GEMM) are time-consuming, laborious, and offer limited spatiotemporal control. Here, we describe the development of a streamlined platform for in vivo gene activation using CRISPR activation (CRISPRa) technology. Unlike conventional GEMMs, this model system allows for flexible, sustained, and timed activation of one or more target genes using single or pooled lentiviral guides. Myc and Yap1 were used as model oncogenes to demonstrate gene activation in primary pancreatic organoid cultures in vitro and enhanced tumorigenic potential in Myc-activated organoids when transplanted orthotopically in vivo. Implementation of this model as an autochthonous lung cancer model showed that transduction-mediated activation of Myc led to accelerated tumor progression and significantly reduced overall survival relative to nontargeted tumor controls. Furthermore, Myc activation led to the acquisition of an immune suppressive, "cold" tumor microenvironment. Cross-species validation of these results using publicly available RNA/DNA-seq datasets linked MYC to a previously described immunosuppressive molecular subtype in patient tumors, thus identifying a patient cohort that may benefit from combined MYC- and immune-targeted therapies. Overall, this work demonstrates how CRISPRa can be used for rapid functional validation of putative oncogenes and may allow for the identification and evaluation of potential metastatic and oncogenic drivers through competitive screening. SIGNIFICANCE A streamlined platform for programmable CRISPR gene activation enables rapid evaluation and functional validation of putative oncogenes in vivo.
Collapse
Affiliation(s)
- Fredrik I. Thege
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
- CORRESPONDANCE: Fredrik I. Thege, , Sonja M. Wörmann, , MD Anderson Cancer Center, Zayed Building, Z3.2065, 6565 MD Anderson Blvd., Houston, TX 77030, USA
| | - Dhwani N. Rupani
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bhargavi B. Barathi
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sara L. Manning
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anirban Maitra
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Andrew D. Rhim
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sonja M. Wörmann
- Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
- CORRESPONDANCE: Fredrik I. Thege, , Sonja M. Wörmann, , MD Anderson Cancer Center, Zayed Building, Z3.2065, 6565 MD Anderson Blvd., Houston, TX 77030, USA
| |
Collapse
|
11
|
Arnal-Estapé A, Foggetti G, Starrett JH, Nguyen DX, Politi K. Preclinical Models for the Study of Lung Cancer Pathogenesis and Therapy Development. Cold Spring Harb Perspect Med 2021; 11:a037820. [PMID: 34518338 PMCID: PMC8634791 DOI: 10.1101/cshperspect.a037820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Experimental preclinical models have been a cornerstone of lung cancer translational research. Work in these model systems has provided insights into the biology of lung cancer subtypes and their origins, contributed to our understanding of the mechanisms that underlie tumor progression, and revealed new therapeutic vulnerabilities. Initially patient-derived lung cancer cell lines were the main preclinical models available. The landscape is very different now with numerous preclinical models for research each with unique characteristics. These include genetically engineered mouse models (GEMMs), patient-derived xenografts (PDXs) and three-dimensional culture systems ("organoid" cultures). Here we review the development and applications of these models and describe their contributions to lung cancer research.
Collapse
Affiliation(s)
- Anna Arnal-Estapé
- Department of Pathology
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | | | | | - Don X Nguyen
- Department of Pathology
- Department of Internal Medicine (Section of Medical Oncology)
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Katerina Politi
- Department of Pathology
- Department of Internal Medicine (Section of Medical Oncology)
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
12
|
Vallabhaneni S, Liu J, Morel M, Wang J, DeMayo FJ, Long W. Conditional ERK3 overexpression cooperates with PTEN deletion to promote lung adenocarcinoma formation in mice. Mol Oncol 2021; 16:1184-1199. [PMID: 34719109 PMCID: PMC8895443 DOI: 10.1002/1878-0261.13132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 11/23/2022] Open
Abstract
ERK3, officially known as mitogen‐activated protein kinase 6 (MAPK6), is a poorly studied mitogen‐activated protein kinase (MAPK). Recent studies have revealed the upregulation of ERK3 expression in cancer and suggest an important role for ERK3 in promoting cancer cell growth and invasion in some cancers, in particular lung cancer. However, it is unknown whether ERK3 plays a role in spontaneous tumorigenesis in vivo. To determine the role of ERK3 in lung tumorigenesis, we created a conditional ERK3 transgenic mouse line in which ERK3 transgene expression is controlled by Cre recombinase. By crossing these transgenic mice with a mouse line harboring a lung tissue–specific Cre recombinase transgene driven by a club cell secretory protein gene promoter (CCSP‐iCre), we have found that conditional ERK3 overexpression cooperates with phosphatase and tensin homolog (PTEN) deletion to induce the formation of lung adenocarcinomas (LUADs). Mechanistically, ERK3 overexpression stimulates activating phosphorylations of erb‐b2 receptor tyrosine kinases 2 and 3 (ERBB2 and ERBB3) by upregulating Sp1 transcription factor (SP1)–mediated gene transcription of neuregulin 1 (NRG1), a potent ligand for ERBB2/ERBB3. Our study has revealed a bona fide tumor‐promoting role for ERK3 using genetically engineered mouse models. Together with previous findings showing the roles of ERK3 in cultured cells and in a xenograft lung tumor model, our findings corroborate that ERK3 acts as an oncoprotein in promoting LUAD development and progression.
Collapse
Affiliation(s)
- Sreeram Vallabhaneni
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Jian Liu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, 314400, China.,Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - Marion Morel
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| | - Jixin Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, 314400, China.,Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - Francesco J DeMayo
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park (RTP), NC, USA
| | - Weiwen Long
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| |
Collapse
|
13
|
Hutton C, Heider F, Blanco-Gomez A, Banyard A, Kononov A, Zhang X, Karim S, Paulus-Hock V, Watt D, Steele N, Kemp S, Hogg EKJ, Kelly J, Jackstadt RF, Lopes F, Menotti M, Chisholm L, Lamarca A, Valle J, Sansom OJ, Springer C, Malliri A, Marais R, Pasca di Magliano M, Zelenay S, Morton JP, Jørgensen C. Single-cell analysis defines a pancreatic fibroblast lineage that supports anti-tumor immunity. Cancer Cell 2021; 39:1227-1244.e20. [PMID: 34297917 PMCID: PMC8443274 DOI: 10.1016/j.ccell.2021.06.017] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 04/19/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022]
Abstract
Fibroblasts display extensive transcriptional heterogeneity, yet functional annotation and characterization of their heterocellular relationships remains incomplete. Using mass cytometry, we chart the stromal composition of 18 murine tissues and 5 spontaneous tumor models, with an emphasis on mesenchymal phenotypes. This analysis reveals extensive stromal heterogeneity across tissues and tumors, and identifies coordinated relationships between mesenchymal and immune cell subsets in pancreatic ductal adenocarcinoma. Expression of CD105 demarks two stable and functionally distinct pancreatic fibroblast lineages, which are also identified in murine and human healthy tissues and tumors. Whereas CD105-positive pancreatic fibroblasts are permissive for tumor growth in vivo, CD105-negative fibroblasts are highly tumor suppressive. This restrictive effect is entirely dependent on functional adaptive immunity. Collectively, these results reveal two functionally distinct pancreatic fibroblast lineages and highlight the importance of mesenchymal and immune cell interactions in restricting tumor growth.
Collapse
Affiliation(s)
- Colin Hutton
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Felix Heider
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Adrian Blanco-Gomez
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Antonia Banyard
- Flow Cytometry Core, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Alexander Kononov
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Xiaohong Zhang
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Saadia Karim
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, UK
| | - Viola Paulus-Hock
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, UK
| | - Dale Watt
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, UK
| | - Nina Steele
- University of Michigan, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, Ann Arbor, MI 48109, USA
| | - Samantha Kemp
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA; Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Elizabeth K J Hogg
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Joanna Kelly
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Rene-Filip Jackstadt
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, UK
| | - Filipa Lopes
- Drug Discovery Unit, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Matteo Menotti
- Cell Signalling, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Luke Chisholm
- Molecular Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - Juan Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Switchback Road, Garscube Estate, Glasgow G61 1QH, UK
| | - Caroline Springer
- Drug Discovery Unit, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Angeliki Malliri
- Cell Signalling, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Richard Marais
- Molecular Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Marina Pasca di Magliano
- University of Michigan, Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Santiago Zelenay
- Cancer Immunity and Inflammation, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK
| | - Jennifer P Morton
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Switchback Road, Garscube Estate, Glasgow G61 1QH, UK
| | - Claus Jørgensen
- Systems Oncology, Cancer Research UK Manchester Institute, Alderley Park, Manchester SK10 4TG, UK.
| |
Collapse
|
14
|
Rosigkeit S, Kruchem M, Thies D, Kreft A, Eichler E, Boegel S, Jansky S, Siegl D, Kaps L, Pickert G, Haehnel P, Kindler T, Hartwig UF, Guerra C, Barbacid M, Schuppan D, Bockamp E. Definitive evidence for Club cells as progenitors for mutant Kras/Trp53-deficient lung cancer. Int J Cancer 2021; 149:1670-1682. [PMID: 34331774 DOI: 10.1002/ijc.33756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022]
Abstract
Accumulating evidence suggests that both the nature of oncogenic lesions and the cell-of-origin can strongly influence cancer histopathology, tumor aggressiveness and response to therapy. Although oncogenic Kras expression and loss of Trp53 tumor suppressor gene function have been demonstrated to initiate murine lung adenocarcinomas (LUADs) in alveolar type II (AT2) cells, clear evidence that Club cells, representing the second major subset of lung epithelial cells, can also act as cells-of-origin for LUAD is lacking. Equally, the exact anatomic location of Club cells that are susceptible to Kras transformation and the resulting tumor histotype remains to be established. Here, we provide definitive evidence for Club cells as progenitors for LUAD. Using in vivo lineage tracing, we find that a subset of Kras12V -expressing and Trp53-deficient Club cells act as precursors for LUAD and we define the stepwise trajectory of Club cell-initiated tumors leading to lineage marker conversion and aggressive LUAD. Our results establish Club cells as cells-of-origin for LUAD and demonstrate that Club cell-initiated tumors have the potential to develop aggressive LUAD.
