Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 30(5): 1038-1053, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38127282

ABSTRACT

PURPOSE: Plexiform neurofibromas (PNF) are benign peripheral nerve sheath tumors (PNST) associated with neurofibromatosis type 1 (NF1). Despite similar histologic appearance, these neoplasms exhibit diverse evolutionary trajectories, with a subset progressing to malignant peripheral nerve sheath tumor (MPNST), the leading cause of premature death in individuals with NF1. Malignant transformation of PNF often occurs through the development of atypical neurofibroma (ANF) precursor lesions characterized by distinct histopathologic features and CDKN2A copy-number loss. Although genomic studies have uncovered key driver events promoting tumor progression, the transcriptional changes preceding malignant transformation remain poorly defined. EXPERIMENTAL DESIGN: Here we resolve gene-expression profiles in PNST across the neurofibroma-to-MPNST continuum in NF1 patients and mouse models, revealing early molecular features associated with neurofibroma evolution and transformation. RESULTS: Our findings demonstrate that ANF exhibit enhanced signatures of antigen presentation and immune response, which are suppressed as malignant transformation ensues. MPNST further displayed deregulated survival and mitotic fidelity pathways, and targeting key mediators of these pathways, CENPF and BIRC5, disrupted the growth and viability of human MPNST cell lines and primary murine Nf1-Cdkn2a-mutant Schwann cell precursors. Finally, neurofibromas contiguous with MPNST manifested distinct alterations in core oncogenic and immune surveillance programs, suggesting that early molecular events driving disease progression may precede histopathologic evidence of malignancy. CONCLUSIONS: If validated prospectively in future studies, these signatures may serve as molecular diagnostic tools to augment conventional histopathologic diagnosis by identifying neurofibromas at high risk of undergoing malignant transformation, facilitating risk-adapted care.


Subject(s)
Nerve Sheath Neoplasms , Neurofibroma , Neurofibromatosis 1 , Neurofibrosarcoma , Animals , Humans , Mice , Gene Expression Profiling , Nerve Sheath Neoplasms/genetics , Neurofibroma/genetics , Neurofibromatosis 1/genetics , Neurofibrosarcoma/genetics
2.
Hum Mol Genet ; 28(16): 2752-2762, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31091306

ABSTRACT

Plexiform neurofibroma (PN) tumors are a hallmark manifestation of neurofibromatosis type 1 (NF1) that arise in the Schwann cell (SC) lineage. NF1 is a common heritable cancer predisposition syndrome caused by germline mutations in the NF1 tumor suppressor, which encodes a GTPase-activating protein called neurofibromin that negatively regulates Ras proteins. Whereas most PN are clinically indolent, a subset progress to atypical neurofibromatous neoplasms of uncertain biologic potential (ANNUBP) and/or to malignant peripheral nerve sheath tumors (MPNSTs). In small clinical series, loss of 9p21.3, which includes the CDKN2A locus, has been associated with the genesis of ANNUBP. Here we show that the Cdkn2a alternate reading frame (Arf) serves as a gatekeeper tumor suppressor in mice that prevents PN progression by inducing senescence-mediated growth arrest in aberrantly proliferating Nf1-/- SC. Conditional ablation of Nf1 and Arf in the neural crest-derived SC lineage allows escape from senescence, resulting in tumors that accurately phenocopy human ANNUBP and progress to MPNST with high penetrance. This animal model will serve as a platform to study the clonal development of ANNUBP and MPNST and to identify new therapies to treat existing tumors and to prevent disease progression.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Neurofibroma/genetics , Neurofibroma/pathology , Neurofibromatosis 1/genetics , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/genetics , Disease Models, Animal , Disease Progression , Genotype , Heterografts , Humans , Immunohistochemistry , Mice , Mutation , Nerve Sheath Neoplasms/genetics , Nerve Sheath Neoplasms/metabolism , Nerve Sheath Neoplasms/pathology , Neurofibroma/metabolism , Neurofibroma/mortality , Neurofibromatosis 1/metabolism , Schwann Cells/metabolism , Schwann Cells/pathology , ras Proteins/metabolism
3.
Arterioscler Thromb Vasc Biol ; 37(1): 75-83, 2017 01.
Article in English | MEDLINE | ID: mdl-27856454

ABSTRACT

OBJECTIVE: Nucleotide P2Y2 receptor (P2Y2R) contributes to vascular inflammation by increasing vascular cell adhesion molecule-1 expression in endothelial cells (EC), and global P2Y2R deficiency prevents fatty streak formation in apolipoprotein E null (ApoE-/-) mice. Because P2Y2R is ubiquitously expressed in vascular cells, we investigated the contribution of endothelial P2Y2R in the pathogenesis of atherosclerosis. APPROACH AND RESULTS: EC-specific P2Y2R-deficient mice were generated by breeding VEcadherin5-Cre mice with the P2Y2R floxed mice. Endothelial P2Y2R deficiency reduced endothelial nitric oxide synthase activity and significantly altered ATP- and UTP (uridine 5'-triphosphate)-induced vasorelaxation without affecting vasodilatory responses to acetylcholine. Telemetric blood pressure and echocardiography measurements indicated that EC-specific P2Y2R-deficient mice did not develop hypertension. We investigated the role of endothelial P2Y2R in the development of atherosclerotic lesions by crossing the EC-specific P2Y2R knockout mice onto an ApoE-/- background and evaluated lesion development after feeding a standard chow diet for 25 weeks. Histopathologic examination demonstrated reduced atherosclerotic lesions in the aortic sinus and entire aorta, decreased macrophage infiltration, and increased smooth muscle cell and collagen content, leading to the formation of a subendothelial fibrous cap in EC-specific P2Y2R-deficient ApoE-/- mice. Expression and proteolytic activity of matrix metalloproteinase-2 was significantly reduced in atherosclerotic lesions from EC-specific P2Y2R-deficient ApoE-/- mice. Furthermore, EC-specific P2Y2R deficiency inhibited nitric oxide production, leading to significant increase in smooth muscle cell migration out of aortic explants. CONCLUSIONS: EC-specific P2Y2R deficiency reduces atherosclerotic burden and promotes plaque stability in ApoE-/- mice through impaired macrophage infiltration acting together with reduced matrix metalloproteinase-2 activity and increased smooth muscle cell migration.


Subject(s)
Aorta, Thoracic/metabolism , Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Endothelial Cells/metabolism , Plaque, Atherosclerotic , Receptors, Purinergic P2Y2/deficiency , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Aorta, Thoracic/physiopathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cell Movement , Cells, Cultured , Collagen/metabolism , Disease Models, Animal , Disease Progression , Endothelial Cells/drug effects , Endothelial Cells/pathology , Fibrosis , Macrophages/metabolism , Macrophages/pathology , Male , Matrix Metalloproteinase 2/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y2/genetics , Rupture, Spontaneous , Signal Transduction , Vasodilation/drug effects , Vasodilator Agents/pharmacology
4.
Atherosclerosis ; 257: 38-46, 2017 02.
Article in English | MEDLINE | ID: mdl-28038380

ABSTRACT

BACKGROUND AND AIMS: Mutations in the 5'-nucleotidase ecto (NT5E) gene that encodes CD73, a nucleotidase that converts AMP to adenosine, are linked to arterial calcification. However, the role of purinergic receptor signaling in the pathology of intimal calcification is not well understood. In this study, we examined whether extracellular nucleotides acting via P2Y2 receptor (P2Y2R) modulate arterial intimal calcification, a condition highly correlated with cardiovascular morbidity. METHODS: Apolipoprotein E, P2Y2R double knockout mice (ApoE-/-P2Y2R-/-) were used to determine the effect of P2Y2R deficiency on vascular calcification in vivo. Vascular smooth muscle cells (VSMC) isolated from P2Y2R-/- mice grown in high phosphate medium were used to assess the role of P2Y2R in the conversion of VSMC into osteoblasts. Luciferase-reporter assays were used to assess the effect of P2Y2R on the transcriptional activity of Runx2. RESULTS: P2Y2R deficiency in ApoE-/- mice caused extensive intimal calcification despite a significant reduction in atherosclerosis and macrophage plaque content. The ectoenzyme apyrase that degrades nucleoside di- and triphosphates accelerated high phosphate-induced calcium deposition in cultured VSMC. Expression of P2Y2R inhibits calcification in vitro inhibited the osteoblastic trans-differentiation of VSMC. Mechanistically, expression of P2Y2R inhibited Runx2 transcriptional activation of an osteocalcin promoter driven luciferase reporter gene. CONCLUSIONS: This study reveals a role for vascular P2Y2R as an inhibitor of arterial intimal calcification and provides a new mechanistic insight into the regulation of the osteoblastic trans-differentiation of SMC through P2Y2R-mediated Runx2 antagonism. Given that calcification of atherosclerotic lesions is a significant clinical problem, activating P2Y2R may be an effective therapeutic approach for treatment or prevention of vascular calcification.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Purinergic P2Y2/metabolism , Vascular Calcification/prevention & control , 5'-Nucleotidase/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Transdifferentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , GPI-Linked Proteins/metabolism , Genetic Predisposition to Disease , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , Phenotype , Promoter Regions, Genetic , Receptors, Purinergic P2Y2/deficiency , Receptors, Purinergic P2Y2/genetics , Transfection , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
5.
J Vasc Res ; 53(3-4): 163-171, 2016.
Article in English | MEDLINE | ID: mdl-27723650

ABSTRACT

OBJECTIVES: Extracellular nucleotide release at the site of arterial injury mediates the proliferation and migration of vascular smooth muscle cells. Our aim was to investigate the role of the P2Y2 nucleotide receptor (P2Y2R) in neointimal hyperplasia. Approach and Results: Vascular injury was induced by the implantation of a polyethylene cuff around the femoral artery in wild-type and P2Y2R-deficient mice (P2Y2R-/-). Electron microscopy was used to analyze monocyte and lymphocyte influx to the intima 36 h after injury. Compared to wild-type littermates, P2Y2R-/- mice exhibited a 3-fold decreased number of mononuclear leukocytes invading the intima (p < 0.05). Concomitantly, the migration of smooth muscle cells was decreased by more than 60% (p < 0.05), resulting in a sharp inhibition of intimal thickening formation in P2Y2R-/- mice (n = 15) 14 days after cuff placement. In vitro, loss of P2Y2R significantly impaired monocyte migration in response to nucleotide agonists. Furthermore, transgenic rats overexpressing the P2Y2R developed accelerated intimal lesions resulting in more than 95% luminal stenosis (p < 0.05, n = 10). CONCLUSIONS: Loss- and gain-of-function approaches established direct evidence for P2Y2R involvement in neointimal hyperplasia. Specific anti-P2Y2R therapies may be used against restenosis and bypass graft failure.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Receptors, Purinergic P2Y2/metabolism , Vascular System Injuries/metabolism , Animals , Cells, Cultured , Chemotaxis, Leukocyte , Constriction, Pathologic , Disease Models, Animal , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/ultrastructure , Genetic Predisposition to Disease , Hyperplasia , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/ultrastructure , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/ultrastructure , Phenotype , Purinergic P2Y Receptor Agonists/pharmacology , Rats, Sprague-Dawley , Rats, Transgenic , Receptors, Purinergic P2Y2/deficiency , Receptors, Purinergic P2Y2/drug effects , Receptors, Purinergic P2Y2/genetics , Time Factors , Vascular System Injuries/genetics , Vascular System Injuries/pathology , Vascular System Injuries/prevention & control
6.
Vascul Pharmacol ; 85: 11-20, 2016 10.
Article in English | MEDLINE | ID: mdl-27355755

ABSTRACT

BACKGROUND: Lymphotoxin alpha (LTα) is expressed in human atherosclerotic lesions and genetic variations in the LTα pathway have been linked to myocardial infarction. Activation of the P2Y2 nucleotide receptor (P2Y2R) regulates the production of LTα. in vitro. We aimed to uncover a potential pathway linking purinergic receptor to LTα-mediated inflammatory processes pivotal to the early stages of atherosclerosis in apolipoprotein E (ApoE(-)(/)(-)) deficient mice. METHODS AND RESULTS: En face immunostaining revealed that P2Y2R and VCAM-1 are preferentially expressed in the atherosclerosis prone site of the mouse aortic sinus. Deletion of the P2Y2R gene suppresses VCAM-1 expression. Compared with ApoE(-)(/)(-) mice, ApoE(-)(/)(-) mice lacking the P2Y2R gene (ApoE(-)(/)(-)/P2Y2R(-)(/)(-)) did not develop fatty streak lesions when fed a standard chow diet for 15weeks. Systemic and CD4(+) T cell production of the pro-inflammatory cytokine lymphotoxin-alpha (LTα) were specifically inhibited in ApoE(-)(/)(-)/P2Y2R(-)(/)(-)mice. Anti-LTα preventive treatment was initiated in ApoE(-)(/)(-)mice with intraperitoneal administration of recombinant human tumor necrosis factor receptor 1 fusion protein (TNFR1-Fc) on 5 consecutive days before the disease onset. Remarkably, none of the TNFR1:Fc-treated ApoE(-)(/)(-)mice exhibited atherosclerotic lesions at any developmental stage. SIGNIFICANCE: ApoE(-)(/)(-) mice deficient in P2Y2R exhibit low endothelial cell VCAM-1 levels, decreased production of LTα and delayed onset of atherosclerosis. These data suggest that targeting this nucleotide receptor could be an effective therapeutic approach in atherosclerosis.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/pathology , Lymphotoxin-alpha/genetics , Receptors, Purinergic P2Y2/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Endothelial Cells/metabolism , Humans , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/administration & dosage , Receptors, Tumor Necrosis Factor, Type I/pharmacology , Vascular Cell Adhesion Molecule-1/genetics
7.
Biochem J ; 375(Pt 3): 673-80, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12908872

ABSTRACT

Diacylglycerol kinases (DAGKs) catalyse ATP-dependent phosphorylation of sn-1,2-diacylglycerol that arises during stimulated phosphatidylinositol turnover. DAGKa is activated in vitro by Ca2+ and by acidic phospholipids. The regulatory region of DAGKa includes an N-terminal RVH motif and EF hands that mediate Ca2+-dependent activation. DAGKa also contains tandem C1 protein kinase C homology domains. We utilized yeast, Saccharomyces cerevisiae, which lacks an endogenous DAGK, to express DAGKa and to determine the enzymic activities of different mutant forms of pig DAGKa in vitro. Six aspartate residues conserved in all DAGKs were individually examined by site-directed mutagenesis. Five of these aspartate residues reside in conserved blocks that correspond to sequences in the catalytic site of phosphofructokinases. Mutation of D434 (Asp434) or D650 abolished all DAGKa activity, whereas substitution of one among D465, D497, D529 and D697 decreased the activity to 6% or less of that for wild-type DAGKa. Roles of homologous residues in phosphofructokinases suggested that the N-terminal half of the DAGK catalytic domain binds Mg-ATP and the C-terminal half binds diacylglycerol. A DAGKa mutant with its entire regulatory region deleted showed a much decreased activity that was not activated by Ca2+, but still exhibited PS (phosphatidylserine)-dependent activation. Moreover, mutations of aspartate residues at the catalytic domain had differential effects on activation by Ca2+ and PS. These results indicate that Ca2+ and PS stimulate DAGKa via distinct mechanisms.


Subject(s)
Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Amino Acid Sequence , Animals , Aspartic Acid/genetics , Binding Sites/genetics , COS Cells , Calcium/pharmacology , Catalytic Domain/genetics , Chlorocebus aethiops , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Phosphatidylserines/pharmacology , Saccharomyces cerevisiae/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Swine
SELECTION OF CITATIONS
SEARCH DETAIL