Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Cell ; 161(7): 1539-1552, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26091037

ABSTRACT

The adenomatous polyposis coli (APC) tumor suppressor is mutated in the vast majority of human colorectal cancers (CRC) and leads to deregulated Wnt signaling. To determine whether Apc disruption is required for tumor maintenance, we developed a mouse model of CRC whereby Apc can be conditionally suppressed using a doxycycline-regulated shRNA. Apc suppression produces adenomas in both the small intestine and colon that, in the presence of Kras and p53 mutations, can progress to invasive carcinoma. In established tumors, Apc restoration drives rapid and widespread tumor-cell differentiation and sustained regression without relapse. Tumor regression is accompanied by the re-establishment of normal crypt-villus homeostasis, such that once aberrantly proliferating cells reacquire self-renewal and multi-lineage differentiation capability. Our study reveals that CRC cells can revert to functioning normal cells given appropriate signals and provide compelling in vivo validation of the Wnt pathway as a therapeutic target for treatment of CRC.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Colorectal Neoplasms/genetics , Disease Models, Animal , Intestine, Large/pathology , Intestine, Small/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Cell Proliferation , Colorectal Neoplasms/pathology , Doxycycline/administration & dosage , Genes, p53 , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Intestine, Large/metabolism , Intestine, Small/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , Wnt Signaling Pathway
2.
Cell ; 158(3): 579-92, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083869

ABSTRACT

The p53 tumor suppressor coordinates a series of antiproliferative responses that restrict the expansion of malignant cells, and as a consequence, p53 is lost or mutated in the majority of human cancers. Here, we show that p53 restricts expression of the stem and progenitor-cell-associated protein nestin in an Sp1/3 transcription-factor-dependent manner and that Nestin is required for tumor initiation in vivo. Moreover, loss of p53 facilitates dedifferentiation of mature hepatocytes into nestin-positive progenitor-like cells, which are poised to differentiate into hepatocellular carcinomas (HCCs) or cholangiocarcinomas (CCs) in response to lineage-specific mutations that target Wnt and Notch signaling, respectively. Many human HCCs and CCs show elevated nestin expression, which correlates with p53 loss of function and is associated with decreased patient survival. Therefore, transcriptional repression of Nestin by p53 restricts cellular plasticity and tumorigenesis in liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nestin/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic , Hepatocytes/metabolism , Humans , Liver Neoplasms/pathology , Mice , Prognosis , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
3.
Cell ; 148(6): 1099-109, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424222

ABSTRACT

Analyses of the human genome have proven extremely successful in identifying changes that contribute to human disease. Genetically engineered mice provide a powerful tool to analyze these changes, although they are slow and costly and do not always recapitulate human biology. Recent advances in genomic technologies, rodent-modeling approaches, and the production of patient-derived reprogrammed cell lines now provide a plethora of complementary systems to study disease states and test new therapies. Continued evolution and integration of these model systems will be the key to realizing the benefits of the genomic revolution and refining our understanding and treatment of human diseases.


Subject(s)
Disease Models, Animal , Animals , Humans , Induced Pluripotent Stem Cells , Mice , Mice, Knockout , Mice, Transgenic , RNA Interference , Rats
4.
Nature ; 597(7875): 263-267, 2021 09.
Article in English | MEDLINE | ID: mdl-34408323

ABSTRACT

Fructose consumption is linked to the rising incidence of obesity and cancer, which are two of the leading causes of morbidity and mortality globally1,2. Dietary fructose metabolism begins at the epithelium of the small intestine, where fructose is transported by glucose transporter type 5 (GLUT5; encoded by SLC2A5) and phosphorylated by ketohexokinase to form fructose 1-phosphate, which accumulates to high levels in the cell3,4. Although this pathway has been implicated in obesity and tumour promotion, the exact mechanism that drives these pathologies in the intestine remains unclear. Here we show that dietary fructose improves the survival of intestinal cells and increases intestinal villus length in several mouse models. The increase in villus length expands the surface area of the gut and increases nutrient absorption and adiposity in mice that are fed a high-fat diet. In hypoxic intestinal cells, fructose 1-phosphate inhibits the M2 isoform of pyruvate kinase to promote cell survival5-7. Genetic ablation of ketohexokinase or stimulation of pyruvate kinase prevents villus elongation and abolishes the nutrient absorption and tumour growth that are induced by feeding mice with high-fructose corn syrup. The ability of fructose to promote cell survival through an allosteric metabolite thus provides additional insights into the excess adiposity generated by a Western diet, and a compelling explanation for the promotion of tumour growth by high-fructose corn syrup.


Subject(s)
Fructose/pharmacology , High Fructose Corn Syrup/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Nutrients/metabolism , Animals , Cell Survival/drug effects , Enzyme Activation , Female , Fructokinases/metabolism , Fructose/metabolism , High Fructose Corn Syrup/metabolism , Hypoxia/diet therapy , Hypoxia/pathology , Intestinal Mucosa/metabolism , Lipid Metabolism/drug effects , Male , Mice , Pyruvate Kinase/metabolism
5.
Cell ; 145(1): 145-58, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21458673

ABSTRACT

RNA interference is a powerful tool for studying gene function, however, the reproducible generation of RNAi transgenic mice remains a significant limitation. By combining optimized fluorescence-coupled miR30-based shRNAs with high efficiency ES cell targeting, we developed a fast, scalable pipeline for the production of shRNA transgenic mice. Using this system, we generated eight tet-regulated shRNA transgenic lines targeting Firefly and Renilla luciferases, Oct4 and tumor suppressors p53, p16(INK4a), p19(ARF) and APC and demonstrate potent gene silencing and GFP-tracked knockdown in a broad range of tissues in vivo. Further, using an shRNA targeting APC, we illustrate how this approach can identify predicted phenotypes and also unknown functions for a well-studied gene. In addition, through regulated gene silencing we validate APC/Wnt and p19(ARF) as potential therapeutic targets in T cell acute lymphoblastic leukemia/lymphoma and lung adenocarcinoma, respectively. This system provides a cost-effective and scalable platform for the production of RNAi transgenic mice targeting any mammalian gene. PAPERCLIP:


Subject(s)
Gene Knockdown Techniques/methods , RNA Interference , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Animals , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques/economics , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mice , Mice, Transgenic , MicroRNAs/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , RNA Processing, Post-Transcriptional , RNA, Small Interfering/genetics , Signal Transduction , Wnt Proteins/metabolism
7.
Am J Pathol ; 194(6): 927-940, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38417696

ABSTRACT

Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract that are largely driven by immune cell activity, and mucosal healing is critical for remission. Serine is a nonessential amino acid that supports epithelial and immune cell metabolism and proliferation; however, whether these roles affect IBD pathogenesis is not well understood. Herein, the study showed that serine synthesis increased selectively in the epithelial cells of colons from patients with IBD and murine models of colitis. Inhibiting serine synthesis impaired colonic mucosal healing and increased susceptibility to acute injury in mice, effects associated with diminished epithelial cell proliferation. Dietary removal of serine similarly sensitized mice to acute chemically induced colitis but ameliorated inflammation in chronic colitis models. The anti-inflammatory effect of exogenous serine depletion in chronic colitis was associated with mitochondrial dysfunction of macrophages, resulting in impaired nucleotide production and proliferation. Collectively, these results suggest that serine plays an important role in both epithelial and immune cell biology in the colon and that modulating its availability could impact IBD pathogenesis.


Subject(s)
Cell Proliferation , Colitis , Epithelial Cells , Intestinal Mucosa , Serine , Animals , Colitis/immunology , Colitis/pathology , Colitis/chemically induced , Mice , Humans , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Serine/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Female , Colon/pathology , Colon/immunology , Colon/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Disease Models, Animal
8.
Proc Natl Acad Sci U S A ; 119(17): e2110557119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35442775

ABSTRACT

Anticancer drug development campaigns often fail due to an incomplete understanding of the therapeutic index differentiating the efficacy of the agent against the cancer and its on-target toxicities to the host. To address this issue, we established a versatile preclinical platform in which genetically defined cancers are produced using somatic tissue engineering in transgenic mice harboring a doxycycline-inducible short hairpin RNA against the target of interest. In this system, target inhibition is achieved by the addition of doxycycline, enabling simultaneous assessment of efficacy and toxicity in the same animal. As proof of concept, we focused on CDK9­a cancer target whose clinical development has been hampered by compounds with poorly understood target specificity and unacceptable toxicities. We systematically compared phenotypes produced by genetic Cdk9 inhibition to those achieved using a recently developed highly specific small molecule CDK9 inhibitor and found that both perturbations led to robust antitumor responses. Remarkably, nontoxic levels of CDK9 inhibition could achieve significant treatment efficacy, and dose-dependent toxicities produced by prolonged CDK9 suppression were largely reversible upon Cdk9 restoration or drug withdrawal. Overall, these results establish a versatile in vivo target validation platform that can be employed for rapid triaging of therapeutic targets and lend support to efforts aimed at advancing CDK9 inhibitors for cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/genetics , RNA Interference
9.
EMBO J ; 39(5): e102169, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31930530