Collapse
Affiliation(s)
- Sebastian Rosigkeit
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Marie Kruchem
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Dorothe Thies
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Andreas Kreft
- Institute of Pathology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Emma Eichler
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Sebastian Boegel
- Department of Internal Medicine, University Center of Autoimmunity, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Sandrine Jansky
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Dominik Siegl
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Leonard Kaps
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Geethanjali Pickert
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Patricia Haehnel
- III. Department of Medicine Hematology, Internal Oncology and Pneumology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Kindler
- III. Department of Medicine Hematology, Internal Oncology and Pneumology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Udo F Hartwig
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,III. Department of Medicine Hematology, Internal Oncology and Pneumology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Carmen Guerra
- Experimental Oncology, Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Mariano Barbacid
- Experimental Oncology, Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Detlef Schuppan
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ernesto Bockamp
- Institute of Translational Immunology (TIM), University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
15
|
Pulmonary Inflammation and KRAS Mutation in Lung Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33788188 DOI: 10.1007/978-3-030-63046-1_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2023]
Abstract
Chronic lung infection and lung cancer are two of the most important pulmonary diseases. Respiratory infection and its associated inflammation have been increasingly investigated for their role in increasing the risk of respiratory diseases including chronic obstructive pulmonary disease (COPD) and lung cancer. Kirsten rat sarcoma viral oncogene (KRAS) is one of the most important regulators of cell proliferation, differentiation, and survival. KRAS mutations are among the most common drivers of cancer. Lung cancer harboring KRAS mutations accounted for ~25% of the incidence but the relationship between KRAS mutation and inflammation remains unclear. In this chapter, we will describe the roles of KRAS mutation in lung cancer and how elevated inflammatory responses may increase KRAS mutation rate and create a vicious cycle of chronic inflammation and KRAS mutation that likely results in persistent potentiation for KRAS-associated lung tumorigenesis. We will discuss in this chapter regarding the studies of KRAS gene mutations in specimens from lung cancer patients and in animal models for investigating the role of inflammation in increasing the risk of lung tumorigenesis driven primarily by oncogenic KRAS.
Collapse
|
16
|
Abstract
Cigarette smoking is the major culprit of chronic lung diseases and the most dominant risk factor for the development of both lung cancer and chronic obstructive pulmonary disease (COPD). In addition, chronic inflammation has been shown to increase the risk of lung cancer and COPD in clinical and epidemiological studies. For pulmonary disease-related research, mice are the most commonly used model system. Multiple lung cancer mouse models driven by targeted genetic alterations are used to evaluate the critical roles of oncogenes and tumor suppressor genes. These models are useful in addressing lung tumorigenesis associated with specific genetic changes, but they are not able to provide a global insight into cigarette smoke-induced carcinogenesis. To fill this knowledge gap, we developed a lung cancer model by treating mice with cigarette smoke carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) with/without repeated lipopolysaccharides (LPS) exposure in order to determine the role of chronic inflammation in lung tumorigenesis. Notably, combined LPS/NNK treatment increased tumor number, tumor incidence, and tumor area compared to NNK treatment alone. Therefore, this model offers a feasible approach to investigate lung cancer development on a more global level, determine the role of inflammation in carcinogenesis, and provide a tool for evaluating chemoprevention and immunotherapy.
Collapse
Affiliation(s)
- Marissa E Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Beth Kahkonen
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Chia-Hsin Liu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yuanpu Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
17
|
Badhai J, Pandey GK, Song JY, Krijgsman O, Bhaskaran R, Chandrasekaran G, Kwon MC, Bombardelli L, Monkhorst K, Grasso C, Zevenhoven J, van der Vliet J, Cozijnsen M, Krimpenfort P, Peeper D, van Lohuizen M, Berns A. Combined deletion of Bap1, Nf2, and Cdkn2ab causes rapid onset of malignant mesothelioma in mice. J Exp Med 2021; 217:151644. [PMID: 32271879 PMCID: PMC7971132 DOI: 10.1084/jem.20191257] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/09/2019] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
We have generated mouse models of malignant mesothelioma (MM) based upon disruption of the Bap1, Nf2, and Cdkn2ab tumor suppressor loci in various combinations as also frequently observed in human MM. Inactivation of all three loci in the mesothelial lining of the thoracic cavity leads to a highly aggressive MM that recapitulates the histological features and gene expression profile observed in human patients. The tumors also show a similar inflammatory phenotype. Bap1 deletion alone does not cause MM but dramatically accelerates MM development when combined with Nf2 and Cdkn2ab (hereafter BNC) disruption. The accelerated tumor development is accompanied by increased Polycomb repression and EZH2-mediated redistribution of H3K27me3 toward promoter sites with concomitant activation of PI3K and MAPK pathways. Treatment of BNC tumor–bearing mice with cisplatin and pemetrexed, the current frontline treatment, prolongs survival. This makes the autochthonous mouse model described here very well suited to explore the pathogenesis of MM and validate new treatment regimens for MM, including immunotherapy.
Collapse
Affiliation(s)
- Jitendra Badhai
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gaurav Kumar Pandey
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ji-Ying Song
- Department of Experimental Animal Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Oscar Krijgsman
- Oncode Institute, Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Rajith Bhaskaran
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Gayathri Chandrasekaran
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Min-Chul Kwon
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Lorenzo Bombardelli
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Kim Monkhorst
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Cristoforo Grasso
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - John Zevenhoven
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jan van der Vliet
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Miranda Cozijnsen
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Paul Krimpenfort
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Daniel Peeper
- Oncode Institute, Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maarten van Lohuizen
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Anton Berns
- Oncode Institute, Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
18
|
Abstract
As one of the most common forms of cancer, lung cancers present as a collection of different histological subtypes. These subtypes are characterized by distinct sets of driver mutations and phenotypic appearance, and they often show varying degrees of heterogenicity, aggressiveness, and response/resistance to therapy. Intriguingly, lung cancers are also capable of showing features of multiple subtypes or converting from one subtype to another. The intertumoral and intratumoral heterogeneity of lung cancers as well as incidences of subtype transdifferentiation raise the question of to what extent the tumor characteristics are dictated by the cell of origin rather than the acquired driver lesions. We provide here an overview of the studies in experimental mouse models that try to address this question. These studies convincingly show that both the cell of origin and the genetic driver lesions play a critical role in shaping the phenotypes of lung tumors. However, they also illustrate that there is far from a direct one-to-one relationship between the cell of origin and the cancer subtype, as most epithelial cells can be reprogrammed toward diverse lung cancer fates when exposed to the appropriate set of driver mutations.
Collapse
Affiliation(s)
- Giustina Ferone
- Division of Molecular Genetics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Myung Chang Lee
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Julien Sage
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Anton Berns
- Division of Molecular Genetics, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
19
|
Bürtin F, Mullins CS, Linnebacher M. Mouse models of colorectal cancer: Past, present and future perspectives. World J Gastroenterol 2020; 26:1394-1426. [PMID: 32308343 PMCID: PMC7152519 DOI: 10.3748/wjg.v26.i13.1394] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common diagnosed malignancy among both sexes in the United States as well as in the European Union. While the incidence and mortality rates in western, high developed countries are declining, reflecting the success of screening programs and improved treatment regimen, a rise of the overall global CRC burden can be observed due to lifestyle changes paralleling an increasing human development index. Despite a growing insight into the biology of CRC and many therapeutic improvements in the recent decades, preclinical in vivo models are still indispensable for the development of new treatment approaches. Since the development of carcinogen-induced rodent models for CRC more than 80 years ago, a plethora of animal models has been established to study colon cancer biology. Despite tenuous invasiveness and metastatic behavior, these models are useful for chemoprevention studies and to evaluate colitis-related carcinogenesis. Genetically engineered mouse models (GEMM) mirror the pathogenesis of sporadic as well as inherited CRC depending on the specific molecular pathways activated or inhibited. Although the vast majority of CRC GEMM lack invasiveness, metastasis and tumor heterogeneity, they still have proven useful for examination of the tumor microenvironment as well as systemic immune responses; thus, supporting development of new therapeutic avenues. Induction of metastatic disease by orthotopic injection of CRC cell lines is possible, but the so generated models lack genetic diversity and the number of suited cell lines is very limited. Patient-derived xenografts, in contrast, maintain the pathological and molecular characteristics of the individual patient’s CRC after subcutaneous implantation into immunodeficient mice and are therefore most reliable for preclinical drug development – even in comparison to GEMM or cell line-based analyses. However, subcutaneous patient-derived xenograft models are less suitable for studying most aspects of the tumor microenvironment and anti-tumoral immune responses. The authors review the distinct mouse models of CRC with an emphasis on their clinical relevance and shed light on the latest developments in the field of preclinical CRC models.
Collapse
Affiliation(s)
- Florian Bürtin
- Department of General, Visceral, Vascular and Transplantation Surgery, University Medical Center Rostock, University of Rostock, Rostock 18057, Germany
| | - Christina S Mullins
- Department of Thoracic Surgery, University Medical Center Rostock, University of Rostock, Rostock 18057, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Department of General, Visceral, Vascular and Transplantation Surgery, University Medical Center Rostock, Rostock 18057, Germany
| |
Collapse
|
20
|
Wiel C, Le Gal K, Ibrahim MX, Jahangir CA, Kashif M, Yao H, Ziegler DV, Xu X, Ghosh T, Mondal T, Kanduri C, Lindahl P, Sayin VI, Bergo MO. BACH1 Stabilization by Antioxidants Stimulates Lung Cancer Metastasis. Cell 2019; 178:330-345.e22. [PMID: 31257027 DOI: 10.1016/j.cell.2019.06.005] [Citation(s) in RCA: 335] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 03/29/2019] [Accepted: 06/03/2019] [Indexed: 01/03/2023]
Abstract
For tumors to progress efficiently, cancer cells must overcome barriers of oxidative stress. Although dietary antioxidant supplementation or activation of endogenous antioxidants by NRF2 reduces oxidative stress and promotes early lung tumor progression, little is known about its effect on lung cancer metastasis. Here, we show that long-term supplementation with the antioxidants N-acetylcysteine and vitamin E promotes KRAS-driven lung cancer metastasis. The antioxidants stimulate metastasis by reducing levels of free heme and stabilizing the transcription factor BACH1. BACH1 activates transcription of Hexokinase 2 and Gapdh and increases glucose uptake, glycolysis rates, and lactate secretion, thereby stimulating glycolysis-dependent metastasis of mouse and human lung cancer cells. Targeting BACH1 normalized glycolysis and prevented antioxidant-induced metastasis, while increasing endogenous BACH1 expression stimulated glycolysis and promoted metastasis, also in the absence of antioxidants. We conclude that BACH1 stimulates glycolysis-dependent lung cancer metastasis and that BACH1 is activated under conditions of reduced oxidative stress.
