Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Nature ; 619(7970): 632-639, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37344599

ABSTRACT

Sex exerts a profound impact on cancer incidence, spectrum and outcomes, yet the molecular and genetic bases of such sex differences are ill-defined and presumptively ascribed to X-chromosome genes and sex hormones1. Such sex differences are particularly prominent in colorectal cancer (CRC) in which men experience higher metastases and mortality. A murine CRC model, engineered with an inducible transgene encoding oncogenic mutant KRASG12D and conditional null alleles of Apc and Trp53 tumour suppressors (designated iKAP)2, revealed higher metastases and worse outcomes specifically in males with oncogenic mutant KRAS (KRAS*) CRC. Integrated cross-species molecular and transcriptomic analyses identified Y-chromosome gene histone demethylase KDM5D as a transcriptionally upregulated gene driven by KRAS*-mediated activation of the STAT4 transcription factor. KDM5D-dependent chromatin mark and transcriptome changes showed repression of regulators of the epithelial cell tight junction and major histocompatibility complex class I complex components. Deletion of Kdm5d in iKAP cancer cells increased tight junction integrity, decreased cell invasiveness and enhanced cancer cell killing by CD8+ T cells. Conversely, iAP mice engineered with a Kdm5d transgene to provide constitutive Kdm5d expression specifically in iAP cancer cells showed an increased propensity for more invasive tumours in vivo. Thus, KRAS*-STAT4-mediated upregulation of Y chromosome KDM5D contributes substantially to the sex differences in KRAS* CRC by means of its disruption of cancer cell adhesion properties and tumour immunity, providing an actionable therapeutic strategy for metastasis risk reduction for men afflicted with KRAS* CRC.


Subject(s)
Colorectal Neoplasms , Histone Demethylases , Minor Histocompatibility Antigens , Sex Characteristics , Animals , Female , Humans , Male , Mice , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Histone Demethylases/genetics , Histone Demethylases/metabolism , Mice, Transgenic , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Up-Regulation
3.
Nat Aging ; 1(12): 1162-1174, 2021 12.
Article in English | MEDLINE | ID: mdl-35036927

ABSTRACT

Amyloid-induced neurodegeneration plays a central role in Alzheimer's disease (AD) pathogenesis. Here, we show that telomerase reverse transcriptase (TERT) haploinsufficiency decreases BDNF and increases amyloid-ß (Aß) precursor in murine brain. Moreover, prior to disease onset, the TERT locus sustains accumulation of repressive epigenetic marks in murine and human AD neurons, implicating TERT repression in amyloid-induced neurodegeneration. To test the impact of sustained TERT expression on AD pathobiology, AD mouse models were engineered to maintain physiological levels of TERT in adult neurons, resulting in reduced Aß accumulation, improved spine morphology, and preserved cognitive function. Mechanistically, integrated profiling revealed that TERT interacts with ß-catenin and RNA polymerase II at gene promoters and upregulates gene networks governing synaptic signaling and learning processes. These TERT-directed transcriptional activities do not require its catalytic activity nor telomerase RNA. These findings provide genetic proof-of-concept for somatic TERT gene activation therapy in attenuating AD progression including cognitive decline.


Subject(s)
Alzheimer Disease , Telomerase , Mice , Humans , Animals , Alzheimer Disease/genetics , Telomerase/genetics , Amyloid beta-Peptides/metabolism , Cognition , Neurons/metabolism
4.
Cancer Discov ; 10(9): 1374-1387, 2020 09.
Article in English | MEDLINE | ID: mdl-32385075

