Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Allergy ; 69(2): 237-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24127643

ABSTRACT

BACKGROUND: Edema represents a key feature of nasal polyp (NP) disease. Members of the vascular endothelial growth factor (VEGF) family may be involved, but the precise role of VEGF-A, VEGF-B, placental growth factor (PlGF), and their receptors VEGFR1 and VEGFR2 in NP edema formation remains elusive. OBJECTIVE: Exploring the expression of VEGF family members and their receptors and their correlation with clinical, radiological, and edema markers in NP. METHODS: The expression of VEGF-A, VEGF-B, PlGF, VEGFR1, and VEGFR2 was measured in NP (n = 23) and control tissue (n = 22) at mRNA and protein level. Edema was evaluated by measuring albumin levels and wet/dry ratios. Computed tomography (CT) scans were scored using the Lund-Mackay scoring system. IL-5 mRNA expression was determined by real-time RT-PCR. Cell suspensions from NP (n = 10) and control tissue (n = 12) were stimulated in vitro with IL-1ß or TNFα. RESULTS: mRNA expression of VEGFR1 and VEGF-B was significantly higher in NP compared with control tissue. Expression levels of VEGF-B and VEGFR1 significantly correlated with NP albumin content (VEGF-B: P = 0.0208; VEGFR1: P = 0.0293), CT scan scores (VEGF-B: P = 0.0075; VEGFR1: P = 0.0068), and IL-5 mRNA (VEGF-B: P = 0.0027; VEGFR1: P = 0.0001). In vitro stimulation of control and NP tissue cell suspensions with IL-1ß or TNFα significantly reduced the expression of VEGFR2 in control tissue, without altering VEGFR1 and VEGF-B expression. hVEGF-B induced nitric oxide production in NP macrophages (P < 0.05). CONCLUSION: Expression levels of VEGFR1 and VEGF-B correlate with edema and clinical markers of NP disease and therefore represent potential therapeutic targets.


Subject(s)
Nasal Polyps/metabolism , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Female , Humans , Immunohistochemistry , Male , Membrane Proteins/analysis , Membrane Proteins/biosynthesis , Nasal Polyps/pathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor B/analysis , Vascular Endothelial Growth Factor B/biosynthesis , Vascular Endothelial Growth Factor Receptor-1/analysis , Vascular Endothelial Growth Factor Receptor-2/analysis , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
4.
Nat Genet ; 17(4): 439-44, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9398846

ABSTRACT

The molecular mechanisms predisposing to atherosclerotic aneurysm formation remain undefined. Nevertheless, rupture of aortic aneurysms is a major cause of death in Western societies, with few available treatments and poor long-term prognosis. Indirect evidence suggests that matrix metalloproteinases (MMPs) and plasminogen activators (PAs) are involved in its pathogenesis. MMPs are secreted as inactive zymogens (pro-MMPs), requiring activation in the extracellular compartment. Plasmin, generated from the zymogen plasminogen by tissue-type plasminogen activator (t-PA) or urokinase-type plasminogen activator (u-PA; refs 14,15), has been proposed as a possible activator in vitro, but evidence for such a role in vivo is lacking. Analysis of atherosclerotic aorta in mice with a deficiency of apoliprotein E (Apoe-/-; ref. 18), singly or combined with a deficiency of t-PA (Apoe-/-:Plat-/-) or of u-PA (Apoe-/-:Plau-/-; ref. 19), indicated that deficiency of u-PA protected against media destruction and aneurysm formation, probably by means of reduced plasmin-dependent activation of pro-MMPs. This genetic evidence suggests that plasmin is a pathophysiologically significant activator of pro-MMPs in vivo and may have implications for the design of therapeutic strategies to prevent aortic-wall destruction by controlling Plau gene function.