Collapse
Affiliation(s)
- Clotilde Wiel
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden; Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Kristell Le Gal
- Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Mohamed X Ibrahim
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | | | - Muhammad Kashif
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Haidong Yao
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Dorian V Ziegler
- Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Xiufeng Xu
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden
| | - Tanushree Ghosh
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Tanmoy Mondal
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Per Lindahl
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Volkan I Sayin
- Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, 405 30 Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden.
| | - Martin O Bergo
- Department of Biosciences and Nutrition, Karolinska Institutet, 141 83 Huddinge, Sweden; Sahlgrenska Cancer Center, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 405 30 Gothenburg, Sweden.
| |
Collapse
|
21
|
A step towards valid detection and quantification of lung cancer volume in experimental mice with contrast agent-based X-ray microtomography. Sci Rep 2019; 9:1325. [PMID: 30718557 PMCID: PMC6362109 DOI: 10.1038/s41598-018-37394-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 11/30/2018] [Indexed: 12/18/2022] Open
Abstract
Tumor volume is a parameter used to evaluate the performance of new therapies in lung cancer research. Conventional methods that are used to estimate tumor size in mouse models fail to provide fast and reliable volumetric data for tumors grown non-subcutaneously. Here, we evaluated the use of iodine-staining combined with micro-computed tomography (micro-CT) to estimate the tumor volume of ex vivo tumor-burdened lungs. We obtained fast high spatial resolution three-dimensional information of the lungs, and we demonstrated that iodine-staining highlights tumors and unhealthy tissue. We processed iodine-stained lungs for histopathological analysis with routine hematoxylin and eosin (H&E) staining. We compared the traditional tumor burden estimation performed manually with H&E histological slices with a semi-automated method using micro-CT datasets. In mouse models that develop lung tumors with well precise boundaries, the method that we describe here enables to perform a quick estimation of tumorous tissue volume in micro-CT images. Our method overestimates the tumor burden in tumors surrounded by abnormal tissue, while traditional histopathological analysis underestimates tumor volume. We propose to embed micro-CT imaging to the traditional workflow of tumorous lung analyses in preclinical cancer research as a strategy to obtain a more accurate estimation of the total lung tumor burden.
Collapse
|
22
|
Akbay EA, Kim J. Autochthonous murine models for the study of smoker and never-smoker associated lung cancers. Transl Lung Cancer Res 2018; 7:464-486. [PMID: 30225211 DOI: 10.21037/tlcr.2018.06.04] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lung cancer accounts for the greatest number of cancer deaths in the world. Tobacco smoke-associated cancers constitute the majority of lung cancer cases but never-smoker cancers comprise a significant and increasing fraction of cases. Recent genomic and transcriptomic sequencing efforts of lung cancers have revealed distinct sets of genetic aberrations of smoker and never-smoker lung cancers that implicate disparate biology and therapeutic strategies. Autochthonous mouse models have contributed greatly to our understanding of lung cancer biology and identified novel therapeutic targets and strategies in the era of targeted therapy. With the emergence of immuno-oncology, mouse models may continue to serve as valuable platforms for novel biological insights and therapeutic strategies. Here, we will review the variety of available autochthonous mouse models of lung cancer, their relation to human smoker and never-smoker lung cancers, and their application to immuno-oncology and immune checkpoint blockade that is revolutionizing lung cancer therapy.
Collapse
Affiliation(s)
- Esra A Akbay
- Department of Pathology, University of Texas Southwestern, Dallas, TX 75208, USA.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern, Dallas, TX 75208, USA
| | - James Kim
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern, Dallas, TX 75208, USA.,Department of Internal Medicine, Division of Hematology-Oncology, University of Texas Southwestern, Dallas, TX 75208, USA
| |
Collapse
|
23
|
Borrego A, Cabrera WHK, Jensen JR, Correa M, Ribeiro OG, Starobinas N, De Franco M, Pettinicchio A, Dragani TA, Ibañez OCM, Manenti G. Germline control of somatic Kras mutations in mouse lung tumors. Mol Carcinog 2018; 57:745-751. [PMID: 29500885 DOI: 10.1002/mc.22796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 02/07/2018] [Accepted: 02/28/2018] [Indexed: 01/07/2023]
Abstract
Somatic KRAS mutations are common in human lung adenocarcinomas and are associated with worse prognosis. In mice, Kras is frequently mutated in both spontaneous and experimentally induced lung tumors, although the pattern of mutation varies among strains, suggesting that such mutations are not random events. We tested if the occurrence of Kras mutations is under genetic control in two mouse intercrosses. Codon 61 mutations were prevalent, but the patterns of nucleotide changes differed between the intercrosses. Whole genome analysis with SNPs in (A/J x C57BL/6)F4 mice revealed a significant linkage between a locus on chromosome 19 and 2 particular codon 61 variants (CTA and CGA). In (AIRmax × AIRmin) F2 mice, there was a significant linkage between SNPs located on distal chromosome 6 (around 135 Mbp) and the frequency of codon 61 mutation. These results reveal the presence of two loci, on chromosomes 6 and 19, that modulate Kras mutation frequency in different mouse intercrosses. These findings indicate that somatic mutation frequency and type are not simple random events, but are under genetic control.
Collapse
Affiliation(s)
- Andrea Borrego
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | - Wafa H K Cabrera
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | - José R Jensen
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | - Mara Correa
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | | | - Nancy Starobinas
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | | | - Angela Pettinicchio
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Tommaso A Dragani
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Olga C M Ibañez
- Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | - Giacomo Manenti
- Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
24
|
Minas TZ, Surdez D, Javaheri T, Tanaka M, Howarth M, Kang HJ, Han J, Han ZY, Sax B, Kream BE, Hong SH, Çelik H, Tirode F, Tuckermann J, Toretsky JA, Kenner L, Kovar H, Lee S, Sweet-Cordero EA, Nakamura T, Moriggl R, Delattre O, Üren A. Combined experience of six independent laboratories attempting to create an Ewing sarcoma mouse model. Oncotarget 2018; 8:34141-34163. [PMID: 27191748 PMCID: PMC5470957 DOI: 10.18632/oncotarget.9388] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 05/05/2016] [Indexed: 12/17/2022] Open
Abstract
Ewing sarcoma (ES) involves a tumor-specific chromosomal translocation that produces the EWS-FLI1 protein, which is required for the growth of ES cells both in vitro and in vivo. However, an EWS-FLI1-driven transgenic mouse model is not currently available. Here, we present data from six independent laboratories seeking an alternative approach to express EWS-FLI1 in different murine tissues. We used the Runx2, Col1a2.3, Col1a3.6, Prx1, CAG, Nse, NEFL, Dermo1, P0, Sox9 and Osterix promoters to target EWS-FLI1 or Cre expression. Additional approaches included the induction of an endogenous chromosomal translocation, in utero knock-in, and the injection of Cre-expressing adenovirus to induce EWS-FLI1 expression locally in multiple lineages. Most models resulted in embryonic lethality or developmental defects. EWS-FLI1-induced apoptosis, promoter leakiness, the lack of potential cofactors, and the difficulty of expressing EWS-FLI1 in specific sites were considered the primary reasons for the failed attempts to create a transgenic mouse model of ES.
Collapse
Affiliation(s)
- Tsion Zewdu Minas
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Didier Surdez
- Genetics and Biology of Cancers Unit, Institut Curie Research Center, PSL Research University, Île-de-France, Paris, France.,INSERM U830, Institut Curie Research Center, Île-de-France, Paris, France
| | | | - Miwa Tanaka
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Michelle Howarth
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Hong-Jun Kang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - Jenny Han
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Zhi-Yan Han
- Genetics and Biology of Cancers Unit, Institut Curie Research Center, PSL Research University, Île-de-France, Paris, France.,INSERM U830, Institut Curie Research Center, Île-de-France, Paris, France
| | - Barbara Sax
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Barbara E Kream
- Department of Medicine, and Genetics and Genome Sciences, University of Connecticut Health Science Center, Farmington, CT, United States of America
| | - Sung-Hyeok Hong
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Haydar Çelik
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Franck Tirode
- Genetics and Biology of Cancers Unit, Institut Curie Research Center, PSL Research University, Île-de-France, Paris, France.,INSERM U830, Institut Curie Research Center, Île-de-France, Paris, France
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Jeffrey A Toretsky
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Lukas Kenner
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria.,Department of Pathology of Laboratory Animals (UPLA), University of Veterinary Medicine, Vienna, Austria
| | - Heinrich Kovar
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria.,Children´s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Sean Lee
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States of America
| | - E Alejandro Sweet-Cordero
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Takuro Nakamura
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria.,Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria.,Medical University of Vienna, Vienna, Austria
| | - Olivier Delattre
- Genetics and Biology of Cancers Unit, Institut Curie Research Center, PSL Research University, Île-de-France, Paris, France.,INSERM U830, Institut Curie Research Center, Île-de-France, Paris, France.,Unité de génétique somatique, Institut Curie, Île-de-France, Paris, France
| | - Aykut Üren
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
25
|
Yang M, Feng Y, Yue C, Xu B, Chen L, Jiang J, Lu B, Zhu Y. Lower expression level of IL-33 is associated with poor prognosis of pulmonary adenocarcinoma. PLoS One 2018; 13:e0193428. [PMID: 29499051 PMCID: PMC5834175 DOI: 10.1371/journal.pone.0193428] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/09/2018] [Indexed: 12/17/2022] Open
Abstract
Objective Lung cancer is one of the deadliest malignancies. The immune checkpoint-blockade (ICB) tumor therapy has led to striking improvement of long-term survival for some lung cancer patients. However, the response rate of immunotherapy is still low for lung cancer. Studying the tumor microenvironment (TME) should shed light on improvement of immunotherapy of lung cancer. Interleukin-33 (IL-33), an “alarmin” cytokine, has been implicated in tumor associated immune responses and inflammatory diseases of the lung. The role of IL-33 in lung cancer progression, however, remains elusive. This study is designed to characterize IL-33 expression in lung tumor tissues and establish the clinical significance of IL-33 in non-small cell lung cancer lung cancer (NSCLC). Materials and methods Tumor tissue specimens from patients suffering from NSCLC were analyzed for expression of IL-33 protein by immunohistochemistry and expression of IL-33 and ST2 mRNA by RT-quantitative PCR (RT-QPCR). The expression data were analyzed for their association with clinical and pathological parameters of NSCLC. In addition, the association between expression levels of IL-33 mRNA and patient survival was determined using 5 independent expression profiling datasets of human lung cancer. Results and conclusion The expression levels of IL-33 and ST2 were significantly down-regulated in both adenocarcinoma and squamous cell carcinoma of the lung when compared to adjacent normal lung tissues. In addition, the level of IL-33 protein was inversely correlated with tumor grade and size. Moreover, analysis of TCGA and GEO lung cancer expression datasets revealed that higher expression levels of IL-33 mRNA were correlated with longer overall survival of patients suffering from adenocarcinoma of the lung. These data indicate that the expression levels of IL-33 are inversely associated with lung cancer progression, consistent with the hypothesis that IL-33 is involved in immune surveillance of NSCLC.
