Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Cell Biol ; 24(1): 35-50, 2022 01.
Article in English | MEDLINE | ID: mdl-35027734

ABSTRACT

Vascular endothelial growth factor receptor type 2 (VEGFR2, also known as KDR and FLK1) signalling in endothelial cells (ECs) is essential for developmental and reparative angiogenesis. Reactive oxygen species and copper (Cu) are also involved in these processes. However, their inter-relationship is poorly understood. Evidence of the role of the endothelial Cu importer CTR1 (also known as SLC31A1) in VEGFR2 signalling and angiogenesis in vivo is lacking. Here, we show that CTR1 functions as a redox sensor to promote angiogenesis in ECs. CTR1-depleted ECs showed reduced VEGF-induced VEGFR2 signalling and angiogenic responses. Mechanistically, CTR1 was rapidly sulfenylated at Cys189 at its cytosolic C terminus after stimulation with VEGF, which induced CTR1-VEGFR2 disulfide bond formation and their co-internalization to early endosomes, driving sustained VEGFR2 signalling. In vivo, EC-specific Ctr1-deficient mice or CRISPR-Cas9-generated redox-dead Ctr1(C187A)-knockin mutant mice had impaired developmental and reparative angiogenesis. Thus, oxidation of CTR1 at Cys189 promotes VEGFR2 internalization and signalling to enhance angiogenesis. Our study uncovers an important mechanism for sensing reactive oxygen species through CTR1 to drive neovascularization.


Subject(s)
Copper Transporter 1/metabolism , Copper/metabolism , Neovascularization, Physiologic/physiology , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cattle , Cell Line , Copper Transporter 1/genetics , Cysteine/metabolism , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Signal Transduction/physiology
2.
Sci Rep ; 11(1): 24345, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934133

ABSTRACT

Mutations in the NF1 tumor suppressor gene are linked to arteriopathy. Nf1 heterozygosity (Nf1+/-) results in robust neointima formation, similar to humans, and myeloid-restricted Nf1+/- recapitulates this phenotype via MEK-ERK activation. Here we define the contribution of myeloid subpopulations to NF1 arteriopathy. Neutrophils from WT and Nf1+/- mice were functionally assessed in the presence of MEK and farnesylation inhibitors in vitro and neutrophil recruitment to lipopolysaccharide was assessed in WT and Nf1+/- mice. Littermate 12-15 week-old male wildtype and Nf1+/- mice were subjected to carotid artery ligation and provided either a neutrophil depleting antibody (1A8), liposomal clodronate to deplete monocytes/macrophages, or PD0325901 and neointima size was assessed 28 days after injury. Bone marrow transplant experiments assessed monocyte/macrophage mobilization during neointima formation. Nf1+/- neutrophils exhibit enhanced proliferation, migration, and adhesion via p21Ras activation of MEK in vitro and in vivo. Neutrophil depletion suppresses circulating Ly6Clow monocytes and enhances neointima size, while monocyte/macrophage depletion and deletion of CCR2 in bone marrow cells abolish neointima formation in Nf1+/- mice. Taken together, these findings suggest that neurofibromin-MEK-ERK activation in circulating neutrophils and monocytes during arterial remodeling is nuanced and points to important cross-talk between these populations in the pathogenesis of NF1 arteriopathy.


Subject(s)
Carotid Artery Injuries/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Myeloid Progenitor Cells/pathology , Neointima/pathology , Neurofibromatosis 1/pathology , Neurofibromin 1/physiology , Receptors, CCR2/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Myeloid Progenitor Cells/metabolism , Neointima/etiology , Neointima/metabolism , Neurofibromatosis 1/etiology , Neurofibromatosis 1/metabolism
3.
Transl Res ; 228: 52-63, 2021 02.
Article in English | MEDLINE | ID: mdl-32781282

ABSTRACT

Neurofibromatosis type 1 (NF1) is a heritable cancer predisposition syndrome resulting from mutations in the NF1 tumor suppressor gene. Genotype-phenotype correlations for NF1 are rare due to the large number of NF1 mutations and role of modifier genes in manifestations of NF1; however, emerging reports suggest that persons with NF1 display a distinct anthropometric and metabolic phenotype featuring short stature, low body mass index, increased insulin sensitivity, and protection from diabetes. Nf1 heterozygous (Nf1+/-) mice accurately reflect the dominant inheritance of NF1 and are regularly employed as a model of NF1. Here, we sought to identify whether Nf1+/- mice recapitulate the anthropometric and metabolic features identified in persons with NF1. Littermate 16-20 week-old male wildtype (WT) and Nf1+/- C57B/6J mice underwent nuclear magnetic resonance (NMR), indirect calorimetry, and glucose/insulin/pyruvate tolerance testing. In some experiments, tissues were harvested for NMR and histologic characterization. Nf1+/- mice are leaner with significantly reduced visceral and subcutaneous fat mass, which corresponds with an increased density of small adipocytes and reduced leptin levels. Additionally, Nf1+/- mice are highly reliant on carbohydrates as an energy substrate and display increased glucose clearance and insulin sensitivity, but normal response to pyruvate suggesting enhanced glucose utilization and preserved gluconeogenesis. Finally, WT and Nf1+/- mice subjected to high glucose diet were protected from diet-induced obesity and hyperglycemia. Our data suggest that Nf1+/- mice closely recapitulate the anthropometric and metabolic phenotype identified in persons with NF1, which will impact the interpretation of previous and future translational studies of NF1.


Subject(s)
Anthropometry , Genes, Neurofibromatosis 1 , Heterozygote , Neurofibromatosis 1/metabolism , Animals , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Neurofibromatosis 1/genetics , Neurofibromatosis 1/pathology
4.
Invest Ophthalmol Vis Sci ; 59(6): 2520-2528, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29847659

ABSTRACT

Purpose: Neurofibromatosis type 1 (NF1) is the result of inherited mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin. Eye manifestations are common in NF1 with recent reports describing a vascular dysplasia in the retina and choroid. Common features of NF1 retinopathy include tortuous and dilated feeder vessels that terminate in capillary tufts, increased endothelial permeability, and neovascularization. Given the retinal vascular phenotype observed in persons with NF1, we hypothesize that preserving neurofibromin may be a novel strategy to control pathologic retinal neovascularization. Methods: Nf1 expression in human endothelial cells (EC) was reduced using small hairpin (sh) RNA and EC proliferation, migration, and capacity to form vessel-like networks were assessed in response to VEGF and hypoxia. Wild-type (WT), Nf1 heterozygous (Nf1+/-), and Nf1flox/+;Tie2cre pups were subjected to hyperoxia/hypoxia using the oxygen-induced retinopathy model. Retinas were analyzed quantitatively for extent of retinal vessel dropout, neovascularization, and capillary branching. Results: Neurofibromin expression was suppressed in response to VEGF, which corresponded with activation of Mek-Erk and PI3-K-Akt signaling. Neurofibromin-deficient EC exhibited enhanced proliferation and network formation in response to VEGF and hypoxia via an Akt-dependent mechanism. In response to hyperoxia/hypoxia, Nf1+/- retinas exhibited increased vessel dropout and neovascularization when compared with WT retinas. Neovascularization was similar between Nf1+/- and Nf1flox/+;Tie2cre retinas, but capillary drop out in Nf1flox/+;Tie2cre retinas was significantly reduced when compared with Nf1+/- retinas. Conclusions: These data suggest that neurofibromin expression is essential for controlling endothelial cell proliferation and retinal neovascularization and therapies targeting neurofibromin-deficient EC may be beneficial.


Subject(s)
Cell Proliferation , Endothelial Cells/pathology , Neurofibromin 1/deficiency , Retinal Neovascularization/etiology , Retinopathy of Prematurity/etiology , Animals , Aorta, Thoracic/pathology , Cell Movement/physiology , Endothelial Cells/metabolism , Gene Silencing/physiology , Humans , Hypoxia/complications , Mice , Mice, Inbred C57BL , Oxygen/toxicity , Retinal Neovascularization/physiopathology , Retinal Vessels/pathology , Retinopathy of Prematurity/physiopathology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/pharmacology
5.
Free Radic Biol Med ; 97: 212-222, 2016 08.
Article in English | MEDLINE | ID: mdl-27266634

ABSTRACT

Neurofibromatosis type 1 (NF1) predisposes individuals to early and debilitating cardiovascular disease. Loss of function mutations in the NF1 tumor suppressor gene, which encodes the protein neurofibromin, leads to accelerated p21(Ras) activity and phosphorylation of multiple downstream kinases, including Erk and Akt. Nf1 heterozygous (Nf1(+/-)) mice develop a robust neointima that mimics human disease. Monocytes/macrophages play a central role in NF1 arterial stenosis as Nf1 mutations in myeloid cells alone are sufficient to reproduce the enhanced neointima observed in Nf1(+/-) mice. Though the molecular mechanisms underlying NF1 arterial stenosis remain elusive, macrophages are important producers of reactive oxygen species (ROS) and Ras activity directly regulates ROS production. Here, we use compound mutant and lineage-restricted mice to demonstrate that Nf1(+/-) macrophages produce excessive ROS, which enhance Nf1(+/-) smooth muscle cell proliferation in vitro and in vivo. Further, use of a specific NADPH oxidase-2 inhibitor to limit ROS production prevents neointima formation in Nf1(+/-) mice. Finally, mononuclear cells from asymptomatic NF1 patients have increased oxidative DNA damage, an indicator of chronic exposure to oxidative stress. These data provide genetic and pharmacologic evidence that excessive exposure to oxidant species underlie NF1 arterial stenosis and provide a platform for designing novels therapies and interventions.


Subject(s)
NADPH Oxidase 2/genetics , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carotid Stenosis/genetics , Carotid Stenosis/physiopathology , Cell Proliferation/genetics , DNA Damage/genetics , Heterozygote , Humans , Macrophages/metabolism , Macrophages/pathology , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 2/metabolism , Neointima/genetics , Neointima/metabolism , Neointima/physiopathology , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/physiopathology , Oxidative Stress/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Reactive Oxygen Species/metabolism
6.
BMC Cancer ; 16: 55, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26847345

ABSTRACT

BACKGROUND: Transgelin is an actin-binding protein that promotes motility in normal cells. Although the role of transgelin in cancer is controversial, a number of studies have shown that elevated levels correlate with aggressive tumor behavior, advanced stage, and poor prognosis. Here we sought to determine the role of transgelin more directly by determining whether experimental manipulation of transgelin levels in colorectal cancer (CRC) cells led to changes in metastatic potential in vivo. METHODS: Isogenic CRC cell lines that differ in transgelin expression were characterized using in vitro assays of growth and invasiveness and a mouse tail vein assay of experimental metastasis. Downstream effects of transgelin overexpression were investigated by gene expression profiling and quantitative PCR. RESULTS: Stable overexpression of transgelin in RKO cells, which have low endogenous levels, led to increased invasiveness, growth at low density, and growth in soft agar. Overexpression also led to an increase in the number and size of lung metastases in the mouse tail vein injection model. Similarly, attenuation of transgelin expression in HCT116 cells, which have high endogenous levels, decreased metastases in the same model. Investigation of mRNA expression patterns showed that transgelin overexpression altered the levels of approximately 250 other transcripts, with over-representation of genes that affect function of actin or other cytoskeletal proteins. Changes included increases in HOOK1, SDCCAG8, ENAH/Mena, and TNS1 and decreases in EMB, BCL11B, and PTPRD. CONCLUSIONS: Increases or decreases in transgelin levels have reciprocal effects on tumor cell behavior, with higher expression promoting metastasis. Chronic overexpression influences steady-state levels of mRNAs for metastasis-related genes.


Subject(s)
Cell Movement/genetics , Colorectal Neoplasms/genetics , Microfilament Proteins/biosynthesis , Muscle Proteins/biosynthesis , Neoplasm Metastasis , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Humans , Mice , Microfilament Proteins/genetics , Muscle Proteins/genetics , RNA, Messenger/biosynthesis
7.
Hum Mol Genet ; 25(6): 1129-39, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26740548

ABSTRACT

Persons with neurofibromatosis type 1 (NF1) have a predisposition for premature and severe arterial stenosis. Mutations in the NF1 gene result in decreased expression of neurofibromin, a negative regulator of p21(Ras), and increases Ras signaling. Heterozygous Nf1 (Nf1(+/-)) mice develop a marked arterial stenosis characterized by proliferating smooth muscle cells (SMCs) and a predominance of infiltrating macrophages, which closely resembles arterial lesions from NF1 patients. Interestingly, lineage-restricted inactivation of a single Nf1 allele in monocytes/macrophages is sufficient to recapitulate the phenotype observed in Nf1(+/-) mice and to mobilize proinflammatory CCR2+ monocytes into the peripheral blood. Therefore, we hypothesized that CCR2 receptor activation by its primary ligand monocyte chemotactic protein-1 (MCP-1) is critical for monocyte infiltration into the arterial wall and neointima formation in Nf1(+/-) mice. MCP-1 induces a dose-responsive increase in Nf1(+/-) macrophage migration and proliferation that corresponds with activation of multiple Ras kinases. In addition, Nf1(+/-) SMCs, which express CCR2, demonstrate an enhanced proliferative response to MCP-1 when compared with WT SMCs. To interrogate the role of CCR2 activation on Nf1(+/-) neointima formation, we induced neointima formation by carotid artery ligation in Nf1(+/-) and WT mice with genetic deletion of either MCP1 or CCR2. Loss of MCP-1 or CCR2 expression effectively inhibited Nf1(+/-) neointima formation and reduced macrophage content in the arterial wall. Finally, administration of a CCR2 antagonist significantly reduced Nf1(+/-) neointima formation. These studies identify MCP-1 as a potent chemokine for Nf1(+/-) monocytes/macrophages and CCR2 as a viable therapeutic target for NF1 arterial stenosis.


Subject(s)
Macrophages/pathology , Monocytes/pathology , Neointima/pathology , Neurofibromatosis 1/pathology , Receptors, CCR2/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Genes, Neurofibromatosis 1 , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/genetics , Neointima/metabolism , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromin 1/genetics , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/genetics , Signal Transduction
8.
Radiat Res ; 180(1): 17-24, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23692479

ABSTRACT

The carcinogenic risk of high-charge and energy (HZE) particle exposure arises from its ability to both induce complex DNA damage and from its ability to evoke deleterious, non-DNA targeted effects. We investigate here whether these nontargeted effects involve dysregulation of double-strand break repair, such that a history of HZE exposure heightens the risks from future injury. We used a new human cell reporter line, in which expression of the I-SceI meganuclease stimulates both translocations on different chromosomes, and deletions on the same chromosome. Exposure to 1.0 Gy of 600 MeV/u (56)Fe ions led to a doubling in the frequency of I-SceI-mediated translocations and a smaller, but nevertheless significant, increase in the frequency of I-SceI-mediated deletions. This mutagenic repair phenotype persisted for up to two weeks and eight population doublings. The phenotype was not induced by low-linear energy transfer radiation or by a lower dose of HZE-particle radiation (0.3 Gy) indicating that the effect is radiation quality and dose dependent. The mutagenic repair phenotype was associated with the presence of micronuclei and persistent DSB repair foci, consistent with a hypothesis that genomic stress is a causative factor.


Subject(s)
DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/genetics , Translocation, Genetic/radiation effects , Cell Line , Chromosome Deletion , Cosmic Radiation , Deoxyribonucleases, Type II Site-Specific/genetics , Humans , Linear Energy Transfer , Saccharomyces cerevisiae Proteins/genetics
9.
Nucleic Acids Res ; 37(20): 6746-53, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19759212

ABSTRACT

Mammalian cells repair DNA double-strand breaks (DSBs) via efficient pathways of direct, nonhomologous DNA end joining (NHEJ) and homologous recombination (HR). Prior work has identified a complex of two polypeptides, PSF and p54(nrb), as a stimulatory factor in a reconstituted in vitro NHEJ system. PSF also stimulates early steps of HR in vitro. PSF and p54(nrb) are RNA recognition motif-containing proteins with well-established functions in RNA processing and transport, and their apparent involvement in DSB repair was unexpected. Here we investigate the requirement for p54(nrb) in DSB repair in vivo. Cells treated with siRNA to attenuate p54(nrb) expression exhibited a delay in DSB repair in a gamma-H2AX focus assay. Stable knockdown cell lines derived by p54(nrb) miRNA transfection showed a significant increase in ionizing radiation-induced chromosomal aberrations. They also showed increased radiosensitivity in a clonogenic survival assay. Together, results indicate that p54(nrb) contributes to rapid and accurate repair of DSBs in vivo in human cells and that the PSF.p54(nrb) complex may thus be a potential target for radiosensitizer development.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Nuclear Matrix-Associated Proteins/metabolism , Octamer Transcription Factors/metabolism , RNA-Binding Proteins/metabolism , Radiation Tolerance , Cell Survival , Chromosome Aberrations , DNA-Binding Proteins , HeLa Cells , Humans , RNA, Small Interfering/metabolism
10.
J Biol Chem ; 278(43): 41631-5, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-12917393

ABSTRACT

Nonhomologous end joining is the most common mechanism of DNA double-strand break repair in human cells. Here we show that nonhomologous end joining can occur by two biochemically distinct pathways. One requires a fraction containing the Mre11-Rad50-NBS1 complex. The other requires a fraction containing a novel, approximately 200-kDa factor that does not correspond to any of the previously described double-strand break repair proteins. The two pathways converge, sharing a common requirement for the DNA ligase IV-XRCC4 complex to catalyze the final step of phosphodiester bond formation. Whereas the Mre11-Rad50-NBS1-dependent pathway does not require, and may be inhibited by, DNA-dependent protein kinase-mediated phosphorylation, the new pathway depends on this phosphorylation for release from a DNA-dependent protein kinase-mediated reaction checkpoint. The existence of two distinct pathways, which are differentially regulated by the DNA-dependent protein kinase, provides a possible explanation for the selective repair defects seen in DNA-dependent protein kinase-deficient mutants.


Subject(s)
DNA Repair Enzymes , DNA Repair , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Acid Anhydride Hydrolases , Cell Cycle Proteins/metabolism , Chromatography, Agarose , DNA Ligase ATP , DNA Ligases/metabolism , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , MRE11 Homologue Protein , Nuclear Proteins/metabolism , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...