Subject(s)
Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Thoracic/enzymology , Fibrinolysin/metabolism , Metalloendopeptidases/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Aortic Aneurysm, Abdominal/etiology , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/pathology , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Collagen/metabolism , Diet, Atherogenic , Elastin/metabolism , Enzyme Activation , Female , Macrophages/enzymology , Male , Mice , Mice, Knockout , Tunica Media/enzymology , Tunica Media/pathology
5.
Nat Genet ; 17(1): 49-57, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9288097

ABSTRACT

The cerebro-hepato-renal syndrome of Zellweger is a fatal inherited disease caused by deficient import of peroxisomal matrix proteins. The pathogenic mechanisms leading to extreme hypotonia, severe mental retardation and early death are unknown. We generated a Zellweger animal model through inactivation of the murine Pxr1 gene (formally known as Pex5) that encodes the import receptor for most peroxisomal matrix proteins. Pxr1-/- mice lacked morphologically identifiable peroxisomes and exhibited the typical biochemical abnormalities of Zellweger patients. They displayed intrauterine growth retardation, were severely hypotonic at birth and died within 72 hours. Analysis of the neocortex revealed impaired neuronal migration and maturation and extensive apoptotic death of neurons.


Subject(s)
Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Zellweger Syndrome/genetics , Animals , Animals, Newborn , Apoptosis , Base Sequence , Brain/metabolism , Brain/pathology , Cerebral Cortex/pathology , DNA/biosynthesis , DNA Primers , Death , Disease Models, Animal , Female , Fetal Growth Retardation , Fibroblasts/metabolism , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/pathology , Neurons/physiology , Peroxisome-Targeting Signal 1 Receptor , Polymerase Chain Reaction , Pregnancy , Receptors, Cytoplasmic and Nuclear/metabolism , Recombination, Genetic , Zellweger Syndrome/pathology , Zellweger Syndrome/physiopathology
6.
Nat Genet ; 28(2): 131-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11381259

ABSTRACT

Hypoxia stimulates angiogenesis through the binding of hypoxia-inducible factors to the hypoxia-response element in the vascular endothelial growth factor (Vegf) promotor. Here, we report that deletion of the hypoxia-response element in the Vegf promotor reduced hypoxic Vegf expression in the spinal cord and caused adult-onset progressive motor neuron degeneration, reminiscent of amyotrophic lateral sclerosis. The neurodegeneration seemed to be due to reduced neural vascular perfusion. In addition, Vegf165 promoted survival of motor neurons during hypoxia through binding to Vegf receptor 2 and neuropilin 1. Acute ischemia is known to cause nonselective neuronal death. Our results indicate that chronic vascular insufficiency and, possibly, insufficient Vegf-dependent neuroprotection lead to the select degeneration of motor neurons.


Subject(s)
Cell Hypoxia/genetics , Endothelial Growth Factors/genetics , Lymphokines/genetics , Motor Neurons/pathology , Nerve Degeneration/genetics , Response Elements/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Axons/physiology , Binding Sites , Electrophysiology , Endothelial Growth Factors/metabolism , Humans , Lymphokines/metabolism , Mice , Mice, Knockout , Motor Neurons/physiology , Muscle Contraction , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1 , Peripheral Nerves/pathology , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Sequence Deletion , Spinal Cord/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
7.
Nat Med ; 6(4): 389-95, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742145

ABSTRACT

Endothelial and smooth muscle cells interact with each other to form new blood vessels. In this review, the cellular and molecular mechanisms underlying the formation of endothelium-lined channels (angiogenesis) and their maturation via recruitment of smooth muscle cells (arteriogenesis) during physiological and pathological conditions are summarized, alongside with possible therapeutic applications.


Subject(s)
Arterioles/physiopathology , Neovascularization, Pathologic , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/growth & development , Humans , Muscle, Smooth, Vascular/physiopathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology
8.
Nat Med ; 7(7): 864-8, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11433354

ABSTRACT

Intravital microscopy coupled with chronic animal window models has provided stunning insight into tumor pathophysiology, including gene expression, angiogenesis, cell adhesion and migration, vascular, interstitial and lymphatic transport, metabolic microenvironment and drug delivery. However, the findings to date have been limited to the tumor surface (< 150 microm). Here, we show that the multiphoton laser-scanning microscope can provide high three-dimensional resolution of gene expression and function in deeper regions of tumors. These insights could be critical to the development of novel therapeutics that target not only the tumor surface, but also internal regions.