Collapse
Affiliation(s)
- Min Yang
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yuehua Feng
- Comprehensive Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Cuihua Yue
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, Soochow University, Suzhou, China
| | - Binfeng Lu
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (BL); (YZ)
| | - Yibei Zhu
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
- Institute of Cell Therapy, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
- * E-mail: (BL); (YZ)
| |
Collapse
|
26
|
Dassano A, Pintarelli G, Cotroneo CE, Pettinicchio A, Forcati E, De Cecco L, Borrego A, Colombo F, Dragani TA, Manenti G. Complex genetic control of lung tumorigenesis in resistant mice strains. Cancer Sci 2017; 108:2281-2286. [PMID: 28796413 PMCID: PMC5666032 DOI: 10.1111/cas.13349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/27/2017] [Accepted: 08/05/2017] [Indexed: 12/18/2022] Open
Abstract
The SM/J mouse strain is resistant to chemically‐induced lung tumorigenesis despite having a haplotype, in the pulmonary adenoma susceptibility locus (Pas1) locus, that confers tumor susceptibility in other strains. To clarify this inconsistent genotype‐phenotype correlation, we crossed SM/J mice with another resistant strain and conducted genome‐wide linkage analysis in the (C57BL/6J × SM/J)F2 progeny exposed to urethane to induce lung tumors. Overall, >80% of F2 mice of both sexes developed from 1 to 20 lung tumors. Genotyping of 372 F2 mice for 744 informative non‐redundant SNPs dispersed over all autosomal chromosomes revealed four quantitative trait loci (QTLs) affecting lung tumor multiplicity, on chromosomes 3 (near rs13477379), 15 (rs6285067), 17 (rs33373629) and 18 (rs3706601), all with logarithm of the odds (LOD) scores >5. Four QTLs modulated total lung tumor volume, on chromosome 3 (rs13477379), 10 (rs13480702), 15 (rs6285067) and 17 (rs3682923), all with LOD scores >4. No QTL modulating lung tumor multiplicity or total volume was detected in Pas1 on chromosome 6. The present study demonstrates that the SM/J strain carries, at the Pas1 locus, the resistance allele: a finding that will facilitate identification of the Pas1 causal element. More generally, it demonstrates that lung tumorigenesis is under complex polygenic control even in a pedigree with low susceptibility to this neoplasia, suggesting that the genetics of lung tumorigenesis is much more complex than evidenced by the pulmonary adenoma susceptibility and resistance loci that have, so far, been mapped in a small number of crosses between a few inbred strains.
Collapse
Affiliation(s)
- Alice Dassano
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Pintarelli
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara E Cotroneo
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Angela Pettinicchio
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Elena Forcati
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Loris De Cecco
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy.,Department of Experimental Oncology and Molecular medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Andrea Borrego
- Laboratory of Immunogenetics, Instituto Butantan, São Paulo, Brazil
| | - Francesca Colombo
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Tommaso A Dragani
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| | - Giacomo Manenti
- Department of Predictive and Preventive Medicine, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
27
|
Annunziato S, Kas SM, Nethe M, Yücel H, Del Bravo J, Pritchard C, Bin Ali R, van Gerwen B, Siteur B, Drenth AP, Schut E, van de Ven M, Boelens MC, Klarenbeek S, Huijbers IJ, van Miltenburg MH, Jonkers J. Modeling invasive lobular breast carcinoma by CRISPR/Cas9-mediated somatic genome editing of the mammary gland. Genes Dev 2017; 30:1470-80. [PMID: 27340177 PMCID: PMC4926868 DOI: 10.1101/gad.279190.116] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/27/2016] [Indexed: 11/25/2022]
Abstract
Annunziato et al. describe a novel strategy for in vivo validation of candidate tumor suppressors implicated in invasive lobular breast carcinoma (ILC). Whereas intraductal injection of Cas9-encoding lentiviruses induced Cas9-specific immune responses and development of tumors that did not resemble ILC, lentiviral delivery of a Pten targeting sgRNA in mice with mammary gland-specific loss of E-cadherin and expression of Cas9 efficiently induced ILC development. Large-scale sequencing studies are rapidly identifying putative oncogenic mutations in human tumors. However, discrimination between passenger and driver events in tumorigenesis remains challenging and requires in vivo validation studies in reliable animal models of human cancer. In this study, we describe a novel strategy for in vivo validation of candidate tumor suppressors implicated in invasive lobular breast carcinoma (ILC), which is hallmarked by loss of the cell–cell adhesion molecule E-cadherin. We describe an approach to model ILC by intraductal injection of lentiviral vectors encoding Cre recombinase, the CRISPR/Cas9 system, or both in female mice carrying conditional alleles of the Cdh1 gene, encoding for E-cadherin. Using this approach, we were able to target ILC-initiating cells and induce specific gene disruption of Pten by CRISPR/Cas9-mediated somatic gene editing. Whereas intraductal injection of Cas9-encoding lentiviruses induced Cas9-specific immune responses and development of tumors that did not resemble ILC, lentiviral delivery of a Pten targeting single-guide RNA (sgRNA) in mice with mammary gland-specific loss of E-cadherin and expression of Cas9 efficiently induced ILC development. This versatile platform can be used for rapid in vivo testing of putative tumor suppressor genes implicated in ILC, providing new opportunities for modeling invasive lobular breast carcinoma in mice.
Collapse
Affiliation(s)
- Stefano Annunziato
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Sjors M Kas
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Micha Nethe
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Hatice Yücel
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Jessica Del Bravo
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Colin Pritchard
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Rahmen Bin Ali
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Bas van Gerwen
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Bjørn Siteur
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Anne Paulien Drenth
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Eva Schut
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Marieke van de Ven
- Preclinical Intervention Unit, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Mirjam C Boelens
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Sjoerd Klarenbeek
- Experimental Animal Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Ivo J Huijbers
- Transgenic Core Facility, Mouse Clinic for Cancer and Aging (MCCA), The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Martine H van Miltenburg
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; Cancer Genomics Netherlands, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
28
|
Bao L, Al-Assar O, Drynan LF, Arends MJ, Tyers P, Barker RA, Rabbitts TH. A non-cell autonomous mouse model of CNS haemangioblastoma mediated by mutant KRAS. Sci Rep 2017; 7:44899. [PMID: 28322325 PMCID: PMC5359595 DOI: 10.1038/srep44899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 02/14/2017] [Indexed: 01/01/2023] Open
Abstract
Haemangioblastoma is a rare malignancy of the CNS where vascular proliferation causes lesions due to endothelial propagation. We found that conditionally expressing mutant Kras, using Rag1-Cre, gave rise to CNS haemangioblastoma in the cortex and cerebellum in mice that present with highly vascular tumours with stromal cells similar to human haemangioblastomas. The aberrant haemangioblastoma endothelial cells do not express mutant Kras but rather the mutant oncogene is expressed in CNS interstitial cells, including neuronal cells and progeny. This demonstrates a non-cell autonomous origin of this disease that is unexpectedly induced via Rag1-Cre expression in CNS interstitial cells. This is the first time that mutant RAS has been shown to stimulate non-cell autonomous proliferation in malignancy and suggests that mutant RAS can control endothelial cell proliferation in neo-vascularisation when expressed in certain cells.
Collapse
Affiliation(s)
- Leyuan Bao
- Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| | - Osama Al-Assar
- Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| | - Lesley F Drynan
- MRC Laboratory of Molecular Biology, Francis Crick Ave, Cambridge CB2 OQH, UK
| | - Mark J Arends
- Centre for Comparative Pathology &Division of Pathology, University of Edinburgh, Institute of Genetics &Molecular Medicine, Crewe Road, Edinburgh, EH4 2XR, UK
| | - Pam Tyers
- John van Geest Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Terence H Rabbitts
- Weatherall Institute of Molecular Medicine, MRC Molecular Haematology Unit, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|
29
|
Antontseva EV, Matveeva MY, Bondar NP, Kashina EV, Leberfarb EY, Bryzgalov LO, Gervas PA, Ponomareva AA, Cherdyntseva NV, Orlov YL, Merkulova TI. Regulatory single nucleotide polymorphisms at the beginning of intron 2 of the human KRAS gene. J Biosci 2016; 40:873-83. [PMID: 26648033 DOI: 10.1007/s12038-015-9567-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
There are two regulatory single nucleotide polymorphisms (rSNPs) at the beginning of the second intron of the mouse K-ras gene that are strongly associated with lung cancer susceptibility. We performed functional analysis of three SNPs (rs12228277: T greater than A, rs12226937: G greater than A, and rs61761074: T greater than G) located in the same region of human KRAS. We found that rs12228277 and rs61761074 result in differential binding patterns of lung nuclear proteins to oligonucleotide probes corresponding two alternative alleles; in both cases, the transcription factor NF-Y is involved. G greater than A substitution (rs12226937) had no effect on the binding of lung nuclear proteins. However, all the nucleotide substitutions under study showed functional effects in a luciferase reporter assay. Among them, rs61761074 demonstrated a significant correlation with allele frequency in non-small-cell lung cancer (NSCLC). Taken together, the results of our study suggest that a T greater than G substitution at nucleotide position 615 in the second intron of the KRAS gene (rs61761074) may represent a promising genetic marker of NSCLC.
Collapse
Affiliation(s)
- Elena V Antontseva
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, Lavrentieva avenue 10, Novosibirsk 630090, Russian Federation
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Malanga D, Belmonte S, Colelli F, Scarfò M, De Marco C, Oliveira DM, Mirante T, Camastra C, Gagliardi M, Rizzuto A, Mignogna C, Paciello O, Papparella S, Fagman H, Viglietto G. AKT1E¹⁷K Is Oncogenic in Mouse Lung and Cooperates with Chemical Carcinogens in Inducing Lung Cancer. PLoS One 2016; 11:e0147334. [PMID: 26859676 PMCID: PMC4747507 DOI: 10.1371/journal.pone.0147334] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 01/01/2016] [Indexed: 11/19/2022] Open
Abstract
The hotspot AKT1E17K mutation in the pleckstrin homology domain of AKT1 occurs in approximately 0.6-2% of human lung cancers. Recently, we have demonstrated that AKT1E17K transforms immortalized human bronchial cells. Here by use of a transgenic Cre-inducible murine strain in the wild type Rosa26 (R26) locus (R26-AKT1E17K mice) we demonstrate that AKT1E17K is a bona-fide oncogene and plays a role in the development of lung cancer in vivo. In fact, we report that mutant AKT1E17K induces bronchial and/or bronchiolar hyperplastic lesions in murine lung epithelium, which progress to frank carcinoma at very low frequency, and accelerates tumor formation induced by chemical carcinogens. In conclusion, AKT1E17K induces hyperplasia of mouse lung epithelium in vivo and cooperates with urethane to induce the fully malignant phenotype.
Collapse
Affiliation(s)
- Donatella Malanga
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy
- BIOGEM-Istituto di Ricerche Genetiche, Ariano Irpino (AV), Italy
- * E-mail: (GV); (DM)
| | | | - Fabiana Colelli
- BIOGEM-Istituto di Ricerche Genetiche, Ariano Irpino (AV), Italy
| | - Marzia Scarfò
- BIOGEM-Istituto di Ricerche Genetiche, Ariano Irpino (AV), Italy
| | - Carmela De Marco
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy
- BIOGEM-Istituto di Ricerche Genetiche, Ariano Irpino (AV), Italy
| | - Duarte Mendes Oliveira
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy
| | - Teresa Mirante
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy
| | - Caterina Camastra
- Dipartimento di Scienze della Salute, Unità di Anatomia Patologica, Università Magna Graecia, Catanzaro, Italy
| | | | - Antonia Rizzuto
- Dipartimento di Scienze Mediche e Chirurgiche, Università Magna Graecia, Catanzaro, Italy
| | - Chiara Mignogna
- Dipartimento di Scienze della Salute, Unità di Anatomia Patologica, Università Magna Graecia, Catanzaro, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Productions, Università Federico II, Napoli, Italy
| | - Serenella Papparella
- Department of Veterinary Medicine and Animal Productions, Università Federico II, Napoli, Italy
| | - Henrik Fagman
- Department of Clinical Pathology and Genetics, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Giuseppe Viglietto
- Dipartimento di Medicina Sperimentale e Clinica, Università Magna Graecia, Catanzaro, Italy
- BIOGEM-Istituto di Ricerche Genetiche, Ariano Irpino (AV), Italy
- * E-mail: (GV); (DM)
| |
Collapse
|
31
|
Fang B. RAS signaling and anti-RAS therapy: lessons learned from genetically engineered mouse models, human cancer cells, and patient-related studies. Acta Biochim Biophys Sin (Shanghai) 2016; 48:27-38. [PMID: 26350096 DOI: 10.1093/abbs/gmv090] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022] Open
Abstract
Activating mutations of oncogenic RAS genes are frequently detected in human cancers. The studies in genetically engineered mouse models (GEMMs) reveal that Kras-activating mutations predispose mice to early onset tumors in the lung, pancreas, and gastrointestinal tract. Nevertheless, most of these tumors do not have metastatic phenotypes. Metastasis occurs when tumors acquire additional genetic changes in other cancer driver genes. Studies on clinical specimens also demonstrated that KRAS mutations are present in premalignant tissues and that most of KRAS mutant human cancers have co-mutations in other cancer driver genes, including TP53, STK11, CDKN2A, and KMT2C in lung cancer; APC, TP53, and PIK3CA in colon cancer; and TP53, CDKN2A, SMAD4, and MED12 in pancreatic cancer. Extensive efforts have been devoted to develop therapeutic agents that target enzymes involved in RAS posttranslational modifications, that inhibit downstream effectors of RAS signaling pathways, and that kill RAS mutant cancer cells through synthetic lethality. Recent clinical studies have revealed that sorafenib, a pan-RAF and VEGFR inhibitor, has impressive benefits for KRAS mutant lung cancer patients. Combination therapy of MEK inhibitors with either docetaxel, AKT inhibitors, or PI3K inhibitors also led to improved clinical responses in some KRAS mutant cancer patients. This review discusses knowledge gained from GEMMs, human cancer cells, and patient-related studies on RAS-mediated tumorigenesis and anti-RAS therapy. Emerging evidence demonstrates that RAS mutant cancers are heterogeneous because of the presence of different mutant alleles and/or co-mutations in other cancer driver genes. Effective subclassifications of RAS mutant cancers may be necessary to improve patients' outcomes through personalized precision medicine.
Collapse
Affiliation(s)
- Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
32
|
Wu K, Zhang X, Li F, Xiao D, Hou Y, Zhu S, Liu D, Ye X, Ye M, Yang J, Shao L, Pan H, Lu N, Yu Y, Liu L, Li J, Huang L, Tang H, Deng Q, Zheng Y, Peng L, Liu G, Gu X, He P, Gu Y, Lin W, He H, Xie G, Liang H, An N, Wang H, Teixeira M, Vieira J, Liang W, Zhao X, Peng Z, Mu F, Zhang X, Xu X, Yang H, Kristiansen K, Wang J, Zhong N, Wang J, Pan-Hammarström Q, He J. Frequent alterations in cytoskeleton remodelling genes in primary and metastatic lung adenocarcinomas. Nat Commun 2015; 6:10131. [PMID: 26647728 PMCID: PMC4682110 DOI: 10.1038/ncomms10131] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/06/2015] [Indexed: 01/04/2023] Open
Abstract
The landscape of genetic alterations in lung adenocarcinoma derived from Asian patients is largely uncharacterized. Here we present an integrated genomic and transcriptomic analysis of 335 primary lung adenocarcinomas and 35 corresponding lymph node metastases from Chinese patients. Altogether 13 significantly mutated genes are identified, including the most commonly mutated gene TP53 and novel mutation targets such as RHPN2, GLI3 and MRC2. TP53 mutations are furthermore significantly enriched in tumours from patients harbouring metastases. Genes regulating cytoskeleton remodelling processes are also frequently altered, especially in metastatic samples, of which the high expression level of IQGAP3 is identified as a marker for poor prognosis. Our study represents the first large-scale sequencing effort on lung adenocarcinoma in Asian patients and provides a comprehensive mutational landscape for both primary and metastatic tumours. This may thus form a basis for personalized medical care and shed light on the molecular pathogenesis of metastatic lung adenocarcinoma. Despite lung adenocarcinoma having a high global mortality, the genetic mutations present in Asian patients are uncharacterized. Here the authors use genomic and transcriptomic analysis to identify thirteen significantly affected genes, including RHPN2, GLI3, MRC2, TP53 and IQGAP3.
Collapse
Affiliation(s)
- Kui Wu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China.,Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Xin Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Fuqiang Li
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Dakai Xiao
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yong Hou
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Shida Zhu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Dongbing Liu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Xiaofei Ye
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Laboratory of Medicine, Karolinska Institutet, Stockholm 14186, Sweden
| | - Mingzhi Ye
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Guangzhou Key Laboratory of Cancer Trans-Omics Research, BGI-Guangzhou, Guangzhou 510006, China
| | - Jie Yang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Libin Shao
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Hui Pan
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Na Lu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Yuan Yu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Liping Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jin Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Liyan Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China
| | - Hailing Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China
| | - Qiuhua Deng
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yue Zheng
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Lihua Peng
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Geng Liu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Xia Gu
- Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Ping He
- Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yingying Gu
- Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Weixuan Lin
- Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Huiming He
- Research Center for Translational Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Guoyun Xie
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Han Liang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Na An
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Hui Wang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Manuel Teixeira
- Genetics Department and Research Center, Portuguese Oncology Institute, Porto 4200-072, Portugal
| | - Joana Vieira
- Genetics Department and Research Center, Portuguese Oncology Institute, Porto 4200-072, Portugal
| | - Wenhua Liang
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Xin Zhao
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Zhiyu Peng
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Guangzhou Key Laboratory of Cancer Trans-Omics Research, BGI-Guangzhou, Guangzhou 510006, China
| | - Feng Mu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,BGI-Wuhan, Wuhan 430075, China
| | - Xiuqing Zhang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Guangzhou Key Laboratory of Cancer Trans-Omics Research, BGI-Guangzhou, Guangzhou 510006, China
| | - Xun Xu
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Huanming Yang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,James D. Watson Institute of Genome Sciences, Zhejiang University, Hangzhou 310058, China
| | - Karsten Kristiansen
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jian Wang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,James D. Watson Institute of Genome Sciences, Zhejiang University, Hangzhou 310058, China
| | - Nanshan Zhong
- Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| | - Jun Wang
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Qiang Pan-Hammarström
- BGI-Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China.,Department of Laboratory of Medicine, Karolinska Institutet, Stockholm 14186, Sweden
| | - Jianxing He
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China.,Guangzhou Institute of Respiratory Disease &State Key Laboratory of Respiratory Disease, Guangzhou 510120, China.,National Clinical Research Center for Respiratory Disease, Guangzhou 510120, China
| |
Collapse
|
33
|
Zhao ZG, Jin JY, Zhang AM, Zhang LP, Wang XX, Sun JG, Chen ZT. MicroRNA profile of tumorigenic cells during carcinogenesis of lung adenocarcinoma. J Cell Biochem 2015; 116:458-66. [PMID: 25359683 DOI: 10.1002/jcb.24999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 10/20/2014] [Indexed: 12/19/2022]
Abstract
To obtain microRNA (miRNA) profile and clarify their biological function in tumorigenic Sca-1(+) CD34(+) cells during carcinogenesis of lung adenocarcinoma. After intranasal infection with recombinant Adeno-Cre viruses (AdV-Cre), lung adenocarcinoma was identified pathologically in Lox-stop-lox Kras (LSL-Kras) G12D mice. Sca-1(+) CD34(+) cells were sorted by flow cytometry and tested for tumor-initiating ability, self-renewal and tumorigenicity. MiRNA profiles were obtained using microarray and further confirmed by real-time RT-PCR (qRT-PCR). MiRNA functions were predicted bioinformatically, and miR-294 function was verified to explore its role in tumor migration and invasion. Lung adenocarcinoma was induced in LSL-Kras G12D mice within 30 days. In vivo, the tumorigenicity of Sca-1(+) CD34(+) cells was 25 times stronger than Sca-1(-) CD34(-) cells. During tumorigenesis of lung adenocarcinoma, the expression of 145 miRNAs in Sca-1(+) CD34(+) cells increased and 72 miRNAs decreased (P < 0.01). Four successively up-regulated miRNAs (miR-15a*, miR-203, miR-294 and miR-295*) and three successively down-regulated ones (miR-19b, miR-483 and miR-615-5p) were identified. Among them, miR-294 could constitutively bind to 3'-UTR of matrix metalloproteinase 3 (MMP3), and down-regulate MMP3 protein expression. MiR-294 also significantly inhibited migration and invasion of Lewis lung cancer cells. MiRNAs are characteristically expressed in tumor-initiating Sca-1(+) CD34(+) cells of lung adenocarcinoma, and may play important roles during the carcinogenesis of lung adenocarcinoma.