ABSTRACT

Genetic inactivation of PTEN is common in prostate cancer and correlates with poorer prognosis. We previously identified CHD1 as an essential gene in PTEN-deficient cancer cells. Here, we sought definitive in vivo genetic evidence for, and mechanistic understanding of, the essential role of CHD1 in PTEN-deficient prostate cancer. In Pten and Pten/Smad4 genetically engineered mouse models, prostate-specific deletion of Chd1 resulted in markedly delayed tumor progression and prolonged survival. Chd1 deletion was associated with profound tumor microenvironment (TME) remodeling characterized by reduced myeloid-derived suppressor cells (MDSC) and increased CD8+ T cells. Further analysis identified IL6 as a key transcriptional target of CHD1, which plays a major role in recruitment of immunosuppressive MDSCs. Given the prominent role of MDSCs in suppressing responsiveness to immune checkpoint inhibitors (ICI), our genetic and tumor biological findings support combined testing of anti-IL6 and ICI therapies, specifically in PTEN-deficient prostate cancer. SIGNIFICANCE: We demonstrate a critical role of CHD1 in MDSC recruitment and discover CHD1/IL6 as a major regulator of the immunosuppressive TME of PTEN-deficient prostate cancer. Pharmacologic inhibition of IL6 in combination with immune checkpoint blockade elicits robust antitumor responses in prostate cancer.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
DNA-Binding Proteins/metabolism , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/genetics , Tumor Escape/genetics , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/immunology , Humans , Male , Mice, Transgenic , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Smad4 Protein/genetics , Tumor Microenvironment/genetics
6.
Nature ; 543(7647): 728-732, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28321130

ABSTRACT

A significant fraction of patients with advanced prostate cancer treated with androgen deprivation therapy experience relapse with relentless progression to lethal metastatic castration-resistant prostate cancer (mCRPC). Immune checkpoint blockade using antibodies against cytotoxic-T-lymphocyte-associated protein 4 (CTLA4) or programmed cell death 1/programmed cell death 1 ligand 1 (PD1/PD-L1) generates durable therapeutic responses in a significant subset of patients across a variety of cancer types. However, mCRPC showed overwhelming de novo resistance to immune checkpoint blockade, motivating a search for targeted therapies that overcome this resistance. Myeloid-derived suppressor cells (MDSCs) are known to play important roles in tumour immune evasion. The abundance of circulating MDSCs correlates with prostate-specific antigen levels and metastasis in patients with prostate cancer. Mouse models of prostate cancer show that MDSCs (CD11b+Gr1+) promote tumour initiation and progression. These observations prompted us to hypothesize that robust immunotherapy responses in mCRPC may be elicited by the combined actions of immune checkpoint blockade agents together with targeted agents that neutralize MDSCs yet preserve T-cell function. Here we develop a novel chimaeric mouse model of mCRPC to efficiently test combination therapies in an autochthonous setting. Combination of anti-CTLA4 and anti-PD1 engendered only modest efficacy. Targeted therapy against mCRPC-infiltrating MDSCs, using multikinase inhibitors such as cabozantinib and BEZ235, also showed minimal anti-tumour activities. Strikingly, primary and metastatic CRPC showed robust synergistic responses when immune checkpoint blockade was combined with MDSC-targeted therapy. Mechanistically, combination therapy efficacy stemmed from the upregulation of interleukin-1 receptor antagonist and suppression of MDSC-promoting cytokines secreted by prostate cancer cells. These observations illuminate a clinical path hypothesis for combining immune checkpoint blockade with MDSC-targeted therapies in the treatment of mCRPC.


Subject(s)
Immunotherapy/methods , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms, Castration-Resistant/therapy , Anilides/pharmacology , Anilides/therapeutic use , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chimera , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Female , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Molecular Targeted Therapy , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms, Castration-Resistant/pathology , Pyridines/pharmacology , Pyridines/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
7.
Nat Genet ; 49(1): 75-86, 2017 01.
Article in English | MEDLINE | ID: mdl-27841882