ABSTRACT

Genetically engineered mouse models (GEMMs) of cancer have proven to be of great value for basic and translational research. Although CRISPR-based gene disruption offers a fast-track approach for perturbing gene function and circumvents certain limitations of standard GEMM development, it does not provide a flexible platform for recapitulating clinically relevant missense mutations in vivo. To this end, we generated knock-in mice with Cre-conditional expression of a cytidine base editor and tested their utility for precise somatic engineering of missense mutations in key cancer drivers. Upon intraductal delivery of sgRNA-encoding vectors, we could install point mutations with high efficiency in one or multiple endogenous genes in situ and assess the effect of defined allelic variants on mammary tumorigenesis. While the system also produces bystander insertions and deletions that can stochastically be selected for when targeting a tumor suppressor gene, we could effectively recapitulate oncogenic nonsense mutations. We successfully applied this system in a model of triple-negative breast cancer, providing the proof of concept for extending this flexible somatic base editing platform to other tissues and tumor types.


Subject(s)
Breast Neoplasms/genetics , CRISPR-Cas Systems , Gene Editing , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Mutation
10.
PLoS Genet ; 17(12): e1009941, 2021 12.
Article in English | MEDLINE | ID: mdl-34879057

ABSTRACT

The retinoblastoma (RB) tumor suppressor is functionally inactivated in a wide range of human tumors where this inactivation promotes tumorigenesis in part by allowing uncontrolled proliferation. RB has been extensively studied, but its mechanisms of action in normal and cancer cells remain only partly understood. Here, we describe a new mouse model to investigate the consequences of RB depletion and its re-activation in vivo. In these mice, induction of shRNA molecules targeting RB for knock-down results in the development of phenotypes similar to Rb knock-out mice, including the development of pituitary and thyroid tumors. Re-expression of RB leads to cell cycle arrest in cancer cells and repression of transcriptional programs driven by E2F activity. Thus, continuous RB loss is required for the maintenance of tumor phenotypes initiated by loss of RB, and this new mouse model will provide a new platform to investigate RB function in vivo.


Subject(s)
Pituitary Neoplasms/genetics , Retinoblastoma Binding Proteins/genetics , Thyroid Neoplasms/genetics , Animals , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Disease Models, Animal , E2F Transcription Factors/metabolism , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Transgenic , NIH 3T3 Cells , Pituitary Neoplasms/pathology , RNA, Small Interfering/metabolism , Thyroid Neoplasms/pathology
11.
J Biol Chem ; 297(1): 100815, 2021 07.
Article in English | MEDLINE | ID: mdl-34023388

ABSTRACT

Cyclic AMP-responsive element-binding protein H (CREBH encoded by Creb3l3) is a transcription factor that regulates the expression of genes that control lipid and glucose metabolism as well as inflammation. CREBH is upregulated in the liver under conditions of overnutrition, and mice globally lacking the gene (CREBH-/-) are highly susceptible to diet-induced obesity, insulin resistance, and hepatic steatosis. The net protective effects of CREBH have been attributed in large part to the activities of fibroblast growth factor (Fgf)-21 (Fgf21), a target gene that promotes weight loss, improves glucose homeostasis, and reduces hepatic lipid accumulation. To explore the possibility that activation of the CREBH-Fgf21 axis could ameliorate established effects of high-fat feeding, we generated an inducible transgenic hepatocyte-specific CREBH overexpression mouse model (Tg-rtTA). Acute overexpression of CREBH in livers of Tg-rtTA mice effectively reversed diet-induced obesity, insulin resistance, and hepatic steatosis. These changes were associated with increased activities of thermogenic brown and beige adipose tissues in Tg-rtTA mice, leading to reductions in fat mass, along with enhanced insulin sensitivity and glucose tolerance. Genetically silencing Fgf21 in Tg-rtTA mice abrogated the CREBH-mediated reductions in body weight loss, but only partially reversed the observed improvements in glucose metabolism. These findings reveal that the protective effects of CREBH activation may be leveraged to mitigate diet-induced obesity and associated metabolic abnormalities in both Fgf21-dependent and Fgf21-independent pathways.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Diet , Fatty Liver/genetics , Fatty Liver/pathology , Insulin Resistance/genetics , Liver/metabolism , Obesity/genetics , Adiposity , Animals , Body Weight , Cyclic AMP Response Element-Binding Protein/metabolism , Energy Metabolism , Feeding Behavior , Fibroblast Growth Factors/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout
12.
BMC Genomics ; 23(1): 792, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36457077

ABSTRACT

Somatic mutations drive colorectal cancer (CRC) by disrupting gene regulatory mechanisms. Distinct combinations of mutations can result in unique changes to regulatory mechanisms leading to variability in the efficacy of therapeutics. MicroRNAs are important regulators of gene expression, and their activity can be altered by oncogenic mutations. However, it is unknown how distinct combinations of CRC-risk mutations differentially affect microRNAs. Here, using genetically-modified mouse intestinal organoid (enteroid) models, we identify 12 different modules of microRNA expression patterns across different combinations of mutations common in CRC. We also show that miR-24-3p is aberrantly upregulated in genetically-modified mouse enteroids irrespective of mutational context. Furthermore, we identify an enrichment of miR-24-3p predicted targets in downregulated gene lists from various mutational contexts compared to WT. In follow-up experiments, we demonstrate that miR-24-3p promotes CRC cell survival in multiple cell contexts. Our novel characterization of genotype-specific patterns of miRNA expression offer insight into the mechanisms that drive inter-tumor heterogeneity and highlight candidate microRNA therapeutic targets for the advancement of precision medicine for CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Animals , Mice , Cell Survival/genetics , Colorectal Neoplasms/genetics , Genotype , MicroRNAs/genetics , Organoids
13.
Nucleic Acids Res ; 48(6): 2841-2852, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32112097

ABSTRACT

Base editing (BE) is a powerful tool for engineering single nucleotide variants (SNVs) and has been used to create targeted mutations in cell lines, organoids and animal models. Recent development of new BE enzymes has provided an extensive toolkit for genome modification; however, identifying and isolating edited cells for analysis has proven challenging. Here we report a 'Gene On' (GO) reporter system that indicates precise cytosine or adenine base editing in situ with high sensitivity and specificity. We test GO using an activatable GFP and use it to measure the kinetics, efficiency and PAM specificity of a range of new BE variants. Further, GO is flexible and can be easily adapted to induce expression of numerous genetically encoded markers, antibiotic resistance genes or enzymes, such as Cre recombinase. With these tools, GO can be exploited to functionally link BE events at endogenous genomic loci to cellular enzymatic activities in human and mouse cell lines and organoids. Thus, GO provides a powerful approach to increase the practicality and feasibility of implementing CRISPR BE in biomedical research.


Subject(s)
Gene Editing , Genes, Reporter , Animals , Base Sequence , Cell Line, Tumor , Drug Resistance, Microbial , HEK293 Cells , Humans , Integrases/metabolism , Mice , NIH 3T3 Cells , Recombination, Genetic/genetics
14.
Proc Natl Acad Sci U S A ; 116(41): 20672-20678, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31551264

ABSTRACT

Radiation-induced gastrointestinal syndrome (RIGS) is a limiting factor for therapeutic abdominopelvic radiation and is predicted to be a major source of morbidity in the event of a nuclear accident or radiological terrorism. In this study, we developed an in vivo mouse-modeling platform that enables spatial and temporal manipulation of potential RIGS targets in mice following whole-abdomen irradiation without the confounding effects of concomitant hematopoietic syndrome that occur following whole-body irradiation. We then tested the utility of this platform to explore the effects of transient Wnt pathway activation on intestinal regeneration and animal recovery following induction of RIGS. Our results demonstrate that intestinal epithelial suppression of adenomatous polyposis coli (Apc) mitigates RIGS lethality in vivo after lethal ionizing radiation injury-induced intestinal epithelial damage. These results highlight the potential of short-term Wnt agonism as a therapeutic target and establish a platform to evaluate other strategies to stimulate intestinal regeneration after ionizing radiation damage.


Subject(s)
Adenomatous Polyposis Coli Protein/antagonists & inhibitors , Gastrointestinal Diseases/prevention & control , Intestines/cytology , Radiation Injuries, Experimental/prevention & control , Regeneration , Whole-Body Irradiation/adverse effects , Wnt Proteins/metabolism , Animals , Gastrointestinal Diseases/etiology , Gastrointestinal Diseases/metabolism , Intestines/radiation effects , Mice , RNA, Small Interfering , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Syndrome
15.
Genes Dev ; 28(1): 85-97, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24395249

ABSTRACT

Genetically engineered mouse models (GEMMs) have greatly expanded our knowledge of pancreatic ductal adenocarcinoma (PDAC) and serve as a critical tool to identify and evaluate new treatment strategies. However, the cost and time required to generate conventional pancreatic cancer GEMMs limits their use for investigating novel genetic interactions in tumor development and maintenance. To address this problem, we developed flexible embryonic stem cell (ESC)-based GEMMs that facilitate the rapid generation of genetically defined multiallelic chimeric mice without further strain intercrossing. The ESCs harbor a latent Kras mutant (a nearly ubiquitous feature of pancreatic cancer), a homing cassette, and other genetic elements needed for rapid insertion and conditional expression of tetracycline-controlled transgenes, including fluorescence-coupled shRNAs capable of efficiently silencing gene function by RNAi. This system produces a disease that recapitulates the progression of pancreatic cancer in human patients and enables the study and visualization of the impact of gene perturbation at any stage of pancreas cancer progression. We describe the use of this approach to dissect temporal roles for the tumor suppressor Pten and the oncogene c-Myc in pancreatic cancer development and maintenance.


Subject(s)
Disease Models, Animal , Embryonic Stem Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Animals, Genetically Modified , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Reproducibility of Results
16.
Nature ; 525(7570): 543-547, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26367798

ABSTRACT

Following the discovery of BRD4 as a non-oncogene addiction target in acute myeloid leukaemia (AML), bromodomain and extra terminal protein (BET) inhibitors are being explored as a promising therapeutic avenue in numerous cancers. While clinical trials have reported single-agent activity in advanced haematological malignancies, mechanisms determining the response to BET inhibition remain poorly understood. To identify factors involved in primary and acquired BET resistance in leukaemia, here we perform a chromatin-focused RNAi screen in a sensitive MLL-AF9;Nras(G12D)-driven AML mouse model, and investigate dynamic transcriptional profiles in sensitive and resistant mouse and human leukaemias. Our screen shows that suppression of the PRC2 complex, contrary to effects in other contexts, promotes BET inhibitor resistance in AML. PRC2 suppression does not directly affect the regulation of Brd4-dependent transcripts, but facilitates the remodelling of regulatory pathways that restore the transcription of key targets such as Myc. Similarly, while BET inhibition triggers acute MYC repression in human leukaemias regardless of their sensitivity, resistant leukaemias are uniformly characterized by their ability to rapidly restore MYC transcription. This process involves the activation and recruitment of WNT signalling components, which compensate for the loss of BRD4 and drive resistance in various cancer models. Dynamic chromatin immunoprecipitation sequencing and self-transcribing active regulatory region sequencing of enhancer profiles reveal that BET-resistant states are characterized by remodelled regulatory landscapes, involving the activation of a focal MYC enhancer that recruits WNT machinery in response to BET inhibition. Together, our results identify and validate WNT signalling as a driver and candidate biomarker of primary and acquired BET resistance in leukaemia, and implicate the rewiring of transcriptional programs as an important mechanism promoting resistance to BET inhibitors and, potentially, other chromatin-targeted therapies.


Subject(s)
Azepines/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/drug effects , Triazoles/pharmacology , Animals , Cell Cycle Proteins , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Genes, myc/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Mice , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , Wnt Signaling Pathway/drug effects
17.
Carcinogenesis ; 41(3): 334-344, 2020 05 14.
Article in English | MEDLINE | ID: mdl-31170286

ABSTRACT

Beyond the nearly uniform presence of KRAS mutations, pancreatic cancer is increasingly recognized as a heterogeneous disease. Preclinical in vivo model systems exist, but with the advent of precision oncology, murine models with enhanced genetic flexibility are needed to functionally annotate genetic alterations found in the human malignancy. Here, we describe the generation of focal gene disruptions and large chromosomal deletions via inducible and pancreas-specific expression of Cas9 in adult mice. Experimental mice are derived on demand directly from genetically engineered embryonic stem cells, without the need for further intercrossing. To provide initial validation of our approach, we show that disruption of the E3 ubiquitin ligase Rnf43 accelerates KrasG12D-dependent tumourigenesis. Moreover, we demonstrate that this system can be used to rapidly interrogate the impact of complex cancer-associated alleles through the generation of a previously unstudied 1.2 megabase deletion surrounding the CDKN2A and CDKN2B tumour suppressors. Thus, our approach is capable of reproducibly generating biallelic and precise loss of large chromosomal fragments that, in conjunction with mutant Kras, leads to development of pancreatic ductal adenocarcinoma with full penetrance.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Editing , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Genome, Human/genetics , Humans , Mice , Mutation/genetics , Pancreas/pathology , Pancreatic Neoplasms/pathology , Precision Medicine , Sequence Deletion/genetics , Pancreatic Neoplasms
18.
Methods ; 164-165: 100-108, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30836137