Collapse
Affiliation(s)
- Zhen-guo Zhao
- Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | | | | | | | | | | | | |
Collapse
|
34
|
Fang B, Mehran RJ, Heymach JV, Swisher SG. Predictive biomarkers in precision medicine and drug development against lung cancer. CHINESE JOURNAL OF CANCER 2015; 34:295-309. [PMID: 26134262 PMCID: PMC4593363 DOI: 10.1186/s40880-015-0028-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/04/2015] [Indexed: 02/06/2023]
Abstract
The molecular characterization of various cancers has shown that cancers with the same origins, histopathologic diagnoses, and clinical stages can be highly heterogeneous in their genetic and epigenetic alterations that cause tumorigenesis. A number of cancer driver genes with functional abnormalities that trigger malignant transformation and that are required for the survival of cancer cells have been identified. Therapeutic agents targeting some of these cancer drivers have been successfully developed, resulting in substantial improvements in clinical symptom amelioration and outcomes in a subset of cancer patients. However, because such therapeutic drugs often benefit only a limited number of patients, the successes of clinical development and applications rely on the ability to identify those patients who are sensitive to the targeted therapies. Thus, biomarkers that can predict treatment responses are critical for the success of precision therapy for cancer patients and of anticancer drug development. This review discusses the molecular heterogeneity of lung cancer pathogenesis; predictive biomarkers for precision medicine in lung cancer therapy with drugs targeting epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 receptor tyrosine kinase (ROS1), and immune checkpoints; biomarkers associated with resistance to these therapeutics; and approaches to identify predictive biomarkers in anticancer drug development. The identification of predictive biomarkers during anticancer drug development is expected to greatly facilitate such development because it will increase the chance of success or reduce the attrition rate. Additionally, such identification will accelerate the drug approval process by providing effective patient stratification strategies in clinical trials to reduce the sample size required to demonstrate clinical benefits.
Collapse
Affiliation(s)
- Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Reza J Mehran
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - John V Heymach
- Department of Thoracic and Head/Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Preclinical Murine Models for Lung Cancer: Clinical Trial Applications. BIOMED RESEARCH INTERNATIONAL 2015; 2015:621324. [PMID: 26064932 PMCID: PMC4433653 DOI: 10.1155/2015/621324] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/24/2014] [Indexed: 12/18/2022]
Abstract
Murine models for the study of lung cancer have historically been the backbone of preliminary preclinical data to support early human clinical trials. However, the availability of multiple experimental systems leads to debate concerning which model, if any, is best suited for a particular therapeutic strategy. It is imperative that these models accurately predict clinical benefit of therapy. This review provides an overview of the current murine models used to study lung cancer and the advantages and limitations of each model, as well as a retrospective evaluation of the uses of each model with respect to accuracy in predicting clinical benefit of therapy. A better understanding of murine models and their uses, as well as their limitations may aid future research concerning the development and implementation of new targeted therapies and chemotherapeutic agents for lung cancer.
Collapse
|
36
|
Abstract
To date a variety of non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) mouse models have been developed that mimic human lung cancer. Chemically induced or spontaneous lung cancer in susceptible inbred strains has been widely used, but the more recent genetically engineered somatic mouse models recapitulate much better the genotype-phenotype correlations found in human lung cancer. Additionally, improved orthotopic transplantation of primary human cancer tissue fragments or cells into lungs of immune-compromised mice can be valuable tools for preclinical research such as antitumor drug tests. Here we give a short overview of most somatic mouse models for lung cancer that are currently in use. We accompany each different model with a description of its practical use and application for all major lung tumor types, as well as the intratracheal injection or direct injection of fresh or freeze-thawed tumor cells or tumor cell lines into lung parenchyma of recipient mice. All here presented somatic mouse models are based on the ability to (in) activate specific alleles at a time, and in a tissue-specific cell type, of choice. This spatial-temporal controlled induction of genetic lesions allows the selective introduction of main genetic lesions in an adult mouse lung as found in human lung cancer. The resulting conditional somatic mouse models can be used as versatile powerful tools in basic lung cancer research and preclinical translational studies alike. These distinctively advanced lung cancer models permit us to investigate initiation (cell of origin) and progression of lung cancer, along with response and resistance to drug therapy. Cre/lox or FLP/frt recombinase-mediated methods are now well-used techniques to develop tissue-restricted lung cancer in mice with tumor-suppressor gene and/or oncogene (in)activation. Intranasal or intratracheal administration of engineered adenovirus-Cre or lentivirus-Cre has been optimized for introducing Cre recombinase activity into pulmonary tissues, and we discuss here the different techniques underlying these applications. Concomitant with Cre/Flp recombinase-based models are the tetracycline (Tet)-inducible bitransgenic systems in which presence or absence of doxycycline can turn the expression of a specific oncogene on or off. The use of several Tet-inducible lung cancer models for NSCLC is presented here in which the reversal of oncogene expression led to complete tumor regression and provided us with important insight of how oncogene dependence influence lung cancer survival and growth. As alternative to Tet-inducible models, we discuss the application of reversible expressed, transgenic mutant estrogen receptor (ER) fusion proteins, which are regulated via systemic tamoxifen administration. Most of the various lung cancer models can be combined through the generation of transgenic compound mice so that the use of these somatic mouse models can be even more enhanced for the study of specific molecular pathways that facilitate growth and maintenance of lung cancer. Finally, this description of the practical application and methodology of mouse models for lung cancer should be helpful in assisting researchers to make the best choices and optimal use of (existing) somatic models that suits the specific experimental needs in their study of lung cancer.
Collapse
Affiliation(s)
- Roghaiyeh Safari
- Health Science Institute, Dokuz Eylul University, Cumhuriyet Bulvari No: 144 35210, Alsancak, Izmir, Turkey
| | | |
Collapse
|
37
|
Terp MG, Ditzel HJ. Application of proteomics in the study of rodent models of cancer. Proteomics Clin Appl 2014; 8:640-52. [DOI: 10.1002/prca.201300084] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/25/2013] [Accepted: 11/27/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Mikkel G. Terp
- Department of Cancer and Inflammation Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
| | - Henrik J. Ditzel
- Department of Cancer and Inflammation Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
- Department of Oncology; Odense University Hospital; Odense Denmark
| |
Collapse
|
38
|
Saito H, Suzuki N. K-rasG12V mediated lung tumor models identified three new quantitative trait loci modifying events post-K-ras mutation. Biochem Biophys Res Commun 2014; 452:1067-70. [PMID: 25245290 DOI: 10.1016/j.bbrc.2014.09.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 10/24/2022]
Abstract
A high incidence of oncogenic K-ras mutations is observed in lung adenocarcinoma of human cases and carcinogen-induced animal models. The process of oncogenic K-ras-mediated lung adenocarcinogenesis can be dissected into two parts: pre- and post-K-ras mutation. Adoption of transgenic lines containing a flox-K-rasG12V transgene eliminates the use of chemical carcinogens and enables us to study directly crucial events post-K-ras mutation without considering the cellular events involved with oncogenic K-ras mutation, e.g., distribution and metabolism of chemical carcinogens, DNA repair, and somatic recombination by host factors. We generated two mouse strains C57BL/6J-Ryr2(tm1Nobs) and A/J-Ryr2(tm1Nobs) in which K-rasG12V can be transcribed from the cytomegalovirus early enhancer/chicken beta actin promoter in virtually any tissue. Upon K-rasG12V induction in lung epithelial cells by an adenovirus expressing the Cre recombinase, the number of tumors in the C57BL/6J-Ryr2(tm1Nobs/+) mouse line was 12.5 times that in the A/J-Ryr2(tm1Nobs/+) mouse line. Quantitative trait locus (QTL) analysis revealed that new three modifier loci, D3Mit19, D3Mit45 and D11Mit20, were involved in the differential susceptibility between the two lines. In addition, we found that differential expression of the wild-type K-ras gene, which was genetically turn out to be anti-oncogenic activity on K-rasG12V, could not account for the different susceptibility in our two K-rasG12V-mediated lung tumor models. Thus, we provide a genetic system that enables us to explore new downstream modifiers post-K-ras mutation.
Collapse
Affiliation(s)
- Hiromitsu Saito
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Noboru Suzuki
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, 2-174 Edobashi, Tsu, Mie 514-8507, Japan.
| |
Collapse
|
39
|
Rappaport A, Johnson L. Genetically engineered knock-in and conditional knock-in mouse models of cancer. Cold Spring Harb Protoc 2014; 2014:897-911. [PMID: 25183823 DOI: 10.1101/pdb.top069799] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Classical transgenic models are useful for quickly gauging the impact of transgene overexpression, but they are limited by the absence of the innate, subtle regulatory elements encoded in introns and other untranslated regions. Moreover, the widespread, high-level expression of oncogenes often leads to tumors that lack the histopathological and acquired genetic features of human cancers. Targeted mutation of endogenous loci, or knock-in (KI) alleles, facilitates more accurate modeling of human tumors by allowing for the expression of mutant alleles under normal physiological regulation. Advanced strategies enable the stochastic activation of such alleles in somatic cells, such that genotypically wild-type cells surround individual mutant cells. More recent technologies, such as site-specific engineered nucleases, have also accelerated the design and implementation of KI strategies. Together, these tools aid in the development of advanced mouse models that better recapitulate the features of human disease.
Collapse
Affiliation(s)
- Amy Rappaport
- Genentech, Inc., South San Francisco, California 94080
| | - Leisa Johnson
- Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
40
|
Lineage tracing of mammary epithelial cells using cell-type-specific cre-expressing adenoviruses. Stem Cell Reports 2014; 2:770-9. [PMID: 24936465 PMCID: PMC4050356 DOI: 10.1016/j.stemcr.2014.04.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 04/08/2014] [Accepted: 04/09/2014] [Indexed: 12/27/2022] Open
Abstract
Lineage tracing using Cre/lox transgenic mice provides a powerful tool for studying normal mammary epithelial cell (MEC) development and the cellular origins of mammary tumors under physiological settings. However, generation of new transgenic mice for lineage-tracing purposes is often time consuming. Here, we report a lineage-tracing tool for MECs based on intraductal injection of lineage-specific Cre-expressing adenovirus (Ad-Cre). Using well-characterized promoters for Keratin 8 and Keratin 14, we generated lineage-specific Ad-Cre lines for luminal and basal MECs, respectively. By pulse-chase lineage tracing using these Ad-Cre lines, we showed that luminal and basal lineages are largely self-sustained and that IRS1 and IRS2 are essential for maintaining the basal lineage; we also showed that heterogeneous mammary tumors can be induced from luminal MECs in mice carrying the Etv6-NTRK3 fusion gene. Overall, we validated the Ad-Cre system as a promising and efficient tool for fate mapping of normal and malignant cells in adult tissues. Adenovirus-Cre can be used for pulse-chase lineage tracing of adult stem cells Mammary luminal and basal lineages in adults are largely self-sustained IRS1 and IRS2 are essential for maintaining the adult mammary basal lineage Multiple adult mammary luminal cell types may serve as breast cancer cellular origins
Collapse
|
41
|
Mouse pulmonary adenoma susceptibility 1 locus is an expression QTL modulating Kras-4A. PLoS Genet 2014; 10:e1004307. [PMID: 24743582 PMCID: PMC3990522 DOI: 10.1371/journal.pgen.1004307] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 02/28/2014] [Indexed: 11/28/2022] Open
Abstract
Pulmonary adenoma susceptibility 1 (Pas1) is the major locus responsible for lung tumor susceptibility in mice; among the six genes mapping in this locus, Kras is considered the best candidate for Pas1 function although how it determines tumor susceptibility remains unknown. In an (A/J×C57BL/6)F4 intercross population treated with urethane to induce lung tumors, Pas1 not only modulated tumor susceptibility (LOD score = 48, 69% of phenotypic variance explained) but also acted, in lung tumor tissue, as an expression quantitative trait locus (QTL) for Kras-4A, one of two alternatively spliced Kras transcripts, but not Kras-4B. Additionally, Kras-4A showed differential allelic expression in lung tumor tissue of (A/J×C57BL/6)F4 heterozygous mice, with significantly higher expression from the A/J-derived allele; these results suggest that cis-acting elements control Kras-4A expression. In normal lung tissue from untreated mice of the same cross, Kras-4A levels were also highly linked to the Pas1 locus (LOD score = 23.2, 62% of phenotypic variance explained) and preferentially generated from the A/J-derived allele, indicating that Pas1 is an expression QTL in normal lung tissue as well. Overall, the present findings shed new light on the genetic mechanism by which Pas1 modulates the susceptibility to lung tumorigenesis, through the fine control of Kras isoform levels. A person's risk of developing cancer depends on both genetic and environmental factors. To study the genetic predisposition to cancer without the influence of environmental variables, scientists study mice treated with urethane, a chemical carcinogen that induces lung tumors. By crossing inbred (genetically identical) strains of mice that are either resistant or susceptible to urethane-induced cancer, researchers can search for genes associated with tumor formation in the offspring. From previous work of this type using second-generation mice, it was already known that a region on chromosome 6 was associated with tumor formation. Now, a new study, carried out in a fourth-generation mouse population, focused to a single gene of chromosome 6 called Kras. This gene forms two different messenger RNA transcripts, called Kras-4A and Kras-4B, that produce two proteins with slightly different structure and, perhaps, function. The study found that mice susceptible to lung tumors have relatively more Kras-4A messenger RNA than resistant mice and that this difference may be due to small variations in the DNA near or within this gene.
Collapse
|
42
|
Gong J, Wang Z, Polejaeva I, Salgia R, Kao CM, Chen CT, Chen G, Chen L. Activating the expression of human K-rasG12D stimulates oncogenic transformation in transgenic goat fetal fibroblast cells. PLoS One 2014; 9:e90059. [PMID: 24594684 PMCID: PMC3942380 DOI: 10.1371/journal.pone.0090059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/27/2014] [Indexed: 01/15/2023] Open
Abstract
Humane use of preclinical large animal cancer models plays a critical role in understanding cancer biology and developing therapeutic treatments. Among the large animal candidates, goats have great potentials as sustainable sources for large animal cancer model development. Goats are easier to handle and cheaper to raise. The genome of the goats has been sequenced recently. It has been known that goats develop skin, adrenal cortex, breast and other types of cancers. Technically, goats are subject to somatic cell nuclear transfer more efficiently and exhibit better viability through the cloning process. Towards the development of a goat cancer model, we created a transgenic goat fetal fibroblast (GFF) cell as the donor cell for SCNT. Human mutated K-ras (hK-rasG12D) was chosen as the transgene, as it is present in 20% of cancers. Both hK-rasG12D and a herpes simplex viral thymidine kinase (HSV1-tk) reporter genes, flanked by a pair of LoxP sites, were knocked in the GFF endogenous K-ras locus through homologous recombination. Following Cre-mediated activation (with a 95% activation efficiency), hK-rasG12D and HSV1-tk were expressed in the transgenic GFF cells, evidently through the presence of corresponding mRNAs, and confirmed by HSV1-tk protein function assay. The hK-rasG12D expressing GFF cells exhibited enhanced proliferation rates and an anchorage-independent growth behavior. They were able to initiate tumor growth in athymic nude mice. In conclusion, after activating hK-rasG12D gene expression, hK-rasG12D transgenic GFF cells were transformed into tumorgenesis cells. Transgenic goats via SCNT using the above-motioned cells as the donor cells have been established.
Collapse
Affiliation(s)
- Jianhua Gong
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah, United States of America
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Irina Polejaeva
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Ravi Salgia
- Department of Radiology, University of Chicago, Chicago, Illinois, United States of America
| | - Chien-Min Kao
- Department of Radiology, University of Chicago, Chicago, Illinois, United States of America
| | - Chin-Tu Chen
- Department of Radiology, University of Chicago, Chicago, Illinois, United States of America
| | - Guangchun Chen
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah, United States of America
| | - Liaohai Chen
- Department of Chemistry and Biochemistry, Utah State University, Logan, Utah, United States of America
| |
Collapse
|
43
|
Abstract
This issue marks the 50th anniversary of the release of the U.S. Surgeon General's Report on Smoking and Health. Perhaps no other singular event has done more to highlight the effects of smoking on the development of cancer. Tobacco exposure is the leading cause of cancers involving the oral cavity, conductive airways, and the lung. Owing to the many carcinogens in tobacco smoke, smoking-related malignancies have a high genome-wide burden of mutations, including in the gene encoding for p53. The p53 protein is the most frequently mutated tumor suppressor in cancer, responsible for a range of critical cellular functions that are compromised by the presence of a mutation. Herein, we review the epidemiologic connection between tobacco exposure and cancer, the molecular basis of p53 mutation in lung cancer, and the normal molecular and cellular roles of p53 that are abrogated during lung tumor development and progression as defined by in vitro and in vivo studies. We also consider the therapeutic potential of targeting mutant p53 in a clinical setting based upon the cellular role of mutant p53 and data from genetic murine models.
Collapse
Affiliation(s)
- Don L Gibbons
- University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Box 0432, Houston, Texas 77030.
| | | | | |
Collapse
|
44
|
Talluri S, Francis SM, Dick FA. Mutation of the LXCXE binding cleft of pRb facilitates transformation by ras in vitro but does not promote tumorigenesis in vivo. PLoS One 2013; 8:e72236. [PMID: 23936539 PMCID: PMC3735560 DOI: 10.1371/journal.pone.0072236] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 07/12/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The Retinoblastoma protein (pRB) is a key tumor suppressor that is functionally inactivated in most cancers. pRB regulates the cell division cycle and cell cycle exit through protein-protein interactions mediated by its multiple binding interfaces. The LXCXE binding cleft region of pRB mediates interactions with cellular proteins that have chromatin regulatory functions. Chromatin regulation mediated by pRB is required for a stress responsive cell cycle arrest, including oncogene induced senescence. The in vivo role of chromatin regulation by pRB during senescence, and its relevance to cancer is not clear. METHODOLOGY/PRINCIPAL FINDINGS Using gene-targeted mice, uniquely defective for pRB mediated chromatin regulation, we investigated its role during transformation and tumor progression in response to activation of oncogenic ras. We report that the pRB(∆L) mutation confers susceptibility to escape from HrasV12 induced senescence and allows transformation in vitro, although these cells possess high levels of DNA damage. Intriguingly, LSL-Kras, Rb1 (∆L/∆L) mice show delayed lung tumor formation compared to controls. This is likely due to the increased apoptosis seen in the early hyperplastic lesions shortly following ras activation that inhibits tumor progression. Furthermore, DMBA treatment to induce sporadic ras mutations in other tissues also failed to reveal greater susceptibility to cancer in Rb1 (∆L/∆L) mice. CONCLUSIONS/SIGNIFICANCE Our data suggests that chromatin regulation by pRB can function to limit proliferation, but its loss fails to contribute to cancer susceptibility in ras driven tumor models because of elevated levels of DNA damage and apoptosis.