ABSTRACT

Stem cells, including cancer stem cells (CSCs), require niches to maintain stemness, yet it is unclear how CSCs maintain stemness in the suboptimal environment outside their niches during invasion. Postnatal co-deletion of Pten and Trp53 in mouse neural stem cells (NSCs) leads to the expansion of these cells in their subventricular zone (SVZ) niches but fails to maintain stemness outside the SVZ. We discovered that Qki is a major regulator of NSC stemness. Qk deletion on a Pten-/-; Trp53-/- background helps NSCs maintain their stemness outside the SVZ in Nes-CreERT2; QkL/L; PtenL/L; Trp53L/L mice, which develop glioblastoma with a penetrance of 92% and a median survival time of 105 d. Mechanistically, Qk deletion decreases endolysosome-mediated degradation and enriches receptors essential for maintaining self-renewal on the cytoplasmic membrane to cope with low ligand levels outside niches. Thus, downregulation of endolysosome levels by Qki loss helps glioma stem cells (GSCs) maintain their stemness in suboptimal environments outside their niches.


Subject(s)
Brain Neoplasms/pathology , Endosomes/metabolism , Glioma/pathology , Lysosomes/metabolism , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , RNA-Binding Proteins/physiology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cells, Cultured , Female , Glioma/genetics , Glioma/metabolism , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , PTEN Phosphohydrolase/physiology , Proteolysis , Receptors, Cell Surface/metabolism , Stem Cell Niche , Tumor Suppressor Protein p53/physiology
8.
Cancer Discov ; 5(12): 1314-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26450788

ABSTRACT

UNLABELLED: Epigenetic regulators have emerged as critical factors governing the biology of cancer. Here, in the context of melanoma, we show that RNF2 is prognostic, exhibiting progression-correlated expression in human melanocytic neoplasms. Through a series of complementary gain-of-function and loss-of-function studies in mouse and human systems, we establish that RNF2 is oncogenic and prometastatic. Mechanistically, RNF2-mediated invasive behavior is dependent on its ability to monoubiquitinate H2AK119 at the promoter of LTBP2, resulting in silencing of this negative regulator of TGFß signaling. In contrast, RNF2's oncogenic activity does not require its catalytic activity nor does it derive from its canonical gene repression function. Instead, RNF2 drives proliferation through direct transcriptional upregulation of the cell-cycle regulator CCND2. We further show that MEK1-mediated phosphorylation of RNF2 promotes recruitment of activating histone modifiers UTX and p300 to a subset of poised promoters, which activates gene expression. In summary, RNF2 regulates distinct biologic processes in the genesis and progression of melanoma via different molecular mechanisms. SIGNIFICANCE: The role of epigenetic regulators in cancer progression is being increasingly appreciated. We show novel roles for RNF2 in melanoma tumorigenesis and metastasis, albeit via different mechanisms. Our findings support the notion that epigenetic regulators, such as RNF2, directly and functionally control powerful gene networks that are vital in multiple cancer processes.


Subject(s)
Melanoma/genetics , Melanoma/pathology , Polycomb Repressive Complex 1/genetics , Animals , Catalysis , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cyclin D2/genetics , Cyclin D2/metabolism , Disease Progression , E1A-Associated p300 Protein/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Humans , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , MAP Kinase Signaling System , Melanoma/metabolism , Mice , Neoplasm Metastasis , Nuclear Proteins/metabolism , Oncogenes , Phosphorylation , Polycomb Repressive Complex 1/metabolism , Prognosis , Promoter Regions, Genetic , Transforming Growth Factor beta/metabolism
9.
Cancer Cell ; 27(5): 644-57, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25965571

ABSTRACT

Myelodysplastic syndrome (MDS) risk correlates with advancing age, therapy-induced DNA damage, and/or shorter telomeres, but whether telomere erosion directly induces MDS is unknown. Here, we provide the genetic evidence that telomere dysfunction-induced DNA damage drives classical MDS phenotypes and alters common myeloid progenitor (CMP) differentiation by repressing the expression of mRNA splicing/processing genes, including SRSF2. RNA-seq analyses of telomere dysfunctional CMP identified aberrantly spliced transcripts linked to pathways relevant to MDS pathogenesis such as genome stability, DNA repair, chromatin remodeling, and histone modification, which are also enriched in mouse CMP haploinsufficient for SRSF2 and in CD34(+) CMML patient cells harboring SRSF2 mutation. Together, our studies establish an intimate link across telomere biology, aberrant RNA splicing, and myeloid progenitor differentiation.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/genetics , Myelodysplastic Syndromes/genetics , Telomere , Animals , Haploinsufficiency , Humans , Mice , Myelodysplastic Syndromes/pathology , Nuclear Proteins/genetics , RNA Splicing , Ribonucleoproteins/genetics , Serine-Arginine Splicing Factors
10.
Cancer Res ; 75(10): 2109-19, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25808866

ABSTRACT

Epidermal growth factor receptor (EGFR) is highly amplified, mutated, and overexpressed in human malignant gliomas. Despite its prevalence and growth-promoting functions, therapeutic strategies to inhibit EGFR kinase activity have not been translated into profound beneficial effects in glioma clinical trials. To determine the roles of oncogenic EGFR signaling in gliomagenesis and tumor maintenance, we generated a novel glioma mouse model driven by inducible expression of a mutant EGFR (EGFR*). Using combined genetic and pharmacologic interventions, we revealed that EGFR*-driven gliomas were insensitive to EGFR tyrosine kinase inhibitors, although they could efficiently inhibit EGFR* autophosphorylation in vitro and in vivo. This is in contrast with the genetic suppression of EGFR* induction that led to significant tumor regression and prolonged animal survival. However, despite their initial response to genetic EGFR* extinction, all tumors would relapse and propagate independent of EGFR*. We further showed that EGFR*-independent tumor cells existed prior to treatment and were responsible for relapse following genetic EGFR* suppression. And, the addition of a PI3K/mTOR inhibitor could significantly delay relapse and prolong animal survival. Our findings shed mechanistic insight into EGFR drug resistance in glioma and provide a platform to test therapies targeting aberrant EGFR signaling in this setting.


Subject(s)
Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Glioma/drug therapy , Animals , Brain Neoplasms/metabolism , Crizotinib , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Doxycycline/pharmacology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gefitinib , Glioma/metabolism , Humans , Imidazoles/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphorylation , Protein Processing, Post-Translational , Pyrazoles/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Quinolines/pharmacology , Tumor Cells, Cultured
11.
PLoS One ; 9(2): e89160, 2014.
Article in English | MEDLINE | ID: mdl-24586561

ABSTRACT

Genetic studies of rare diseases can identify genes of unknown function that strongly impact human physiology. Prolyl endopeptidase-like (PREPL) is an uncharacterized member of the prolyl peptidase family that was discovered because of its deletion in humans with hypotonia-cystinuria syndrome (HCS). HCS is characterized by a number of physiological changes including diminished growth and neonatal hypotonia or low muscle tone. HCS patients have deletions in other genes as well, making it difficult to tease apart the specific role of PREPL. Here, we develop a PREPL null (PREPL(-/-)) mouse model to address the physiological role of this enzyme. Deletion of exon 11 from the Prepl gene, which encodes key catalytic amino acids, leads to a loss of PREPL protein as well as lower Prepl mRNA levels. PREPL(-/-) mice have a pronounced growth phenotype, being significantly shorter and lighter than their wild type (PREPL(+/+)) counterparts. A righting assay revealed that PREPL(-/-) pups took significantly longer than PREPL(+/+) pups to right themselves when placed on their backs. This deficit indicates that PREPL(-/-) mice suffer from neonatal hypotonia. According to these results, PREPL regulates growth and neonatal hypotonia in mice, which supports the idea that PREPL causes diminished growth and neonatal hypotonia in humans with HCS. These animals provide a valuable asset in deciphering the underlying biochemical, cellular and physiological pathways that link PREPL to HCS, and this may eventually lead to new insights in the treatment of this disease.


Subject(s)
Serine Endopeptidases/deficiency , Animals , Chromosome Deletion , Chromosomes, Human, Pair 21/genetics , Craniofacial Abnormalities/etiology , Craniofacial Abnormalities/genetics , Cystinuria/etiology , Cystinuria/genetics , Exons/genetics , Intellectual Disability/etiology , Intellectual Disability/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mitochondrial Diseases/etiology , Mitochondrial Diseases/genetics , Muscle Hypotonia/etiology , Muscle Hypotonia/genetics , Prolyl Oligopeptidases , Serine Endopeptidases/genetics
12.
Cell ; 155(2): 397-409, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120138

ABSTRACT

The pyruvate kinase M2 isoform (PKM2) is expressed in cancer and plays a role in regulating anabolic metabolism. To determine whether PKM2 is required for tumor formation or growth, we generated mice with a conditional allele that abolishes PKM2 expression without disrupting PKM1 expression. PKM2 deletion accelerated mammary tumor formation in a Brca1-loss-driven model of breast cancer. PKM2 null tumors displayed heterogeneous PKM1 expression, with PKM1 found in nonproliferating tumor cells and no detectable pyruvate kinase expression in proliferating cells. This suggests that PKM2 is not necessary for tumor cell proliferation and implies that the inactive state of PKM2 is associated with the proliferating cell population within tumors, whereas nonproliferating tumor cells require active pyruvate kinase. Consistent with these findings, variable PKM2 expression and heterozygous PKM2 mutations are found in human tumors. These data suggest that regulation of PKM2 activity supports the different metabolic requirements of proliferating and nonproliferating tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Gene Deletion , Mammary Neoplasms, Experimental/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Exons , Female , Gene Knockout Techniques , Heterografts , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutagenesis , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , RNA Splicing
13.
Cell ; 148(5): 896-907, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22341455

ABSTRACT

To determine the role of telomere dysfunction and telomerase reactivation in generating pro-oncogenic genomic events and in carcinoma progression, an inducible telomerase reverse transcriptase (mTert) allele was crossed onto a prostate cancer-prone mouse model null for Pten and p53 tumor suppressors. Constitutive telomerase deficiency and associated telomere dysfunction constrained cancer progression. In contrast, telomerase reactivation in the setting of telomere dysfunction alleviated intratumoral DNA-damage signaling and generated aggressive cancers with rearranged genomes and new tumor biological properties (bone metastases). Comparative oncogenomic analysis revealed numerous recurrent amplifications and deletions of relevance to human prostate cancer. Murine tumors show enrichment of the TGF-ß/SMAD4 network, and genetic validation studies confirmed the cooperative roles of Pten, p53, and Smad4 deficiencies in prostate cancer progression, including skeletal metastases. Thus, telomerase reactivation in tumor cells experiencing telomere dysfunction enables full malignant progression and provides a mechanism for acquisition of cancer-relevant genomic events endowing new tumor biological capabilities.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Telomerase/metabolism , Telomere/metabolism , Animals , Bone Neoplasms/secondary , Cell Line, Tumor , Crosses, Genetic , DNA Copy Number Variations , Disease Models, Animal , Female , Genomic Instability , Humans , Male , Mice , Tumor Suppressor Protein p53/metabolism
14.
FASEB J ; 25(3): 979-89, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21135040

ABSTRACT

Telomerase is tightly regulated in humans relative to mice, owing to the differential regulation of TERT genes. To explore hTERT regulation in vivo, we engineered mice with a 160-kb transgenic bacterial artificial chromosome (BAC) spanning the hTERT locus with a Renilla luciferase (Rluc) cassette downstream of its promoter. Analysis of multiple founder lines revealed that the Rluc expression profile from the transgenic hTERT reporter locus reproduced that of the native hTERT gene in all tissues and organs examined, demonstrating that genetic sequence determined the species-specific developmental regulation of the hTERT gene and that mouse epigenetic and transcription machineries faithfully regulated hTERT transcription. Thus, these mice allowed detailed analyses of developmental hTERT regulation. Both the transgenic hTERT reporter and the endogenous mTERT locus were expressed in early embryonic stages, and their mRNA levels progressively decreased throughout embryonic and postnatal development. Whereas hTERT transcription was much lower than mTERT expression in most organs, it increased significantly during postnatal development of thymus, testis, and ovary. In testis, the Rluc mRNA was enriched in elongating spermatids of seminiferous tubules. In addition, the transcription of transgenic hTERT reporter, but surprisingly not the endogenous mTERT gene, was activated during Wnt1-induced mammary tumorigenesis, allowing the monitoring of tumor development via noninvasive bioluminescent imaging. Collectively, our results demonstrate that the hTERT transgenic reporter system recapitulates the developmental regulation of the hTERT gene in a chromosomal position-independent manner and serves as a legitimate model to explore telomerase regulation in the development of normal and neoplastic tissues in vivo.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Genes, Reporter/genetics , Mammary Neoplasms, Animal/genetics , Telomerase/genetics , Age Factors , Animals , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Silencing , Genetic Complementation Test , Humans , Luciferases/genetics , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Telomerase/metabolism , Wnt1 Protein/metabolism
15.
Nature ; 469(7328): 102-6, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-21113150

ABSTRACT

An ageing world population has fuelled interest in regenerative remedies that may stem declining organ function and maintain fitness. Unanswered is whether elimination of intrinsic instigators driving age-associated degeneration can reverse, as opposed to simply arrest, various afflictions of the aged. Such instigators include progressively damaged genomes. Telomerase-deficient mice have served as a model system to study the adverse cellular and organismal consequences of wide-spread endogenous DNA damage signalling activation in vivo. Telomere loss and uncapping provokes progressive tissue atrophy, stem cell depletion, organ system failure and impaired tissue injury responses. Here, we sought to determine whether entrenched multi-system degeneration in adult mice with severe telomere dysfunction can be halted or possibly reversed by reactivation of endogenous telomerase activity. To this end, we engineered a knock-in allele encoding a 4-hydroxytamoxifen (4-OHT)-inducible telomerase reverse transcriptase-oestrogen receptor (TERT-ER) under transcriptional control of the endogenous TERT promoter. Homozygous TERT-ER mice have short dysfunctional telomeres and sustain increased DNA damage signalling and classical degenerative phenotypes upon successive generational matings and advancing age. Telomerase reactivation in such late generation TERT-ER mice extends telomeres, reduces DNA damage signalling and associated cellular checkpoint responses, allows resumption of proliferation in quiescent cultures, and eliminates degenerative phenotypes across multiple organs including testes, spleens and intestines. Notably, somatic telomerase reactivation reversed neurodegeneration with restoration of proliferating Sox2(+) neural progenitors, Dcx(+) newborn neurons, and Olig2(+) oligodendrocyte populations. Consistent with the integral role of subventricular zone neural progenitors in generation and maintenance of olfactory bulb interneurons, this wave of telomerase-dependent neurogenesis resulted in alleviation of hyposmia and recovery of innate olfactory avoidance responses. Accumulating evidence implicating telomere damage as a driver of age-associated organ decline and disease risk and the marked reversal of systemic degenerative phenotypes in adult mice observed here support the development of regenerative strategies designed to restore telomere integrity.


Subject(s)
Aging/metabolism , Aging/pathology , Telomerase/deficiency , Telomerase/metabolism , Aging/drug effects , Animals , Avoidance Learning/drug effects , Brain/anatomy & histology , Brain/cytology , Brain/drug effects , Brain/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , DNA Damage/drug effects , Doublecortin Protein , Enzyme Activation/drug effects , Enzyme Reactivators/pharmacology , Mice , Mice, Inbred C57BL , Models, Animal , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Organ Size/drug effects , Phenotype , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Regenerative Medicine , Smell/drug effects , Smell/physiology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Telomerase/genetics , Telomere/drug effects , Telomere/metabolism , Telomere/pathology
16.
Nat Biotechnol ; 28(1): 71-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20023657