ABSTRACT

Base editing is a powerful technology that enables programmable conversion of single nucleotides in the mammalian genome. Base editors consist of a partially active Cas9 nuclease (Cas9D10A) tethered to a natural or synthetic DNA modifying enzyme. Though only recently described, BE has already shown enormous potential for basic and translational research, allowing the creation or repair of disease alleles in a variety of cell types and model organisms. In the past 2 years, a vast array of new and modified base editor variants have been described, expanding the flexibility and usefulness of the approach. Though simple in concept, effective implementation of base editing requires an understanding of the advantages and limitations of each of these tools. Here, we provide an overview of the concepts of DNA base editing, and discuss the recent progress toward the development of optimized base editing systems for mammalian cells. In addition, we highlight key technical aspects of designing and executing BE experiments, and provide detailed experimental examples of successful base editing in cell lines and organoids to help guide the effective use of these tools for genome modification.


Subject(s)
Base Sequence/genetics , CRISPR-Cas Systems/genetics , Gene Editing/methods , Animals , Cell Culture Techniques/methods , Cell Line , DNA/genetics , RNA, Guide, Kinetoplastida/genetics
19.
Proc Natl Acad Sci U S A ; 113(11): 3030-5, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929372

ABSTRACT

Loss of the tumor suppressor gene PTEN is implicated in breast cancer progression and resistance to targeted therapies, and is thought to promote tumorigenesis by activating PI3K signaling. In a transgenic model of breast cancer, Pten suppression using a tetracycline-regulatable short hairpin (sh)RNA cooperates with human epidermal growth factor receptor 2 (HER2/neu), leading to aggressive and metastatic disease with elevated signaling through PI3K and, surprisingly, the mitogen-activated protein kinase (MAPK) pathway. Restoring Pten function is sufficient to down-regulate both PI3K and MAPK signaling and triggers dramatic tumor regression. Pharmacologic inhibition of MAPK signaling produces similar effects to Pten restoration, suggesting that the MAPK pathway contributes to the maintenance of advanced breast cancers harboring Pten loss.


Subject(s)
MAP Kinase Signaling System/physiology , Mammary Neoplasms, Experimental/physiopathology , Neoplasm Proteins/physiology , PTEN Phosphohydrolase/deficiency , Receptor, ErbB-2/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genes, erbB-2 , Humans , MAP Kinase Kinase 1/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Proteins/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/physiology , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/physiology
20.
PLoS Biol ; 13(3): e1002107, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25807062

ABSTRACT

The development and regeneration of myelin by oligodendrocytes, the myelin-forming cells of the central nervous system (CNS), requires profound changes in cell shape that lead to myelin sheath initiation and formation. Here, we demonstrate a requirement for the basal polarity complex protein Scribble in CNS myelination and remyelination. Scribble is expressed throughout oligodendroglial development and is up-regulated in mature oligodendrocytes where it is localised to both developing and mature CNS myelin sheaths. Knockdown of Scribble expression in cultured oligodendroglia results in disrupted morphology and myelination initiation. When Scribble expression is conditionally eliminated in the myelinating glia of transgenic mice, myelin initiation in CNS is disrupted, both during development and following focal demyelination, and longitudinal extension of the myelin sheath is disrupted. At later stages of myelination, Scribble acts to negatively regulate myelin thickness whilst suppressing the extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAP) kinase pathway, and localises to non-compact myelin flanking the node of Ranvier where it is required for paranodal axo-glial adhesion. These findings demonstrate an essential role for the evolutionarily-conserved regulators of intracellular polarity in myelination and remyelination.


Subject(s)
Central Nervous System/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Oligodendroglia/metabolism , Animals , Cell Polarity , Central Nervous System/ultrastructure , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Oligodendroglia/ultrastructure , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL