Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37786677

ABSTRACT

MITF, a basic-Helix-Loop-Helix Zipper (bHLHZip) transcription factor, plays vital roles in melanocyte development and functions as an oncogene. To explore MITF regulation and its role in melanoma, we conducted a genetic screen for suppressors of the Mitf-associated pigmentation phenotype. An intragenic Mitf mutation was identified, leading to termination of MITF at the K316 SUMOylation site and loss of the C-end intrinsically disordered region (IDR). The resulting protein is more nuclear but less stable than wild-type MITF and retains DNA-binding ability. Interestingly, as a dimer, it can translocate wild-type and mutant MITF partners into the nucleus, improving its own stability and ensuring an active nuclear MITF supply. Interactions between K316 SUMOylation and S409 phosphorylation sites across monomers largely explain the observed effects. Notably, the recurrent melanoma-associated E318K mutation in MITF, which affects K316 SUMOylation, also alters protein regulation in concert with S409, unraveling a novel regulatory mechanism with unexpected disease insights.

2.
Sci Transl Med ; 13(604)2021 07 28.
Article in English | MEDLINE | ID: mdl-34321320

ABSTRACT

Accumulation of the parkin-interacting substrate (PARIS; ZNF746), due to inactivation of parkin, contributes to Parkinson's disease (PD) through repression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α; PPARGC1A) activity. Here, we identify farnesol as an inhibitor of PARIS. Farnesol promoted the farnesylation of PARIS, preventing its repression of PGC-1α via decreasing PARIS occupancy on the PPARGC1A promoter. Farnesol prevented dopaminergic neuronal loss and behavioral deficits via farnesylation of PARIS in PARIS transgenic mice, ventral midbrain transduction of AAV-PARIS, adult conditional parkin KO mice, and the α-synuclein preformed fibril model of sporadic PD. PARIS farnesylation is decreased in the substantia nigra of patients with PD, suggesting that reduced farnesylation of PARIS may play a role in PD. Thus, farnesol may be beneficial in the treatment of PD by enhancing the farnesylation of PARIS and restoring PGC-1α activity.


Subject(s)
Parkinson Disease , Animals , Dopamine , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Prenylation , Repressor Proteins/metabolism , Substantia Nigra/metabolism
3.
Cancers (Basel) ; 13(2)2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33435458

ABSTRACT

A central challenge in cancer genomics is the systematic identification of single and cooperating tumor suppressor gene mutations driving cellular transformation and tumor progression in the absence of oncogenic driver mutation(s). Multiple in vitro and in vivo gene inactivation screens have enhanced our understanding of the tumor suppressor gene landscape in various cancers. However, these studies are limited to single or combination gene effects, specific organs, or require sensitizing mutations. In this study, we developed and utilized a Sleeping Beauty transposon mutagenesis system that functions only as a gene trap to exclusively inactivate tumor suppressor genes. Using whole body transposon mobilization in wild type mice, we observed that cumulative gene inactivation can drive tumorigenesis of solid cancers. We provide a quantitative landscape of the tumor suppressor genes inactivated in these cancers and show that, despite the absence of oncogenic drivers, these genes converge on key biological pathways and processes associated with cancer hallmarks.

4.
Nat Commun ; 11(1): 1950, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32327658

ABSTRACT

BDNF signaling in hypothalamic circuitries regulates mammalian food intake. However, whether BDNF exerts metabolic effects on peripheral organs is currently unknown. Here, we show that the BDNF receptor TrkB.T1 is expressed by pancreatic ß-cells where it regulates insulin release. Mice lacking TrkB.T1 show impaired glucose tolerance and insulin secretion. ß-cell BDNF-TrkB.T1 signaling triggers calcium release from intracellular stores, increasing glucose-induced insulin secretion. Additionally, BDNF is secreted by skeletal muscle and muscle-specific BDNF knockout phenocopies the ß-cell TrkB.T1 deletion metabolic impairments. The finding that BDNF is also secreted by differentiated human muscle cells and induces insulin secretion in human islets via TrkB.T1 identifies a new regulatory function of BDNF on metabolism that is independent of CNS activity. Our data suggest that muscle-derived BDNF may be a key factor mediating increased glucose metabolism in response to exercise, with implications for the treatment of diabetes and related metabolic diseases.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Calcium/metabolism , Cells, Cultured , Glucose/metabolism , Glucose Intolerance , Humans , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, trkB/chemistry , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction
6.
Brain ; 142(8): 2380-2401, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31237944

ABSTRACT

α-Synuclein misfolding and aggregation plays a major role in the pathogenesis of Parkinson's disease. Although loss of function mutations in the ubiquitin ligase, parkin, cause autosomal recessive Parkinson's disease, there is evidence that parkin is inactivated in sporadic Parkinson's disease. Whether parkin inactivation is a driver of neurodegeneration in sporadic Parkinson's disease or a mere spectator is unknown. Here we show that parkin in inactivated through c-Abelson kinase phosphorylation of parkin in three α-synuclein-induced models of neurodegeneration. This results in the accumulation of parkin interacting substrate protein (zinc finger protein 746) and aminoacyl tRNA synthetase complex interacting multifunctional protein 2 with increased parkin interacting substrate protein levels playing a critical role in α-synuclein-induced neurodegeneration, since knockout of parkin interacting substrate protein attenuates the degenerative process. Thus, accumulation of parkin interacting substrate protein links parkin inactivation and α-synuclein in a common pathogenic neurodegenerative pathway relevant to both sporadic and familial forms Parkinson's disease. Thus, suppression of parkin interacting substrate protein could be a potential therapeutic strategy to halt the progression of Parkinson's disease and related α-synucleinopathies.


Subject(s)
Parkinson Disease/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/metabolism , Animals , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Parkinson Disease/pathology
7.
Neuron ; 100(4): 816-830.e7, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30344044

ABSTRACT

Through the generation of humanized FUS mice expressing full-length human FUS, we identify that when expressed at near endogenous murine FUS levels, both wild-type and ALS-causing and frontotemporal dementia (FTD)-causing mutations complement the essential function(s) of murine FUS. Replacement of murine FUS with mutant, but not wild-type, human FUS causes stress-mediated induction of chaperones, decreased expression of ion channels and transporters essential for synaptic function, and reduced synaptic activity without loss of nuclear FUS or its cytoplasmic aggregation. Most strikingly, accumulation of mutant human FUS is shown to activate an integrated stress response and to inhibit local, intra-axonal protein synthesis in hippocampal neurons and sciatic nerves. Collectively, our evidence demonstrates that human ALS/FTD-linked mutations in FUS induce a gain of toxicity that includes stress-mediated suppression in intra-axonal translation, synaptic dysfunction, and progressive age-dependent motor and cognitive disease without cytoplasmic aggregation, altered nuclear localization, or aberrant splicing of FUS-bound pre-mRNAs. VIDEO ABSTRACT.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Axons/physiology , Frontotemporal Dementia/genetics , Loss of Function Mutation/genetics , Protein Biosynthesis/physiology , RNA-Binding Protein FUS/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/pathology , Cells, Cultured , Female , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , RNA-Binding Protein FUS/biosynthesis
8.
J Clin Invest ; 128(7): 2927-2943, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29863500

ABSTRACT

Although nonmalignant stromal cells facilitate tumor growth and can occupy up to 90% of a solid tumor mass, better strategies to exploit these cells for improved cancer therapy are needed. Here, we describe a potent MMAE-linked antibody-drug conjugate (ADC) targeting tumor endothelial marker 8 (TEM8, also known as ANTXR1), a highly conserved transmembrane receptor broadly overexpressed on cancer-associated fibroblasts, endothelium, and pericytes. Anti-TEM8 ADC elicited potent anticancer activity through an unexpected killing mechanism we term DAaRTS (drug activation and release through stroma), whereby the tumor microenvironment localizes active drug at the tumor site. Following capture of ADC prodrug from the circulation, tumor-associated stromal cells release active MMAE free drug, killing nearby proliferating tumor cells in a target-independent manner. In preclinical studies, ADC treatment was well tolerated and induced regression and often eradication of multiple solid tumor types, blocked metastatic growth, and prolonged overall survival. By exploiting TEM8+ tumor stroma for targeted drug activation, these studies reveal a drug delivery strategy with potential to augment therapies against multiple cancer types.


Subject(s)
Immunoconjugates/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Brentuximab Vedotin , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, SCID , Microfilament Proteins , Neoplasms/metabolism , Receptors, Peptide/antagonists & inhibitors , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Stromal Cells/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 115(7): 1635-1640, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29386392

ABSTRACT

Mutations in LRRK2 are known to be the most common genetic cause of sporadic and familial Parkinson's disease (PD). Multiple lines of LRRK2 transgenic or knockin mice have been developed, yet none exhibit substantial dopamine (DA)-neuron degeneration. Here we develop human tyrosine hydroxylase (TH) promoter-controlled tetracycline-sensitive LRRK2 G2019S (GS) and LRRK2 G2019S kinase-dead (GS/DA) transgenic mice and show that LRRK2 GS expression leads to an age- and kinase-dependent cell-autonomous neurodegeneration of DA and norepinephrine (NE) neurons. Accompanying the loss of DA neurons are DA-dependent behavioral deficits and α-synuclein pathology that are also LRRK2 GS kinase-dependent. Transmission EM reveals that that there is an LRRK2 GS kinase-dependent significant reduction in synaptic vesicle number and a greater abundance of clathrin-coated vesicles in DA neurons. These transgenic mice indicate that LRRK2-induced DA and NE neurodegeneration is kinase-dependent and can occur in a cell-autonomous manner. Moreover, these mice provide a substantial advance in animal model development for LRRK2-associated PD and an important platform to investigate molecular mechanisms for how DA neurons degenerate as a result of expression of mutant LRRK2.


Subject(s)
Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/physiology , Neurodegenerative Diseases/pathology , Norepinephrine/metabolism , Age Factors , Animals , Behavior, Animal , Dopaminergic Neurons/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Activity , Mutation , Neurodegenerative Diseases/metabolism , alpha-Synuclein/metabolism
10.
Genetics ; 207(4): 1335-1345, 2017 12.
Article in English | MEDLINE | ID: mdl-29021281

ABSTRACT

BRCA2 loss-of-heterozygosity (LOH) is frequently observed in BRCA2-mutated tumors, but its biallelic loss causes embryonic lethality in mice and inhibits proliferation of normal somatic cells. Therefore, it remains unclear how loss of BRCA2 contributes to tumorigenesis. One possibility is that mutation in potential genetic interactors of BRCA2, such as TRP53, is required for cell survival/proliferation in the absence of BRCA2. In this study, using an insertional mutagenesis screen in mouse embryonic stem cells (mESC), we have identified GIPC3 (GAIP-interacting protein C-terminus 3) as a BRCA2 genetic interactor that contributes to survival of Brca2-null mESC. GIPC3 does not compensate for BRCA2 loss in the repair of double-strand breaks. Mass-spectrometric analysis resulted in the identification of G-protein signaling transducers, APPL1 and APPL2, as potential GIPC3-binding proteins. A mutant GIPC3 (His155Ala) that does not bind to APPL1/2 failed to rescue the lethality of Brca2-null mESC, suggesting that the cell viability by GIPC3 is mediated via APPL1/2. Finally, the physiological significance of GIPC3 as a genetic interactor of BRCA2 is supported by the observation that Brca2-null embryos with Gipc3 overexpression are developmentally more advanced than their control littermates. Taken together, we have uncovered a novel role for GIPC3 as a BRCA2 genetic interactor.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Carcinogenesis/genetics , Animals , BRCA2 Protein/deficiency , Breast Neoplasms/pathology , Carrier Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Loss of Heterozygosity/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , Mutagenesis, Insertional , Mutation
11.
Cancer Cell ; 31(4): 501-515.e8, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399408

ABSTRACT

Targeting the tumor vasculature with antibody-drug conjugates (ADCs) is a promising anti-cancer strategy that in order to be realized must overcome several obstacles, including identification of suitable targets and optimal warheads. Here, we demonstrate that the cell-surface protein CD276/B7-H3 is broadly overexpressed by multiple tumor types on both cancer cells and tumor-infiltrating blood vessels, making it a potentially ideal dual-compartment therapeutic target. In preclinical studies CD276 ADCs armed with a conventional MMAE warhead destroyed CD276-positive cancer cells, but were ineffective against tumor vasculature. In contrast, pyrrolobenzodiazepine-conjugated CD276 ADCs killed both cancer cells and tumor vasculature, eradicating large established tumors and metastases, and improving long-term overall survival. CD276-targeted dual-compartment ablation could aid in the development of highly selective broad-acting anti-cancer therapies.


Subject(s)
B7 Antigens/genetics , B7 Antigens/metabolism , Immunoconjugates/pharmacology , Neoplasms/blood supply , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , B7 Antigens/immunology , Benzodiazepines/pharmacology , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Line, Tumor , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Immunoconjugates/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy/methods , Neoplasms/pathology , Neoplasms/therapy , Oligopeptides/pharmacology , Pyrroles/pharmacology , Rabbits
12.
Cell Rep ; 16(3): 793-804, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27373150

ABSTRACT

The neural network of the temporal lobe is thought to provide a cognitive map of our surroundings. Functional analysis of this network has been hampered by coarse tools that often result in collateral damage to other circuits. We developed a chemogenetic system to temporally control electrical input into the hippocampus. When entorhinal input to the perforant path was acutely silenced, hippocampal firing patterns became destabilized and underwent extensive remapping. We also found that spatial memory acquired prior to neural silencing was impaired by loss of input through the perforant path. Together, our experiments show that manipulation of entorhinal activity destabilizes spatial coding and disrupts spatial memory. Moreover, we introduce a chemogenetic model for non-invasive neuronal silencing that offers multiple advantages over existing strategies in this setting.


Subject(s)
Hippocampus/physiology , Nerve Net/physiology , Spatial Memory/physiology , Temporal Lobe/physiology , Animals , Entorhinal Cortex/physiology , Female , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Models, Neurological , Perforant Pathway/physiology
13.
Neuron ; 90(3): 535-50, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27112497

ABSTRACT

Hexanucleotide expansions in C9ORF72 are the most frequent genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Disease mechanisms were evaluated in mice expressing C9ORF72 RNAs with up to 450 GGGGCC repeats or with one or both C9orf72 alleles inactivated. Chronic 50% reduction of C9ORF72 did not provoke disease, while its absence produced splenomegaly, enlarged lymph nodes, and mild social interaction deficits, but not motor dysfunction. Hexanucleotide expansions caused age-, repeat-length-, and expression-level-dependent accumulation of RNA foci and dipeptide-repeat proteins synthesized by AUG-independent translation, accompanied by loss of hippocampal neurons, increased anxiety, and impaired cognitive function. Single-dose injection of antisense oligonucleotides (ASOs) that target repeat-containing RNAs but preserve levels of mRNAs encoding C9ORF72 produced sustained reductions in RNA foci and dipeptide-repeat proteins, and ameliorated behavioral deficits. These efforts identify gain of toxicity as a central disease mechanism caused by repeat-expanded C9ORF72 and establish the feasibility of ASO-mediated therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Frontotemporal Dementia/drug therapy , Guanine Nucleotide Exchange Factors/genetics , Oligonucleotides, Antisense/pharmacology , RNA/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , C9orf72 Protein , DNA Repeat Expansion/genetics , Frontotemporal Dementia/genetics , Mice, Transgenic , Neurons/metabolism , Oligonucleotides, Antisense/adverse effects , Oligonucleotides, Antisense/genetics
14.
Proc Natl Acad Sci U S A ; 112(46): E6321-30, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578792

ABSTRACT

Centrosomes are microtubule-organizing centers that facilitate bipolar mitotic spindle assembly and chromosome segregation. Recognizing that centrosome amplification is a common feature of aneuploid cancer cells, we tested whether supernumerary centrosomes are sufficient to drive tumor development. To do this, we constructed and analyzed mice in which centrosome amplification can be induced by a Cre-recombinase-mediated increase in expression of Polo-like kinase 4 (Plk4). Elevated Plk4 in mouse fibroblasts produced supernumerary centrosomes and enhanced the expected mitotic errors, but proliferation continued only after inactivation of the p53 tumor suppressor. Increasing Plk4 levels in mice with functional p53 produced centrosome amplification in liver and skin, but this did not promote spontaneous tumor development in these tissues or enhance the growth of chemically induced skin tumors. In the absence of p53, Plk4 overexpression generated widespread centrosome amplification, but did not drive additional tumors or affect development of the fatal thymic lymphomas that arise in animals lacking p53. We conclude that, independent of p53 status, supernumerary centrosomes are not sufficient to drive tumor formation.


Subject(s)
Asymmetric Cell Division/physiology , Centrosome/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Dev Cell ; 35(3): 322-32, 2015 Nov 09.
Article in English | MEDLINE | ID: mdl-26555052

ABSTRACT

The mammalian lung forms its elaborate tree-like structure following a largely stereotypical branching sequence. While a number of genes have been identified to play essential roles in lung branching, what coordinates the choice between branch growth and new branch formation has not been elucidated. Here we show that loss of FGF-activated transcription factor genes, Etv4 and Etv5 (collectively Etv), led to prolonged branch tip growth and delayed new branch formation. Unexpectedly, this phenotype is more similar to mutants with increased rather than decreased FGF activity. Indeed, an increased Fgf10 expression is observed, and reducing Fgf10 dosage can attenuate the Etv mutant phenotype. Further evidence indicates that ETV inhibits Fgf10 via directly promoting Shh expression. SHH in turn inhibits local Fgf10 expression and redirects growth, thereby initiating new branches. Together, our findings establish ETV as a key node in the FGF-ETV-SHH inhibitory feedback loop that dictates branching periodicity.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Lung/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Animals , Body Patterning/genetics , Fibroblast Growth Factor 10/metabolism , Hedgehog Proteins/metabolism , Mice , Mice, Transgenic , Morphogenesis/genetics , Signal Transduction/genetics
16.
J Mol Cell Cardiol ; 88: 1-13, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26386426

ABSTRACT

Gap junctions (GJ) are intercellular channels composed of connexin subunits that play a critical role in a diverse number of cellular processes in all tissue types. In the heart, GJs mediate electrical coupling between cardiomyocytes and display mislocalization and/or downregulation in cardiac disease (a process known as GJ remodeling), producing an arrhythmogenic substrate. The main constituent of GJs in the ventricular myocardium is Connexin 43 (Cx43), an integral membrane protein that is rapidly turned over and shows decreased expression or function with age. We hypothesized that Wwp1, an ubiquitin ligase whose expression in known to increase in aging-related pathologies, may regulate Cx43 in vivo by targeting it for ubiquitylation and degradation and yield tissue-specific Cx43 loss of function phenotypes. When Wwp1 was globally overexpressed in mice under the control of a ß-actin promoter, the highest induction of Wwp1 expression was observed in the heart which was associated with a 90% reduction in cardiac Cx43 protein levels, left ventricular hypertrophy (LVH), and the development of lethal ventricular arrhythmias around 8weeks of age. This phenotype was completely penetrant in two independent founder lines. Cardiomyocyte-specific overexpression of Wwp1 confirmed that this phenotype was cell autonomous and delineated Cx43-dependent and -independent roles for Wwp1 in arrhythmogenesis and LVH, respectively. Using a cell-based system, it was determined that Wwp1 co-immunoprecipitates with and ubiquitylates Cx43, causing a decrease in the steady state levels of Cx43 protein. These findings offer new mechanistic insights into the regulation of Cx43 which may be exploitable in various gap junctionopathies.


Subject(s)
Arrhythmias, Cardiac/genetics , Connexin 43/genetics , Hypertrophy, Left Ventricular/genetics , Myocytes, Cardiac/metabolism , Ubiquitin-Protein Ligases/genetics , Actins/genetics , Actins/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Connexin 43/metabolism , Disease Models, Animal , Female , Gap Junctions/metabolism , Gap Junctions/pathology , Gene Expression Regulation , Heart Ventricles/metabolism , Heart Ventricles/pathology , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Male , Mice , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , Phenotype , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
17.
Cell Rep ; 10(2): 123-30, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25558062

ABSTRACT

G protein-coupled receptor 124 (GPR124) is an orphan receptor in the adhesion family of GPCRs, and previous global or endothelial-specific disruption of Gpr124 in mice led to defective CNS angiogenesis and blood-brain barriergenesis. Similar developmental defects were observed following dual deletion of Wnt7a/Wnt7b or deletion of ß-catenin in endothelial cells, suggesting a possible relationship between GPR124 and canonical WNT signaling. Here, we show using in vitro reporter assays, mutation analysis, and genetic interaction studies in vivo that GPR124 functions as a WNT7A/WNT7B-specific costimulator of ß-catenin signaling in brain endothelium. WNT7-stimulated ß-catenin signaling was dependent upon GPR124's intracellular PDZ binding motif and a set of leucine-rich repeats in its extracellular domain. This study reveals a vital role for GPR124 in potentiation of WNT7-induced canonical ß-catenin signaling with important implications for understanding and manipulating CNS-specific angiogenesis and blood-brain barrier-genesis.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Amino Acid Motifs , Animals , Blood-Brain Barrier/metabolism , Brain/cytology , Brain/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Mice, Transgenic , PDZ Domains , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/deficiency
18.
Sci Transl Med ; 6(242): 242ra84, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24964992

ABSTRACT

Antiangiogenic agents that block vascular endothelial growth factor (VEGF) signaling are important components of current cancer treatment modalities but are limited by alternative ill-defined angiogenesis mechanisms that allow persistent tumor vascularization in the face of continued VEGF pathway blockade. We identified prostaglandin E2 (PGE2) as a soluble tumor-derived angiogenic factor associated with VEGF-independent angiogenesis. PGE2 production in preclinical breast and colon cancer models was tightly controlled by cyclooxygenase-2 (COX-2) expression, and COX-2 inhibition augmented VEGF pathway blockade to suppress angiogenesis and tumor growth, prevent metastasis, and increase overall survival. These results demonstrate the importance of the COX-2/PGE2 pathway in mediating resistance to VEGF pathway blockade and could aid in the rapid development of more efficacious anticancer therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Mammary Neoplasms, Experimental/prevention & control , Mammary Neoplasms, Experimental/secondary , Xenograft Model Antitumor Assays , Angiogenesis Inhibitors/pharmacology , Animals , Axitinib , Carcinogenesis/pathology , Cell Line, Tumor , Clone Cells , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/metabolism , Female , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , Liver Neoplasms/blood supply , Liver Neoplasms/drug therapy , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mice , Neoadjuvant Therapy , Signal Transduction/drug effects , Survival Analysis , Vascular Endothelial Growth Factor A/metabolism
19.
PLoS One ; 9(1): e85883, 2014.
Article in English | MEDLINE | ID: mdl-24465765

ABSTRACT

The LMO2 oncogene is deregulated in the majority of human T-cell leukemia cases and in most gene therapy-induced T-cell leukemias. We made transgenic mice with enforced expression of Lmo2 in T-cells by the CD2 promoter/enhancer. These transgenic mice developed highly penetrant T-ALL by two distinct patterns of gene expression: one in which there was concordant activation of Lyl1, Hhex, and Mycn or alternatively, with Notch1 target gene activation. Most strikingly, this gene expression clustering was conserved in human Early T-cell Precursor ALL (ETP-ALL), where LMO2, HHEX, LYL1, and MYCN were most highly expressed. We discovered that HHEX is a direct transcriptional target of LMO2 consistent with its concordant gene expression. Furthermore, conditional inactivation of Hhex in CD2-Lmo2 transgenic mice markedly attenuated T-ALL development, demonstrating that Hhex is a crucial mediator of Lmo2's oncogenic function. The CD2-Lmo2 transgenic mice offer mechanistic insight into concordant oncogene expression and provide a model for the highly treatment-resistant ETP-ALL subtype.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/metabolism , LIM Domain Proteins/metabolism , Leukemia, T-Cell/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD2 Antigens/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , E-Box Elements/genetics , Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , Humans , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Mice , Mice, Transgenic , Molecular Sequence Data , Neoplasm Proteins/metabolism , Oncogenes , Penetrance , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Promoter Regions, Genetic/genetics , Protein Binding , Transcription Factors/genetics , Transcription, Genetic , Up-Regulation/genetics
20.
PLoS One ; 8(4): e62479, 2013.
Article in English | MEDLINE | ID: mdl-23638095

ABSTRACT

The study of axis extension and somitogenesis has been greatly advanced through the use of genetic tools such as the TCre mouse line. In this line, Cre is controlled by a fragment of the T (Brachyury) promoter that is active in progenitor cells that reside within the primitive streak and tail bud and which give rise to lineages emerging from these tissues as the embryonic axis extends. However, because TCre-mediated recombination occurs early in development, gene inactivation can result in an axis truncation that precludes the study of gene function in later or more posterior tissues. To address this limitation, we have generated an inducible TCre transgenic mouse line, called TCreERT2, that provides temporal control, through tamoxifen administration, in all cells emerging from the primitive streak or tail bud throughout development. TCreERT2 activity is mostly silent in the absence of tamoxifen and, in its presence, results in near complete recombination of emerging mesoderm from E7.5 through E13.5. We demonstrate the utility of the TCreERT2 line for determining rate of posterior axis extension and somite formation, thus providing the first in vivo tool for such measurements. To test the usefulness of TCreERT2 for genetic manipulation, we demonstrate that an early deletion of ß-Catenin via TCreERT2 induction phenocopies the TCre-mediated deletion of ß-Catenin defect, whereas a later induction bypasses this early phenotype and produces a similar defect in more caudal tissues. TCreERT2 provides a useful and novel tool for the control of gene expression of emerging embryonic lineages throughout development.


Subject(s)
Mice, Transgenic/genetics , Primitive Streak/embryology , Recombination, Genetic , Animals , Estrogen Antagonists/administration & dosage , Female , Fetal Proteins/genetics , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Primitive Streak/cytology , Primitive Streak/metabolism , T-Box Domain Proteins/genetics , Tamoxifen/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...