ABSTRACT

To recapitulate the stochastic nature of human cancer development, we have devised a strategy for generating mouse tumor models that involves stepwise genetic manipulation of embryonic stem (ES) cells and chimera generation. Tumors in the chimeric animals develop from engineered cells in the context of normal tissue. Adenocarcinomas arising in an allelic series of lung cancer models containing HER2 (also known as ERBB2), KRAS or EGFR oncogenes exhibit features of advanced malignancies. Treatment of EGFR(L858R) and KRAS(G12V) chimeric models with an EGFR inhibitor resulted in near complete tumor regression and no response to the treatment, respectively, accurately reflecting previous clinical observations. Transcriptome and immunohistochemical analyses reveal that PI3K pathway activation is unique to ERBB family tumors whereas KRAS-driven tumors show activation of the JNK/SAP pathway, suggesting points of therapeutic intervention for this difficult-to-treat tumor category.


Subject(s)
Adenocarcinoma/metabolism , Chimera/metabolism , ErbB Receptors/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Adenocarcinoma/pathology , Animals , Disease Models, Animal , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Phenotype , Piperazines/pharmacology , Quinazolines/pharmacology , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/pathology , Signal Transduction/drug effects
17.
Cell ; 128(2): 309-23, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17254969

ABSTRACT

Activated phosphoinositide 3-kinase (PI3K)-AKT signaling appears to be an obligate event in the development of cancer. The highly related members of the mammalian FoxO transcription factor family, FoxO1, FoxO3, and FoxO4, represent one of several effector arms of PI3K-AKT signaling, prompting genetic analysis of the role of FoxOs in the neoplastic phenotypes linked to PI3K-AKT activation. While germline or somatic deletion of up to five FoxO alleles produced remarkably modest neoplastic phenotypes, broad somatic deletion of all FoxOs engendered a progressive cancer-prone condition characterized by thymic lymphomas and hemangiomas, demonstrating that the mammalian FoxOs are indeed bona fide tumor suppressors. Transcriptome and promoter analyses of differentially affected endothelium identified direct FoxO targets and revealed that FoxO regulation of these targets in vivo is highly context-specific, even in the same cell type. Functional studies validated Sprouty2 and PBX1, among others, as FoxO-regulated mediators of endothelial cell morphogenesis and vascular homeostasis.


Subject(s)
Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Endothelial Cells/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Transformation, Neoplastic/metabolism , Drosophila Proteins , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Hemangioma/genetics , Hemangioma/metabolism , Homeodomain Proteins/genetics , Homeostasis/genetics , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 103(20): 7817-22, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16672372

ABSTRACT

The tyrosine kinase inhibitors gefitinib (Iressa) and erlotinib (Tarceva) have shown anti-tumor activity in the treatment of non-small cell lung cancer (NSCLC). Dramatic and durable responses have occurred in NSCLC tumors with mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR). In contrast, these inhibitors have shown limited efficacy in glioblastoma, where a distinct EGFR mutation, the variant III (vIII) in-frame deletion of exons 2-7, is commonly found. In this study, we determined that EGFRvIII mutation was present in 5% (3/56) of analyzed human lung squamous cell carcinoma (SCC) but was not present in human lung adenocarcinoma (0/123). We analyzed the role of the EGFRvIII mutation in lung tumorigenesis and its response to tyrosine kinase inhibition. Tissue-specific expression of EGFRvIII in the murine lung led to the development of NSCLC. Most importantly, these lung tumors depend on EGFRvIII expression for maintenance. Treatment with an irreversible EGFR inhibitor, HKI-272, dramatically reduced the size of these EGFRvIII-driven murine tumors in 1 week. Similarly, Ba/F3 cells transformed with the EGFRvIII mutant were relatively resistant to gefitinib and erlotinib in vitro but proved sensitive to HKI-272. These findings suggest a therapeutic strategy for cancers harboring the EGFRvIII mutation.


Subject(s)
ErbB Receptors/metabolism , Lung Neoplasms , Protein Isoforms/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases , Quinazolines/therapeutic use , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Erlotinib Hydrochloride , Gefitinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Knockout , Mutation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Kinase Inhibitors/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Quinazolines/metabolism , Quinolines/metabolism , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p14ARF/metabolism
19.
Genesis ; 44(5): 262-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16676322

ABSTRACT

Conditional Cre-mediated recombination has emerged as a robust method of introducing somatic genetic alterations in an organ-specific manner in the mouse. Here, we generated and characterized mice harboring a 4-hydroxytamoxifen (OHT)-inducible Cre recombinase-estrogen receptor fusion transgene under the control of the melanocyte-specific tyrosinase promoter, designated Tyr::CreER(T2). Cre-mediated recombination was induced in melanocytes in a spatially and temporally controlled manner upon administration of OHT and was documented in embryonic melanoblasts, follicular bulb melanocytes, dermal dendritic melanocytes, epidermal melanocytes of tail skin, and in putative melanocyte stem cells located within the follicular bulge. Functional evidence suggestive of recombination in follicular melanocyte stem cells included the presence of Cre-mediated recombination in follicular bulb melanocytes 1 year after topical OHT administration, by which time several hair cycles have elapsed and the melanocytes residing in this location have undergone multiple rounds of apoptosis and replenishment. These Tyr:: CreER(T2) transgenic mice represent a useful resource for the evaluation of melanocyte developmental genetics, the characterization of melanocyte stem cell function and dynamics, and the construction of refined mouse models of malignant melanoma.


Subject(s)
Gene Expression Regulation , Integrases , Melanocytes/physiology , Mice, Transgenic , Recombination, Genetic , Animals , Central Nervous System , Embryo, Mammalian , Enhancer Elements, Genetic , Female , Integrases/metabolism , Melanocytes/chemistry , Melanocytes/cytology , Mice , Monophenol Monooxygenase/genetics , Pregnancy , Promoter Regions, Genetic , Receptors, Estrogen/genetics , Stem Cells/physiology , Tamoxifen/administration & dosage , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
20.
Proc Natl Acad Sci U S A ; 103(1): 105-10, 2006 Jan 03.
Article in English | MEDLINE | ID: mdl-16373502

ABSTRACT

Many human diseases are characterized by the development of tissue hypoxia. Inadequate oxygenation can cause cellular dysfunction and death. Tissues use many strategies, including induction of angiogenesis and alterations in metabolism, to survive under hypoxic conditions. The heterodimeric transcription factor hypoxia-inducible factor (HIF) is a master regulator of genes that promote adaptation to hypoxia. HIF activity is linked to oxygen availability because members of the EGLN family hydroxylate HIFalpha subunits on specific prolyl residues when oxygen is present, which marks them for ubiquitination and proteasomal degradation. We created a mouse that ubiquitously expresses a bioluminescent reporter consisting of firefly luciferase fused to a region of HIF that is sufficient for oxygen-dependent degradation. Our validation studies suggest that this mouse will be useful for monitoring hypoxic tissues and evaluating therapeutic agents that stabilize HIF. One such agent, the HIF prolyl hydroxylase inhibitor FG-4383, was active in the liver and kidney after systemic administration as determined by bioluminescence imaging, transcription profiling, and production of erythropoietin, indicating that the HIF transcriptional program can be manipulated in vivo with orally active organic small molecules.


Subject(s)
Dioxygenases/antagonists & inhibitors , Dioxygenases/metabolism , Enzyme Inhibitors , Erythropoietin/biosynthesis , Hypoxia-Inducible Factor 1/metabolism , Models, Animal , Animals , Cell Hypoxia/physiology , Cells, Cultured , DNA Primers , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Luciferases , Mice , Molecular Probe Techniques , Plasmids/genetics , Proteins/genetics , Proteins/metabolism , RNA, Untranslated
SELECTION OF CITATIONS
SEARCH DETAIL
...