Collapse
Affiliation(s)
- Srikanth Talluri
- London Regional Cancer Program, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| | - Sarah M. Francis
- London Regional Cancer Program, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| | - Frederick A. Dick
- London Regional Cancer Program, Western University, London, Ontario, Canada
- Children’s Health Research Institute, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
45
|
Asselin-Labat ML, Filby CE. Adult lung stem cells and their contribution to lung tumourigenesis. Open Biol 2013; 2:120094. [PMID: 22977734 PMCID: PMC3438537 DOI: 10.1098/rsob.120094] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 07/23/2012] [Indexed: 12/14/2022] Open
Abstract
The isolation and characterization of lung stem and progenitor cells represent an important step towards the understanding of lung repair after injury, lung disease pathogenesis and the identification of the target cells of transformation in lung carcinogenesis. Different approaches using prospective isolation of progenitor cells by flow cytometry or lineage-tracing experiments in mouse models of lung injury have led to the identification of distinct progenitor subpopulations in different morphological regions of the adult lung. Genetically defined mouse models of lung cancer are offering new perspectives on the cells of origin of different subtypes of lung cancer. These mouse models pave the way to further investigate human lung progenitor cells at the origin of lung cancers, as well as to define the nature of the lung cancer stem cells. It will be critical to establish the link between oncogenic driver mutations recently discovered in lung cancers, target cells of transformation and subtypes of lung cancers to enable better stratification of patients for improved therapeutic strategies.
Collapse
Affiliation(s)
- Marie-Liesse Asselin-Labat
- ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
| | | |
Collapse
|
46
|
König K, Meder L, Kröger C, Diehl L, Florin A, Rommerscheidt-Fuss U, Kahl P, Wardelmann E, Magin TM, Buettner R, Heukamp LC. Loss of the keratin cytoskeleton is not sufficient to induce epithelial mesenchymal transition in a novel KRAS driven sporadic lung cancer mouse model. PLoS One 2013; 8:e57996. [PMID: 23536778 PMCID: PMC3594220 DOI: 10.1371/journal.pone.0057996] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 01/30/2013] [Indexed: 01/30/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT), the phenotypical change of cells from an epithelial to a mesenchymal type, is thought to be a key event in invasion and metastasis of adenocarcinomas. These changes involve loss of keratin expression as well as loss of cell polarity and adhesion. We here aimed to determine whether the loss of keratin expression itself drives increased invasion and metastasis in adenocarcinomas and whether keratin loss leads to the phenotypic changes associated with EMT. Therefore, we employed a recently described murine model in which conditional deletion of the Keratin cluster II by Cre-recombinase leads to the loss of the entire keratinmultiprotein family. These mice were crossed into a newly generated Cre-recombinase inducible KRAS-driven murine lung cancer model to examine the effect of keratin loss on morphology, invasion and metastasis as well as expression of EMT related genes in the resulting tumors. We here clearly show that loss of a functional keratin cytoskeleton did not significantly alter tumor morphology or biology in terms of invasion, metastasis, proliferation or tumor burden and did not lead to induction of EMT. Further, tumor cells did not induce synchronously expression of vimentin, which is often seen in EMT, to compensate for keratin loss. In summary, our data suggest that changes in cell shape and migration that underlie EMT are dependent on changes in signaling pathways that cause secondary changes in keratin expression and organization. Thus, we conclude that loss of the keratin cytoskeleton per se is not sufficient to causally drive EMT in this tumor model.
Collapse
Affiliation(s)
- Katharina König
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Lydia Meder
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Cornelia Kröger
- Whitehead Institute of Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Linda Diehl
- Institutes of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn, Germany
| | | | | | - Philip Kahl
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Eva Wardelmann
- Institute of Pathology, University of Cologne, Cologne, Germany
| | - Thomas M. Magin
- Translational Centre for Regenerative Medicine and Institute of Biology, University of Leipzig, Leipzig, Germany
| | | | - Lukas C. Heukamp
- Institute of Pathology, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
47
|
Saxena M, Christofori G. Rebuilding cancer metastasis in the mouse. Mol Oncol 2013; 7:283-96. [PMID: 23474222 DOI: 10.1016/j.molonc.2013.02.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 02/06/2013] [Indexed: 12/17/2022] Open
Abstract
Most cancer deaths are due to the systemic dissemination of cancer cells and the formation of secondary tumors (metastasis) in distant organs. Recent years have brought impressive progress in metastasis research, yet we still lack sufficient insights into how cancer cells migrate out of primary tumors and invade into neighboring tissue, intravasate into the blood or the lymphatic circulation, survive in the blood stream, and target specific organs to initiate metastatic outgrowth. While a large number of cellular and animal models of cancer have been crucial in delineating the molecular mechanisms underlying tumor initiation and progression, experimental models that faithfully recapitulate the multiple stages of metastatic disease are still scarce. The advent of sophisticated genetic engineering in mice, in particular the ability to manipulate gene expression in specific tissue and at desired time points at will, have allowed to rebuild the metastatic process in mice. Here, we describe a selection of cellular experimental systems, tumor transplantation mouse models and genetically engineered mouse models that are used for monitoring specific processes involved in metastasis, such as cell migration and invasion, and for investigating the full metastatic process. Such models not only aid in deciphering the pathomechanisms of metastasis, but are also instrumental for the preclinical testing of anti-metastatic therapies and further refinement and generation of improved models.
Collapse
Affiliation(s)
- Meera Saxena
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | | |
Collapse
|
48
|
Kwon MC, Berns A. Mouse models for lung cancer. Mol Oncol 2013; 7:165-77. [PMID: 23481268 DOI: 10.1016/j.molonc.2013.02.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/06/2013] [Indexed: 01/24/2023] Open
Abstract
Lung cancer is a devastating disease and a major therapeutic burden with poor survival rates. It is responsible for 30% of all cancer deaths. Lung cancer is strongly associated with smoking, although some subtypes are also seen in non-smokers. Tumors in the latter group are mostly adenocarcinomas with many carrying mutations in the epidermal growth factor receptor (EGFR). Survival statistics of lung cancer are grim because of its late detection and frequent local and distal metastases. Although DNA sequence information from tumors has revealed a number of frequently occurring mutations, affecting well-known tumor suppressor genes and proto-oncogenes, many of the driver mutations remain ill defined. This is likely due to the involvement of numerous rather infrequently occurring driver mutations that are difficult to distinguish from the very large number of passenger mutations detected in smoking-related lung cancers. Therefore, experimental model systems are indispensable to validate putative driver lesions and to gain insight into their mechanisms of action. Whereas a large fraction of these analyzes can be performed in cell cultures in vitro, in many cases the consequences of the mutations have to be assessed in the context of an intact organism, as this is the context in which the Mendelian selection process of the tumorigenic process took place and the advantages of particular mutations become apparent. Current mouse models for cancer are very suitable for this as they permit mimicking many of the salient features of human tumors. The capacity to swiftly re-engineer complex sets of lesions found in human tumors in mice enables us to assess the contribution of defined combinations of lesions to distinct tumor characteristics such as metastatic behavior and response to therapy. In this review we will describe mouse models of lung cancer and how they are used to better understand the disease and how they are exploited to develop better intervention strategies.
Collapse
Affiliation(s)
- Min-chul Kwon
- Division of Molecular Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | |
Collapse
|
49
|
[The mouse as preclinical models of lung cancer]. Bull Cancer 2012; 99:1017-27. [PMID: 23131302 DOI: 10.1684/bdc.2012.1657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The high incidence and poor prognosis of lung cancer represent a major health problem. Currently, about 20% of lung cancer patients can benefit from targeted therapy after identification of EGFR, ALK or HER2 somatic mutations or rearrangements. Other mutations, such as KRas oncogenic mutation, are still orphans of validated targeted therapy. In this review, we describe the different mouse models of lung carcinoma. We then illustrate the interests of such models for the identification and validation of new therapeutic targets, for the study of secondary resistance and for their use as preclinical models and for new therapeutic strategy tests.
Collapse
|
50
|
Heukamp LC, Thor T, Schramm A, De Preter K, Kumps C, De Wilde B, Odersky A, Peifer M, Lindner S, Spruessel A, Pattyn F, Mestdagh P, Menten B, Kuhfittig-Kulle S, Künkele A, König K, Meder L, Chatterjee S, Ullrich RT, Schulte S, Vandesompele J, Speleman F, Büttner R, Eggert A, Schulte JH. Targeted expression of mutated ALK induces neuroblastoma in transgenic mice. Sci Transl Med 2012; 4:141ra91. [PMID: 22764207 DOI: 10.1126/scitranslmed.3003967] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Activating anaplastic lymphoma kinase (ALK) mutations were recently detected in most familial and 10% of sporadic neuroblastomas. However, the role of mutated ALK in tumorigenesis remains elusive. We demonstrate that targeted expression of the most frequent and aggressive variant, ALK(F1174L), is tumorigenic in mice. Tumors resembled human neuroblastomas in morphology, metastasis pattern, gene expression, and the presence of neurosecretory vesicles as well as synaptic structures. This ALK-driven neuroblastoma mouse model precisely recapitulated the genetic spectrum of the disease. Chromosomal aberrations were syntenic to those in human neuroblastoma, including 17q gain and MYCN oncogene amplification. Targeted ALK(F1174L) and MYCN coexpression revealed a strong synergism in inducing neuroblastoma with minimal chromosomal aberrations, suggesting that fewer secondary hits are required for tumor induction if both oncoproteins are targeted. Treatment of ALK(F1174L) transgenic mice with the ALK inhibitor TAE-684 induced complete tumor regression, indicating that tumor cells were addicted to ALK(F1174L) activity. We conclude that an activating mutation within the ALK kinase domain is sufficient to induce neuroblastoma development, and ALK inhibitors show promise for treating human neuroblastomas harboring ALK mutations.
Collapse
Affiliation(s)
- Lukas C Heukamp
- Institute of Pathology, University Hospital Cologne, Kerpenerstrasse 62, 50924 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|