Subject(s)
Gene Expression , Microscopy/methods , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic , Cell Adhesion , Hemodynamics , Lasers , Leukocytes/cytology , Photons
9.
Nat Med ; 7(1): 59-64, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135617

ABSTRACT

Tissue-plasminogen activator (t-PA) is now available for the treatment of thrombo-embolic stroke but adverse effects have been reported in some patients, particularly hemorrhaging. In contrast, the results of animal studies have indicated that t-PA could increase neuronal damage after focal cerebral ischemia. Here we report for the first time that t-PA potentiates signaling mediated by glutamatergic receptors by modifying the properties of the N-methyl-D-aspartate (NMDA) receptor. When depolarized, cortical neurons release bio-active t-PA that interacts with and cleaves the NR1 subunit of the NMDA receptor. Moreover, the treatment with recombinant t-PA leads to a 37% increase in NMDA-stimulated fura-2 fluorescence, which may reflect an increased NMDA-receptor function. These results were confirmed in vivo by the intrastriatal injection of recombinant-PA, which potentiated the excitotoxic lesions induced by NMDA. These data provide insight into the regulation of NMDA-receptor-mediated signaling and could initiate therapeutic strategies to improve the efficacy of t-PA treatment in man.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction , Tissue Plasminogen Activator/metabolism , Animals , Calcium/metabolism , Cell Death , Hydrolysis , Ion Transport , Membrane Potentials , Neurons/metabolism , Neurons/physiology
10.
Nat Med ; 7(2): 215-21, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175853

ABSTRACT

The growth arrest-specific gene 6 product (Gas6) is a secreted protein related to the anticoagulant protein S but its role in hemostasis is unknown. Here we show that inactivation of the Gas6 gene prevented venous and arterial thrombosis in mice, and protected against fatal collagen/epinephrine-induced thrombo embolism. Gas6-/- mice did not, however, suffer spontaneous bleeding and had normal bleeding after tail clipping. In addition, we found that Gas6 antibodies inhibited platelet aggregation in vitro and protected mice against fatal thrombo embolism without causing bleeding in vivo. Gas6 amplified platelet aggregation and secretion in response to known agonists. Platelet dysfunction in Gas6-/- mice resembled that of patients with platelet signaling transduction defects. Thus, Gas6 is a platelet-response amplifier that plays a significant role in thrombosis. These findings warrant further evaluation of the possible therapeutic use of Gas6 inhibition for prevention of thrombosis.


Subject(s)
Blood Platelets/physiology , Intercellular Signaling Peptides and Proteins , Proteins/physiology , Thrombosis/prevention & control , Animals , Blood Platelets/metabolism , Cell Line , Disease Models, Animal , Female , Gene Expression , Hemostasis , Humans , Male , Mice , Mice, Knockout , Phenotype , Platelet Aggregation , Proteins/genetics , Proteins/immunology , Proteins/pharmacology , Receptors, Cell Surface/genetics , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Thrombosis/etiology
11.
Nat Med ; 6(1): 49-55, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10613823

ABSTRACT

Although tissue injury and inflammation are considered essential for the induction of angiogenesis, the molecular controls of this cascade are mostly unknown. Here we show that a macrophage-derived peptide, PR39, inhibited the ubiquitin-proteasome-dependent degradation of hypoxia-inducible factor-1alpha protein, resulting in accelerated formation of vascular structures in vitro and increased myocardial vasculature in mice. For the latter, coronary flow studies demonstrated that PR39-induced angiogenesis resulted in the production of functional blood vessels. These findings show that PR39 and related compounds can be used as potent inductors of angiogenesis, and that selective inhibition of hypoxia-inducible factor-1alpha degradation may underlie the mechanism of inflammation-induced angiogenesis.


Subject(s)
Antimicrobial Cationic Peptides , DNA-Binding Proteins/metabolism , Endothelium, Vascular/physiology , Myocardial Ischemia/physiopathology , Neovascularization, Physiologic/physiology , Nuclear Proteins/metabolism , Peptides/physiology , Animals , Aorta , Capillaries/drug effects , Capillaries/physiology , Cattle , Cell Hypoxia , Cells, Cultured , Coronary Vessels/drug effects , Cysteine Endopeptidases/metabolism , Endothelium, Vascular/cytology , Heart/physiology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , In Vitro Techniques , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multienzyme Complexes/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/pathology , Peptides/genetics , Peptides/pharmacology , Proteasome Endopeptidase Complex , Recombinant Proteins/metabolism , Swine , Transcription Factors/metabolism , Ubiquitins/metabolism , Umbilical Veins , von Willebrand Factor/genetics
12.
Nat Med ; 4(8): 923-8, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9701244

ABSTRACT

Acquisition of invasive/metastatic potential through protease expression is an essential event in tumor progression. High levels of components of the plasminogen activation system, including urokinase, but paradoxically also its inhibitor, plasminogen activator inhibitor 1 (PAI1), have been correlated with a poor prognosis for some cancers. We report here that deficient PAI1 expression in host mice prevented local invasion and tumor vascularization of transplanted malignant keratinocytes. When this PAI1 deficiency was circumvented by intravenous injection of a replication-defective adenoviral vector expressing human PAI1, invasion and associated angiogenesis were restored. This experimental evidence demonstrates that host-produced PAI is essential for cancer cell invasion and angiogenesis.


Subject(s)
Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic/prevention & control , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/deficiency , Skin Neoplasms/pathology , Adenoviridae , Animals , Cell Transformation, Neoplastic , Cells, Cultured , Disease Progression , Female , Genetic Vectors , Genotype , Humans , Keratinocytes/pathology , Male , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Plasminogen Activator Inhibitor 1/genetics , Skin Neoplasms/blood supply , Transfection
13.
Nat Med ; 7(9): 1021-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533705

ABSTRACT

Deletion of amino-acid residues 1505-1507 (KPQ) in the cardiac SCN5A Na(+) channel causes autosomal dominant prolongation of the electrocardiographic QT interval (long-QT syndrome type 3 or LQT3). Excessive prolongation of the action potential at low heart rates predisposes individuals with LQT3 to fatal arrhythmias, typically at rest or during sleep. Here we report that mice heterozygous for a knock-in KPQ-deletion (SCN5A(Delta/+)) show the essential LQT3 features and spontaneously develop life-threatening polymorphous ventricular arrhythmias. Unexpectedly, sudden accelerations in heart rate or premature beats caused lengthening of the action potential with early afterdepolarization and triggered arrhythmias in Scn5a(Delta/+) mice. Adrenergic agonists normalized the response to rate acceleration in vitro and suppressed arrhythmias upon premature stimulation in vivo. These results show the possible risk of sudden heart-rate accelerations. The Scn5a(Delta/+) mouse with its predisposition for pacing-induced arrhythmia might be useful for the development of new treatments for the LQT3 syndrome.


Subject(s)
Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Sodium Channels/genetics , Adrenergic beta-Agonists/pharmacology , Animals , Arrhythmias, Cardiac/drug therapy , Cardiac Pacing, Artificial , Electrocardiography , Humans , Isoproterenol/pharmacology , Long QT Syndrome/genetics , Membrane Potentials , Mice , Mice, Mutant Strains , Myocardium/cytology , Myocardium/metabolism , NAV1.5 Voltage-Gated Sodium Channel , Sequence Deletion , Sodium/metabolism
14.
Nat Med ; 5(10): 1135-42, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10502816

ABSTRACT

Cardiac rupture is a fatal complication of acute myocardial infarction lacking treatment. Here, acute myocardial infarction resulted in rupture in wild-type mice and in mice lacking tissue-type plasminogen activator, urokinase receptor, matrix metalloproteinase stromelysin-1 or metalloelastase. Instead, deficiency of urokinase-type plasminogen activator (u-PA-/-) completely protected against rupture, whereas lack of gelatinase-B partially protected against rupture. However, u-PA-/- mice showed impaired scar formation and infarct revascularization, even after treatment with vascular endothelial growth factor, and died of cardiac failure due to depressed contractility, arrhythmias and ischemia. Temporary administration of PA inhibitor-1 or the matrix metalloproteinase-inhibitor TIMP-1 completely protected wild-type mice against rupture but did not abort infarct healing, thus constituting a new approach to prevent cardiac rupture after acute myocardial infarction.


Subject(s)
Cardiac Output, Low/etiology , Heart Rupture/etiology , Metalloendopeptidases/antagonists & inhibitors , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Plasminogen Inactivators/therapeutic use , Protease Inhibitors/therapeutic use , Animals , Arrhythmias, Cardiac , Bone Marrow Transplantation , Cell Movement , Collagenases/metabolism , Gene Transfer Techniques , Leukocytes/cytology , Leukocytes/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 9 , Mice , Mice, Mutant Strains , Neovascularization, Physiologic/drug effects , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activators/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
15.
Nat Med ; 7(5): 575-83, 2001 May.
Article in English | MEDLINE | ID: mdl-11329059

ABSTRACT

Vascular endothelial growth factor (VEGF) stimulates angiogenesis by activating VEGF receptor-2 (VEGFR-2). The role of its homolog, placental growth factor (PlGF), remains unknown. Both VEGF and PlGF bind to VEGF receptor-1 (VEGFR-1), but it is unknown whether VEGFR-1, which exists as a soluble or a membrane-bound type, is an inert decoy or a signaling receptor for PlGF during angiogenesis. Here, we report that embryonic angiogenesis in mice was not affected by deficiency of PlGF (Pgf-/-). VEGF-B, another ligand of VEGFR-1, did not rescue development in Pgf-/- mice. However, loss of PlGF impaired angiogenesis, plasma extravasation and collateral growth during ischemia, inflammation, wound healing and cancer. Transplantation of wild-type bone marrow rescued the impaired angiogenesis and collateral growth in Pgf-/- mice, indicating that PlGF might have contributed to vessel growth in the adult by mobilizing bone-marrow-derived cells. The synergism between PlGF and VEGF was specific, as PlGF deficiency impaired the response to VEGF, but not to bFGF or histamine. VEGFR-1 was activated by PlGF, given that anti-VEGFR-1 antibodies and a Src-kinase inhibitor blocked the endothelial response to PlGF or VEGF/PlGF. By upregulating PlGF and the signaling subtype of VEGFR-1, endothelial cells amplify their responsiveness to VEGF during the 'angiogenic switch' in many pathological disorders.


Subject(s)
Capillary Permeability , Endothelial Growth Factors/physiology , Lymphokines/physiology , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic , Pregnancy Proteins/physiology , Animals , Base Sequence , DNA Primers , Embryonic and Fetal Development , Mice , Placenta Growth Factor , Plasma , Pregnancy Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Wound Healing/physiology
16.
J Exp Med ; 185(5): 963-8, 1997 Mar 03.
Article in English | MEDLINE | ID: mdl-9120402

ABSTRACT

The plasminogen/plasmin system has the potential to affect the outcome of inflammatory diseases by regulating accumulation of fibrin and other matrix proteins. In human and experimental crescentic glomerulonephritis (GN), fibrin is an important mediator of glomerular injury and renal impairment. Glomerular deposition of matrix proteins is a feature of progressive disease. To study the role of plasminogen and plasminogen activators in the development of inflammatory glomerular injury, GN was induced in mice in which the genes for these proteins had been disrupted by homologous recombination. Deficiency of plasminogen or combined deficiency of tissue type plasminogen activator (tPA) and urokinase type plasminogen activator (uPA) was associated with severe functional and histological exacerbation of glomerular injury. Deficiency of tPA, the predominant plasminogen activator expressed in glomeruli, also exacerbated disease. uPA deficiency reduced glomerular macrophage infiltration and did not significantly exacerbate disease. uPA receptor deficiency did not effect the expression of GN. These studies demonstrate that plasminogen plays an important role in protecting the glomerulus from acute inflammatory injury and that tPA is the major protective plasminogen activator.


Subject(s)
Glomerulonephritis/etiology , Kidney/pathology , Plasminogen/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Fibrin/metabolism , Glomerulonephritis/immunology , Kidney Glomerulus/pathology , Mice , Mice, Mutant Strains , Plasminogen/genetics , Renal Insufficiency/etiology , Tissue Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/genetics
17.
J Med Genet ; 46(12): 840-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-18413368

ABSTRACT

BACKGROUND: Targeted delivery of the angiogenic factor, vascular endothelial growth factor (VEGF), to motor neurons prolongs survival in rodent models of amyotrophic lateral sclerosis (ALS), while mice expressing reduced VEGF concentrations develop motor neuron degeneration reminiscent of ALS, raising the question whether VEGF contributes to the pathogenesis of ALS. An initial association study reported that VEGF haplotypes conferred increased susceptibility to ALS in humans, but later studies challenged this initial finding. METHODS AND FINDINGS: A meta-analysis was undertaken to critically reappraise whether any of the three common VEGF gene variations (-2578C/A, -1154G/A and -634G/C) increase the risk of ALS. Over 7000 subjects from eight European and three American populations were included in the analysis. Pooled odds ratios were calculated using fixed-effects and random-effects models, and four potential sources of heterogeneity (location of disease onset, gender, age at disease onset and disease duration) were assessed. After correction, none of the genotypes or haplotypes was significantly associated with ALS. Subgroup analysis by gender revealed, however, that the -2578AA genotype, which lowers VEGF expression, increased the risk of ALS in males (OR = 1.46 males vs females; 95% CI = 1.19 to 1.80; p = 7.8 10E-5), even after correction for publication bias and multiple testing. CONCLUSIONS: This meta-analysis does not support the original conclusion that VEGF haplotypes increase the risk of ALS in humans, but the significant association of the low-VEGF -2578AA genotype with increased susceptibility to ALS in males reappraises the link between reduced VEGF concentrations and ALS, as originally revealed by the fortuitous mouse genetic studies.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Vascular Endothelial Growth Factor A/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Heterozygote , Humans , Male , Mice , Motor Neurons/pathology , Polymorphism, Single Nucleotide , Sex Factors
18.
J Cell Biol ; 140(1): 233-45, 1998 Jan 12.
Article in English | MEDLINE | ID: mdl-9425170

ABSTRACT

It has been proposed that the urokinase receptor (u-PAR) is essential for the various biological roles of urokinase-type plasminogen activator (u-PA) in vivo, and that smooth muscle cells require u-PA for migration during arterial neointima formation. The present study was undertaken to evaluate the role of u-PAR during this process in mice with targeted disruption of the u-PAR gene (u-PAR-/-). Surprisingly, u-PAR deficiency did not affect arterial neointima formation, neointimal cell accumulation, or migration of smooth muscle cells. Indeed, topographic analysis of arterial wound healing after electric injury revealed that u-PAR-/- smooth muscle cells, originating from the uninjured borders, migrated over a similar distance and at a similar rate into the necrotic center of the wound as wild-type (u-PAR+/+) smooth muscle cells. In addition, u-PAR deficiency did not impair migration of wounded cultured smooth muscle cells in vitro. There were no genotypic differences in reendothelialization of the vascular wound. The minimal role of u-PAR in smooth muscle cell migration was not because of absent expression, since wild-type smooth muscle cells expressed u-PAR mRNA and functional receptor in vitro and in vivo. Pericellular plasmin proteolysis, evaluated by degradation of 125I-labeled fibrin and activation of zymogen matrix metalloproteinases, was similar for u-PAR-/- and u-PAR+/+ cells. Immunoelectron microscopy of injured arteries in vivo revealed that u-PA was bound on the cell surface of u-PAR+/+ cells, whereas it was present in the pericellular space around u-PAR-/- cells. Taken together, these results suggest that binding of u-PA to u-PAR is not required to provide sufficient pericellular u-PA-mediated plasmin proteolysis to allow cellular migration into a vascular wound.


Subject(s)
Femoral Artery/physiology , Fibrinolysin/metabolism , Metalloendopeptidases/metabolism , Muscle, Smooth, Vascular/physiology , Receptors, Cell Surface/physiology , Tunica Intima/physiology , Urokinase-Type Plasminogen Activator/metabolism , Wound Healing/physiology , Animals , Cells, Cultured , Chemotaxis/genetics , Chemotaxis/physiology , Female , Femoral Artery/cytology , Femoral Artery/injuries , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microscopy, Immunoelectron , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/injuries , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Urokinase Plasminogen Activator , Regeneration , Tunica Intima/cytology , Tunica Intima/injuries , Wound Healing/genetics
19.
J Cell Biol ; 137(4): 953-63, 1997 May 19.
Article in English | MEDLINE | ID: mdl-9151696

ABSTRACT

The middle T antigen of murine Polyomavirus (PymT) rapidly transforms endothelial cells, leading to the formation of vascular tumors in newborn mice. Transformed endothelial (End.) cell lines established from such tumors exhibit altered proteolytic activity as a result of increased expression of urokinase-type plasminogen activator (uPA) and are capable of inducing vascular tumors efficiently when injected into adult mice. In this study we have used mice lacking components of the PA/plasmin system to analyze the role of this system in the transformation process and in tumor growth. We found that the proteolytic status of the host is not a critical determinant for PymT-induced vascular tumor formation. In addition, the lack of either uPA or tissue-type PA (tPA) activity is not limiting for the establishment and proliferation of End. cells in vitro, although the combined loss of both PA activities leads to a marked reduction in proliferation rates. Furthermore, the in vitro morphogenetic properties of mutant End. cells in fibrin gels could only be correlated with an altered proteolytic status in cells lacking both uPA and tPA. However, in contrast with tumors induced by PymT itself, the tumorigenic potential of mutant and wild-type End. cell lines was found to be highly dependent on the proteolytic status of both the tumor cells and the host. Thus, genetic alterations in the PA/plasmin system affect vascular tumor development, indicating that this system is a causal component in PymTmediated oncogenesis.


Subject(s)
Antigens, Polyomavirus Transforming , Cell Transformation, Viral , Endothelium, Vascular/cytology , Fibrinolysin/physiology , Plasminogen Activators/physiology , Vascular Neoplasms/etiology , Animals , Animals, Newborn , Cell Line , Endothelium, Vascular/enzymology , Fibrin , Gels , Gene Expression , Mice , Mice, Knockout , Morphogenesis , Plasminogen Activator Inhibitor 1/deficiency , RNA, Messenger/genetics , Tissue Plasminogen Activator/deficiency , Urokinase-Type Plasminogen Activator/deficiency
20.
J Cell Biol ; 152(4): 777-84, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11266468

ABSTRACT

The plasminogen (Plg)/plasminogen activator (PA) system plays a key role in cancer progression, presumably via mediating extracellular matrix degradation and tumor cell migration. Consequently, urokinase-type PA (uPA)/plasmin antagonists are currently being developed for suppression of tumor growth and angiogenesis. Paradoxically, however, high levels of PA inhibitor 1 (PAI-1) are predictive of a poor prognosis for survival of patients with cancer. We demonstrated previously that PAI-1 promoted tumor angiogenesis, but by an unresolved mechanism. We anticipated that PAI-1 facilitated endothelial cell migration via its known interaction with vitronectin (VN) and integrins. However, using adenoviral gene transfer of PAI-1 mutants, we observed that PAI-1 promoted tumor angiogenesis, not by interacting with VN, but rather by inhibiting proteolytic activity, suggesting that excessive plasmin proteolysis prevents assembly of tumor vessels. Single deficiency of uPA, tissue-type PA (tPA), uPA receptor, or VN, as well as combined deficiencies of uPA and tPA did not impair tumor angiogenesis, whereas lack of Plg reduced it. Overall, these data indicate that plasmin proteolysis, even though essential, must be tightly controlled during tumor angiogenesis, probably to allow vessel stabilization and maturation. These data provide insights into the clinical paradox whereby PAI-1 promotes tumor progression and warrant against the uncontrolled use of uPA/plasmin antagonists as tumor angiogenesis inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endopeptidases/metabolism , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Plasminogen Activator Inhibitor 1/pharmacology , Vitronectin/metabolism , Animals , Endothelium, Vascular/drug effects , Fibrinolysin/metabolism , Keratinocytes/pathology , Mice , Mice, Mutant Strains , Muscle Neoplasms/blood supply , Neoplasm Invasiveness , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/etiology , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Vitronectin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL