Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Brain Res ; 1775: 147743, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34843701

ABSTRACT

Dravet Syndrome (DS) is a severe developmental and epileptic encephalopathy typically caused by loss-of-function de novo mutations in the SCN1A gene which encodes the voltage-gated sodium channel isoform NaV1.1. Decreased NaV1.1 expression results in impaired excitability of inhibitory interneurons and seizure onset. To date, there are no clinically available treatments for DS that directly address the core mechanism of disease; reduced NaV1.1 expression levels in interneurons. Recently, Targeted Augmentation of Nuclear Gene Output (TANGO) of SCN1A by the antisense oligonucleotide (ASO) STK-001, was shown to increase Scn1a mRNA levels, increase NaV1.1 protein expression, reduce seizures, and improve survival in the Scn1a+/- mouse model of DS. However, it remains unknown whether STK-001 treatment rescues the reduced intrinsic excitability of parvalbumin-positive (PV) inhibitory interneurons associated with DS. In this study, we demonstrate that STK-001 treatment reduces seizures, prolongs survival, and rescues PV interneuron excitability in Scn1a+/- mice to levels observed in WT littermates. Together, these results support the notion that TANGO-mediated augmentation of NaV1.1 levels directly targets and rescues one of the core disease mechanisms of DS.


Subject(s)
Action Potentials/physiology , Epilepsies, Myoclonic/genetics , Interneurons/metabolism , NAV1.1 Voltage-Gated Sodium Channel/genetics , Parvalbumins/metabolism , Seizures/genetics , Animals , Disease Models, Animal , Epilepsies, Myoclonic/physiopathology , Mice , Oligonucleotides, Antisense , Seizures/physiopathology
2.
Bio Protoc ; 11(3): e3910, 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33732797

ABSTRACT

Densitometric analysis is often used to quantify NaV1.1 protein on immunoblots, although the sensitivity and dilution linearity of the method are usually poor. Here we present a protocol for quantification of NaV1.1 in mouse brain tissues using a Meso Scale Discovery-Electrochemiluminescence (MSD-ECL) method. MSD-ECL is based on ELISA (enzyme-linked immunosorbent assay) and uses electrochemiluminescence to produce measurable signals. Two different antibodies are used in this assay to capture and detect NaV1.1 respectively in brain tissue lysate. The specificity of the antibodies is confirmed by Scn1a gene knock-out tissue. The calibration curve standards used in this assay were generated with mouse liver lysate spiked with mouse brain lysate, instead of using a recombinant protein. We showed that this method was qualified and used for quantification of NaV1.1 in mouse brain tissues with specificity, accuracy and precision.

3.
Sci Transl Med ; 12(558)2020 08 26.
Article in English | MEDLINE | ID: mdl-32848094

ABSTRACT

Dravet syndrome (DS) is an intractable developmental and epileptic encephalopathy caused largely by de novo variants in the SCN1A gene, resulting in haploinsufficiency of the voltage-gated sodium channel α subunit NaV1.1. Here, we used Targeted Augmentation of Nuclear Gene Output (TANGO) technology, which modulates naturally occurring, nonproductive splicing events to increase target gene and protein expression and ameliorate disease phenotype in a mouse model. We identified antisense oligonucleotides (ASOs) that specifically increase the expression of productive Scn1a transcript in human cell lines, as well as in mouse brain. We show that a single intracerebroventricular dose of a lead ASO at postnatal day 2 or 14 reduced the incidence of electrographic seizures and sudden unexpected death in epilepsy (SUDEP) in the F1:129S-Scn1a +/- × C57BL/6J mouse model of DS. Increased expression of productive Scn1a transcript and NaV1.1 protein was confirmed in brains of treated mice. Our results suggest that TANGO may provide a unique, gene-specific approach for the treatment of DS.


Subject(s)
Epilepsies, Myoclonic , Sudden Unexpected Death in Epilepsy , Animals , Epilepsies, Myoclonic/genetics , Incidence , Mice , Mice, Inbred C57BL , NAV1.1 Voltage-Gated Sodium Channel/genetics , Oligonucleotides, Antisense , Seizures/genetics
4.
Nat Commun ; 11(1): 3501, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647108

ABSTRACT

While most monogenic diseases are caused by loss or reduction of protein function, the need for technologies that can selectively increase levels of protein in native tissues remains. Here we demonstrate that antisense-mediated modulation of pre-mRNA splicing can increase endogenous expression of full-length protein by preventing naturally occurring non-productive alternative splicing and promoting generation of productive mRNA. Bioinformatics analysis of RNA sequencing data identifies non-productive splicing events in 7,757 protein-coding human genes, of which 1,246 are disease-associated. Antisense oligonucleotides targeting multiple types of non-productive splicing events lead to increases in productive mRNA and protein in a dose-dependent manner in vitro. Moreover, intracerebroventricular injection of two antisense oligonucleotides in wild-type mice leads to a dose-dependent increase in productive mRNA and protein in the brain. The targeting of natural non-productive alternative splicing to upregulate expression from wild-type or hypomorphic alleles provides a unique approach to treating genetic diseases.


Subject(s)
Alternative Splicing , Gene Expression Regulation , Oligonucleotides, Antisense/pharmacology , Alleles , Animals , Animals, Newborn , Brain/metabolism , Computational Biology , Exons , Female , Gene Expression/drug effects , HEK293 Cells , Humans , Introns , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Transcriptional Activation/drug effects , Up-Regulation
5.
Biochem J ; 406(2): 203-7, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17608623

ABSTRACT

PCSK9 (proprotein convertase subtilisin/kexin 9) is a secreted serine protease that regulates cholesterol homoeostasis by inducing post-translational degradation of hepatic LDL-R [LDL (low-density lipoprotein) receptor]. Intramolecular autocatalytic processing of the PCSK9 zymogen in the endoplasmic reticulum results in a tightly associated complex between the prodomain and the catalytic domain. Although the autocatalytic processing event is required for proper secretion of PCSK9, the requirement of proteolytic activity in the regulation of LDL-R is currently unknown. Co-expression of the prodomain and the catalytic domain in trans allowed for production of a catalytically inactive secreted form of PCSK9. This catalytically inactive PCSK9 was characterized and shown to be functionally equivalent to the wild-type protein in lowering cellular LDL uptake and LDL-R levels. These findings suggest that, apart from autocatalytic processing, the protease activity of PCSK9 is not necessary for LDL-R regulation.


Subject(s)
Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Cell Line , Humans , Mutation/genetics , Serine/genetics , Serine/metabolism , Serine Endopeptidases/genetics
6.
Cancer Res ; 66(1): 221-31, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397235

ABSTRACT

FTY720, a potent immunomodulator, becomes phosphorylated in vivo (FTY-P) and interacts with sphingosine-1-phosphate (S1P) receptors. Recent studies showed that FTY-P affects vascular endothelial growth factor (VEGF)-induced vascular permeability, an important aspect of angiogenesis. We show here that FTY720 has antiangiogenic activity, potently abrogating VEGF- and S1P-induced angiogenesis in vivo in growth factor implant and corneal models. FTY720 administration tended to inhibit primary and significantly inhibited metastatic tumor growth in a mouse model of melanoma growth. In combination with a VEGFR tyrosine kinase inhibitor PTK787/ZK222584, FTY720 showed some additional benefit. FTY720 markedly inhibited tumor-associated angiogenesis, and this was accompanied by decreased tumor cell proliferation and increased apoptosis. In transfected HEK293 cells, FTY-P internalized S1P1 receptors, inhibited their recycling to the cell surface, and desensitized S1P receptor function. Both FTY720 and FTY-P apparently failed to impede VEGF-produced increases in mitogen-activated protein kinase activity in human umbilical vascular endothelial cells (HUVEC), and unlike its activity in causing S1PR internalization, FTY-P did not result in a decrease of surface VEGFR2 levels in HUVEC cells. Pretreatment with FTY720 or FTY-P prevented S1P-induced Ca2+ mobilization and migration in vascular endothelial cells. These data show that functional antagonism of vascular S1P receptors by FTY720 potently inhibits angiogenesis; therefore, this may provide a novel therapeutic approach for pathologic conditions with dysregulated angiogenesis.


Subject(s)
Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/antagonists & inhibitors , Animals , Calcium/metabolism , Cell Growth Processes/drug effects , Cell Movement/drug effects , Cornea/blood supply , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Female , Fingolimod Hydrochloride , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/drug effects , Phosphorylation , Phthalazines/pharmacology , Propylene Glycols/pharmacokinetics , Pyridines/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
FASEB J ; 17(8): 896-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12670875

ABSTRACT

Endostatin, a proteolytic fragment of collagen XVIII, is an endogenous inhibitor of tumor angiogenesis that also inhibits choroidal neovascularization. In this study, we assessed the effects of increased intraocular expression of endostatin on vascular endothelial growth factor (VEGF)-induced changes in the retina. After subretinal injection of a pair of gutless adenoviral vectors (AGV) designed to provide tamoxifen-inducible expression of endostatin, diffuse endostatin immunoreactivity was induced thoroughout the retina by administration of tamoxifen. Induction of endostatin in double transgenic mice with doxycycline-induced expression of VEGF in the retina resulted in significant suppression of leakage of intravascular [3H]mannitol into the retina. The ability of endostatin to reduce VEGF-induced retinal vascular permeability was confirmed by using [3H]mannitol leakage and two other parameters, fluorescein leakage and retinal thickness, after subretinal injection of a bovine immunodeficiency lentiviral vector coding for endostatin (BIV-vectored endostatin, or BIVendostatin). Subretinal injection of BIVendostatin resulted in more discrete, less intense staining for endostatin in the retina than that seen with the inducible AGV system, which suggested lower levels and allowed visualization of sites where endostatin was concentrated. Endostatin staining outlined retinal blood vessels, which suggested endostatin binding to a component of vessel walls. More prolonged or higher level expression of VEGF in the retina resulted in neovascularization and retinal detachment, both of which were also significantly reduced by BIVendostatin. These data suggest that endostatin may be an endogenous inhibitor of vasopermeability as well as neovascularization. In patients with diabetic retinopathy, endostatin gene transfer may provide a way to decrease the risk of three causes of visual loss: macular edema, neovascularization, and retinal detachment.


Subject(s)
Capillary Permeability/drug effects , Collagen/physiology , Endothelial Growth Factors/pharmacology , Eye/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphokines/pharmacology , Peptide Fragments/physiology , Retinal Detachment/pathology , Retinal Neovascularization/pathology , Animals , Collagen/biosynthesis , Collagen/genetics , Collagen Type XVIII , Endostatins , Endothelial Growth Factors/genetics , Eye/blood supply , Eye/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Lymphokines/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Detachment/chemically induced , Retinal Neovascularization/chemically induced , Transfection/methods , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
8.
Coron Artery Dis ; 16(7): 451-6, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16205454

ABSTRACT

OBJECTIVES: We previously demonstrated that sphingosine kinase (SPK) increases the level of extracellular sphingosine-1-phosphate and promotes neovascularization in a mouse matrigel model. In this study, we tested the hypothesis that SPK gene transfer using a novel adenoviral 'gutless' vector (AGV) can enhance arteriogenesis in a rabbit hindlimb ischemia model. METHODS: Thirty-five male New Zealand white rabbits were randomized to the AGV-SPK group (n=13), AGV-null group (n=13), and control group (n=9). On day 10, after the induction of unilateral hindlimb ischemia, gene vectors or buffer were introduced and the effect examined on day 30, using calf blood pressure, quantitative angiographic analysis, and histology. RESULTS: Calf systolic blood pressure ratios of the ischemic limb to the normal limb on day 30 were 0.77+/-0.13 in control groups, including the AGV-null group, and 0.91+/-0.14 in the AGV-SPK group (P<0.05). Angiographic vessel counts were significantly increased (8.0+/-2.1 at baseline and 11.8+/-3.2 on day 30, P<0.001) in the AGV-SPK group. Histologic analysis showed that microscopic total vessel counts on day 30 were 3.5+/-1.8/field in the control and AGV-null group and 5.4+/-1.0/field in the AGV-SPK group. Arterioles (AGV-SPK; 3.0+/-0.8 versus control and AGV-null; 2.1+/-1.1, P<0.05) were significantly increased in the AGV-SPK group. CONCLUSIONS: This study shows that SPK promotes arteriogenesis, as evidenced by the maximal improvement in the blood pressure restoration and collateral vessel counts. SPK may be an important angiogenic target to improve perfusion in ischemic tissues.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Genetic Therapy/methods , Ischemia/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adenoviridae/genetics , Angiogenesis Inducing Agents/therapeutic use , Angiography , Animals , Femoral Artery/drug effects , Femoral Artery/growth & development , Gene Transfer Techniques , Genetic Vectors/biosynthesis , Hindlimb/blood supply , Male , Models, Animal , Neovascularization, Physiologic/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/therapeutic use , Rabbits , Regional Blood Flow/drug effects
9.
Br J Pharmacol ; 165(6): 2000-2008, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22014125

ABSTRACT

BACKGROUND AND PURPOSE: Inhibition of the renin angiotensin system (RAS) has been consistently demonstrated to reduce atherosclerosis. However, there has been no direct comparison among the three available pharmacological modes of inhibiting the RAS, which are inhibitors of renin, ACE and angiotensin II type 1 receptor. The purpose of this study was to determine the relative effects of these three modes of pharmacological RAS inhibition in reducing atherosclerosis by determining the dose-response relationships. EXPERIMENTAL APPROACH: Male LDL receptor -/- mice were administered either vehicle or any of three doses of aliskiren, enalapril or losartan through s.c. infusion for 12 weeks. All mice were fed a saturated fat-enriched diet during drug infusions. Systolic and diastolic BPs were measured during the study using a non-invasive tail-cuff system. Plasma cholesterol and renin concentrations, atherosclerotic lesions, and renal angiotensin II concentrations were determined at the termination of the study. KEY RESULTS: Plasma renin concentrations were increased by all three drugs. None of the drugs changed plasma cholesterol concentrations. All drugs produced a dose-related decrease in BP. All three drugs also profoundly reduced atherosclerosis in a dose-dependent manner. The highest dose of each drug markedly attenuated lesion size, with no significant differences between the different drugs. The highest dose of each drug also similarly reduced renal angiotensin II concentrations. CONCLUSION AND IMPLICATIONS: Drugs that inhibit the RAS, irrespective of their mode of inhibition, profoundly affect atherosclerotic lesion development in a dose-dependent manner.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Atherosclerosis/drug therapy , Hypercholesterolemia/drug therapy , Renin/antagonists & inhibitors , Amides/pharmacology , Amides/therapeutic use , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aorta/pathology , Atherosclerosis/etiology , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Blood Pressure/drug effects , Cholesterol/blood , Diet, High-Fat , Enalapril/pharmacology , Enalapril/therapeutic use , Fumarates/pharmacology , Fumarates/therapeutic use , Hypercholesterolemia/complications , Hypercholesterolemia/etiology , Hypercholesterolemia/physiopathology , Kidney/metabolism , Losartan/pharmacology , Male , Mice , Mice, Knockout , Receptor, Angiotensin, Type 1/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Renin/blood , Renin-Angiotensin System/drug effects
10.
J Hypertens ; 30(1): 107-16, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22134386

ABSTRACT

OBJECTIVE: Aliskiren is the first commercially available, orally active, direct renin inhibitor approved to treat hypertension. The renin-angiotensin system has been shown to be a significant contributor to the development of hypercholesterolemia-induced atherosclerosis. The aim of this study was to evaluate the antiatherosclerotic and plaque stabilization effects of aliskiren alone and in combination with atorvastatin. METHODS: APOE*3Leiden.CETP mice (n = 14-17/group) were fed a western-type diet (containing 0.25% cholesterol) alone or were treated with either aliskiren (15 mg/kg per day), atorvastatin (3.6 mg/kg per day) or a combination of aliskiren and atorvastatin. Effects on SBP, total cholesterol, inflammation markers and atherosclerotic size and composition were assessed. RESULTS: Aliskiren reduced SBP (-19%, P < 0.001) and atorvastatin reduced total cholesterol (-24%, P < 0.001). Atherosclerotic lesion area was reduced by aliskiren (-40%, P < 0.01), atorvastatin (-61%, P < 0.001) and the combination treatment (-69%, P < 0.001). Aliskiren alone and together with atorvastatin decreased the number of T cells in the aortic root area (-60%, P < 0.01; -41%, P < 0.05), as well as macrophage (-64%, P < 0.001; -72%, P < 0.001) and necrotic area (-52%, P = 0.071; -84%, P < 0.001) in the lesion. Atorvastatin alone and together with aliskiren decreased monocyte adherence (-43%, P < 0.05 and -51%, P < 0.01) and monocyte chemoattractant protein-1 (both -36%, P < 0.01). The combination treatment decreased the number of lesions (-17%, P < 0.05) and E-selectin (-17%, P < 0.05). CONCLUSION: Aliskiren inhibited atherosclerosis development and improved plaque stability alone and in combination with atorvastatin, possibly via a mechanism involving T cells. These results suggest a potential benefit of using aliskiren in a clinical setting, particularly in combination with statin treatment.


Subject(s)
Amides/pharmacology , Apolipoproteins E/genetics , Atherosclerosis/prevention & control , Fumarates/pharmacology , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Atherosclerosis/genetics , Atorvastatin , Female , Mice , Mice, Transgenic
11.
J Cell Biochem ; 91(4): 766-76, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14991768

ABSTRACT

The amino-terminal domain of the extracellular matrix (ECM) protein thrombospondin-1 (TSP-1) mediates binding to cell surface heparan sulfate proteoglycans (HSPG) as well as binding to the endocytic receptor, low density lipoprotein-related protein (LRP-1). We previously found that recombinant TSP-1 containing the amino-terminal residues 1-214, retained both of these interactions (Mikhailenko et al. [1997]: J Biol Chem 272:6784-6791). Here, we examined the activity of a recombinant protein containing amino-terminal residues 1-90 of TSP-1 and found that this domain did not retain high-affinity heparin-binding. The loss of heparin-binding correlated with decreased binding to the fibroblast cell surface. However, both ligand blotting and solid phase binding studies indicate that this truncated fragment of TSP-1 retained high-affinity binding to LRP-1. Consistent with this, it also retained the ability to block the uptake and degradation of (125)I-TSP-1. However, TSP-1(1-90) itself was poorly endocytosed and this truncated amino-terminal domain was considerably more effective than the full-length heparin-binding domain (HBD) of TSP-1 in blocking the catabolism of endogenously expressed TSP-1. These results indicate that TSP-1 binding to LRP-1 does not require prior or concomitant interaction with cell surface HSPG but suggest subsequent endocytosis requires high-affinity heparin-binding.


Subject(s)
Endocytosis , Heparan Sulfate Proteoglycans/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Thrombospondin 1/chemistry , Thrombospondin 1/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/metabolism , Endocytosis/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/genetics , Heparin/metabolism , Humans , Mice , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Protein Binding/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/genetics , Thrombospondin 1/pharmacology
12.
Kidney Int ; 63(5): 1658-70, 2003 May.
Article in English | MEDLINE | ID: mdl-12675841

ABSTRACT

BACKGROUND: Imprinted genes, mesodermal specific cDNA or transcript (MEST) and H19, are implicated in peri-implantation embryogenesis, and their expression was assessed in embryonic kidneys undergoing glucose-induced dysmorphogenesis. METHODS: MEST and H19 mRNA expression was assessed by Northern blot analysis in embryonic kidneys of mice harvested at day 15 to day 19 of gestation and of 1-week-old mice obtained from hyperglycemic mothers. A full-length mouse MEST cDNA was isolated, subcloned into an expression vector, a recombinant protein prepared and an antibody raised; the latter was used to assess protein expression by immunoprecipitation and immunofluorescence microscopy in day 13 metanephric explants subjected to high glucose ambience. Also, MEST mRNA expression was assessed in high d glucose-treated explants by competitive reverse transcription-polymerase chain reaction (RT-PCR) analyses and by in situ tissue autoradiography. RESULTS: A high expression of MEST and H19 with respective transcript size of approximately 2.7 and approximately 2.4 kb was observed in fetal kidneys, and their expression decreased during the successive stages of gestation and was undetectable in the postnatal period. At day 13, the MEST mRNA was expressed in the mesenchyme, while H19 was expressed in the ureteric bud branches and epithelial elements of the metanephros. Their expression decreased with progression of gestation. By competitive RT-PCR and Northern blot and in situ autoradiographic analyses, both MEST and H19 expressions decreased in day 13 explants treated with high glucose and in the kidneys of fetuses obtained from diabetic mothers. The MEST protein expression was observed in the metanephric epithelial elements and ureteric bud branches instead of in the mesenchyme, and its expression decreased in glucose-treated dysmorphogenetic explants, as assessed by immunofluorescence and immunoprecipitation methods. CONCLUSION: MEST and H19 imprinted genes are strategically located in the mammalian embryonic metanephros. They are developmentally regulated and their concomitant decreased expression in high glucose ambience or diabetic state did not follow the prevailing dogma of reciprocal inactivation/activation of imprinted genes, and such a decrease may be responsible for the perturbed epithelial:mesenchymal interactions leading to dysmorphogenesis of the mammalian metanephros.


Subject(s)
Diabetic Nephropathies/physiopathology , Genomic Imprinting , Kidney/physiology , Proteins/genetics , RNA, Untranslated/genetics , Animals , Animals, Newborn , Antibodies/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Glucose/pharmacology , Hyperglycemia/physiopathology , Kidney/embryology , Male , Mice , Mice, Inbred ICR , Organ Culture Techniques , Pregnancy , Pregnancy in Diabetics/physiopathology , Proteins/immunology , Proteins/pharmacology , RNA, Long Noncoding , RNA, Messenger/analysis , Recombinant Fusion Proteins/pharmacology
13.
J Biol Chem ; 277(8): 6667-75, 2002 Feb 22.
Article in English | MEDLINE | ID: mdl-11741921

ABSTRACT

The enzyme sphingosine kinase (SK) catalyzes the formation of sphingosine 1-phosphate (S1P), a bioactive lipid that acts extracellularly on G protein-coupled receptors of the S1P(1)/EDG-1 subfamily. Although S1P is formed in the cytosol of various cells, S1P release is not understood and is controversial because this lipid mediator is also regarded as a second messenger. In this report, we describe the existence of an extracellular S1P-generating system in vascular endothelial cells. Endothelial cells release SK constitutively and form S1P in the range of receptor stimulation. Levels of sphingosine but not ATP in the extracellular environment are rate-limiting. Treatment of endothelial cells with small interfering RNA for SK-1 transcript specifically inhibited SK export, and SK-1-transfected human embryonic kidney 293 cells exhibited enhanced release of SK-1. The export of SK-1 is constitutive and is inhibited by cytochalasin D and treatment at 4 degrees C but not by brefeldin A or nocodazole, suggesting that a nonclassical secretory pathway that requires the actin cytoskeleton dynamics is involved. Because S1P regulates angiogenesis and vascular maturation, we overexpressed SK-1 using an adenoviral vector in vivo in the Matrigel system of angiogenesis. Overexpression of SK-1 resulted in enhanced release of SK activity and induced angiogenesis and vascular maturation. These findings suggest that S1P is made in the extracellular milieu and that extracellular export of SK contributes to the action of S1P in the vascular system.


Subject(s)
Endothelium, Vascular/metabolism , Gene Expression Regulation, Enzymologic , Lysophospholipids , Neovascularization, Physiologic , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Cell Surface , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Xenopus Proteins , Animals , Calcium/metabolism , Cell Line , Culture Media, Conditioned , Cytosol/metabolism , Female , Gene Silencing , Homeodomain Proteins/metabolism , Humans , Immediate-Early Proteins/metabolism , Kidney , Kinetics , Oocytes/physiology , Protein Transport , RNA, Small Interfering , RNA, Untranslated/genetics , Receptors, Lysophospholipid , Repressor Proteins/metabolism , Transfection , Xenopus , Zinc Finger E-box Binding Homeobox 2
14.
J Cell Physiol ; 199(3): 399-411, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15095287

ABSTRACT

Vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF-1) have been implicated as important stimulatory factors for retinal neovascularization. In this study, we used intraocular gene transfer with gutless adenoviral (AGV) vectors to determine the effect of increased intraocular expression of VEGF, IGF-1, or sphingosine kinase (SPK), which produces sphingosine-1-phosphate, another angiogenic factor. Retinal neovascularization did not occur from intravitreous AGV-vectored VEGF, IGF-1, SPK, or combined VEGF and IGF-1, except occasionally adjacent to the retinal penetration site from the injection. However, corneal and iris neovascularization occurred after 2 weeks in all eyes injected with AGV.VEGF, but not those injected with only AGV.IGF-1 or AGV.SPK. These data suggest that the superficial capillary bed of the retina is relatively insensitive to VEGF, IGF-1, or SPK in adult mice, except when combined with retinal trauma. However, AGV-vectored VEGF is sufficient to consistently cause severe corneal and iris neovascularization. This provides a model for anterior segment neovascularization, which unlike previous models is relatively inexpensive and is not plagued by spontaneous regression, and therefore, may be useful for identification of new treatments.


Subject(s)
Adenoviridae , Eye/blood supply , Retinal Neovascularization , Transduction, Genetic , Vascular Endothelial Growth Factor A/biosynthesis , Adenoviridae/genetics , Animals , DNA Primers , Genetic Vectors , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Insulin-Like Growth Factor I/biosynthesis , Insulin-Like Growth Factor I/genetics , Lac Operon , Lysophospholipids/metabolism , Mice , Neovascularization, Physiologic/genetics , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL