Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Hyperthermia ; 41(1): 2331704, 2024.
Article in English | MEDLINE | ID: mdl-38503326

ABSTRACT

OBJECTIVES: To assess the safety and efficacy of radiofrequency ablation (RFA) guidance software that incorporated patient-specific physics-based simulation of each ablation volume. MATERIALS AND METHODS: Patients referred for curative ablation of hepatocellular carcinoma (HCC) of 2-5 cm diameter were prospectively enrolled. RFA was performed under general anesthesia. Procedure planning and intraprocedural modifications were guided by computer simulation of each ablation. The segmented target (tumor with 5 mm margin) was registered to and superimposed on subsequent 3D multiplanar images. The applied RF energy was used to calculate a simulated ablation volume which was displayed relative to the electrode and segmented target, to depict any untreated target tissue. After each additional ablation, the software updated the accumulated simulated ablation volume in relation to the target. The primary endpoints were technical efficacy and rate of local tumor progression (LTP). RESULTS: Sixty-eight tumors were ablated during 57 procedures in 52 patients (68.3 ± 9.2 years old, 78.8% male); 15 (26.3%) had multiple lesions and 23 (39.1%) had prior HCC treatment. The mean tumor diameter was 2.73 (±0.64) cm. The intraprocedural simulation directed additional overlapping ablations in 75.9% of tumors. Technical success and efficacy were 100% at 3-month contrast enhanced CT or MRI follow-up after the single treatment session. Cumulative incidence function estimates for 1- and 2-year LTP were 3.9% and 20.2%, respectively. CONCLUSION: This prospective study found computer-assisted guidance that simulated each ablation was both safe and efficacious. The low rate of LTP was similar to studies that employed stereotactic guidance and ablation confirmation, without requiring a second contrast enhanced study.


Subject(s)
Carcinoma, Hepatocellular , Catheter Ablation , Liver Neoplasms , Radiofrequency Ablation , Humans , Male , Middle Aged , Aged , Female , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/surgery , Liver Neoplasms/pathology , Prospective Studies , Computer Simulation , Catheter Ablation/methods , Radiofrequency Ablation/methods , Treatment Outcome , Retrospective Studies
2.
Pract Radiat Oncol ; 12(1): 28-51, 2022.
Article in English | MEDLINE | ID: mdl-34688956

ABSTRACT

PURPOSE: This guideline provides evidence-based recommendations for the indications and technique-dose of external beam radiation therapy (EBRT) in hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (IHC). METHODS: The American Society for Radiation Oncology convened a task force to address 5 key questions focused on the indications, techniques, and outcomes of EBRT in HCC and IHC. This guideline is intended to cover the definitive, consolidative, salvage, preoperative (including bridge to transplant), and adjuvant settings as well as palliative EBRT for symptomatic primary lesions. Recommendations were based on a systematic literature review and created using a predefined consensus-building methodology and system for grading evidence quality and recommendation strength. RESULTS: Strong recommendations are made for using EBRT as a potential first-line treatment in patients with liver-confined HCC who are not candidates for curative therapy, as consolidative therapy after incomplete response to liver-directed therapies, and as a salvage option for local recurrences. The guideline conditionally recommends EBRT for patients with liver-confined multifocal or unresectable HCC or those with macrovascular invasion, sequenced with systemic or catheter-based therapies. Palliative EBRT is conditionally recommended for symptomatic primary HCC and/or macrovascular tumor thrombi. EBRT is conditionally recommended as a bridge to transplant or before surgery in carefully selected patients. For patients with unresectable IHC, consolidative EBRT with or without chemotherapy should be considered, typically after systemic therapy. Adjuvant EBRT is conditionally recommended for resected IHC with high-risk features. Selection of dose-fractionation regimen and technique should be based on disease extent, disease location, underlying liver function, and available technologies. CONCLUSIONS: The task force has proposed recommendations to inform best clinical practices on the use of EBRT for HCC and IHC with strong emphasis on multidisciplinary care. Future studies should focus on further defining the role of EBRT in the context of liver-directed and systemic therapies and refining optimal regimens and techniques.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Radiation Oncology , Carcinoma, Hepatocellular/radiotherapy , Consensus , Dose Fractionation, Radiation , Humans , Liver Neoplasms/radiotherapy
3.
Hepatology ; 69(4): 1847-1848, 2019 04.
Article in English | MEDLINE | ID: mdl-30734955
5.
JCI Insight ; 2(24)2017 12 21.
Article in English | MEDLINE | ID: mdl-29263300

ABSTRACT

The scaffold protein synectin plays a critical role in the trafficking and regulation of membrane receptor pathways. As platelet-derived growth factor receptor (PDGFR) is essential for hepatic stellate cell (HSC) activation and liver fibrosis, we sought to determine the role of synectin on the PDGFR pathway and development of liver fibrosis. Mice with deletion of synectin from HSC were found to be protected from liver fibrosis. mRNA sequencing revealed that knockdown of synectin in HSC demonstrated reductions in the fibrosis pathway of genes, including PDGFR-ß. Chromatin IP assay of the PDGFR-ß promoter upon synectin knockdown revealed a pattern of histone marks associated with decreased transcription, dependent on p300 histone acetyltransferase. Synectin knockdown was found to downregulate PDGFR-α protein levels, as well, but through an alternative mechanism: protection from autophagic degradation. Site-directed mutagenesis revealed that ubiquitination of specific PDGFR-α lysine residues was responsible for its autophagic degradation. Furthermore, functional studies showed decreased PDGF-dependent migration and proliferation of HSC after synectin knockdown. Finally, human cirrhotic livers demonstrated increased synectin protein levels. This work provides insight into differential transcriptional and posttranslational mechanisms of synectin regulation of PDGFRs, which are critical to fibrogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Liver Cirrhosis/metabolism , Receptors, Platelet-Derived Growth Factor/biosynthesis , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Autophagy/physiology , Cell Movement/physiology , Down-Regulation/physiology , Gene Knockdown Techniques , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatic Stellate Cells/physiology , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Mice, Knockout , Pulmonary Fibrosis/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/physiology , Ubiquitin/metabolism , Up-Regulation/physiology
7.
Lab Invest ; 95(6): 684-96, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25867762

ABSTRACT

Cholangiocytes are the target of a heterogeneous group of liver diseases known as the cholangiopathies. An evolving understanding of the mechanisms driving biliary development provides the theoretical underpinnings for rational development of induced pluripotent stem cell (iPSC)-derived cholangiocytes (iDCs). Therefore, the aims of this study were to develop an approach to generate iDCs and to fully characterize the cells in vitro and in vivo. Human iPSC lines were generated by forced expression of the Yamanaka pluripotency factors. We then pursued a stepwise differentiation strategy toward iDCs, using precise temporal exposure to key biliary morphogens, and we characterized the cells, using a variety of morphologic, molecular, cell biologic, functional, and in vivo approaches. Morphology shows a stepwise phenotypic change toward an epithelial monolayer. Molecular analysis during differentiation shows appropriate enrichment in markers of iPSC, definitive endoderm, hepatic specification, hepatic progenitors, and ultimately cholangiocytes. Immunostaining, western blotting, and flow cytometry demonstrate enrichment of multiple functionally relevant biliary proteins. RNA sequencing reveals that the transcriptome moves progressively toward that of human cholangiocytes. iDCs generate intracellular calcium signaling in response to ATP, form intact primary cilia, and self-assemble into duct-like structures in three-dimensional culture. In vivo, the cells engraft within mouse liver, following retrograde intrabiliary infusion. In summary, we have developed a novel approach to generate mature cholangiocytes from iPSCs. In addition to providing a model of biliary differentiation, iDCs represent a platform for in vitro disease modeling, pharmacologic testing, and individualized, cell-based, regenerative therapies for the cholangiopathies.


Subject(s)
Bile Ducts/cytology , Epithelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Animals , Bile Ducts/chemistry , Bile Ducts/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Calcium Signaling , Cell Differentiation , Cell Engineering , Cell Line , Epithelial Cells/chemistry , Epithelial Cells/metabolism , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/metabolism , Liver/chemistry , Liver/cytology , Liver/metabolism , Mice , Real-Time Polymerase Chain Reaction
8.
Am J Physiol Cell Physiol ; 307(7): C622-33, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25080486

ABSTRACT

Hepatic stellate cells (HSCs) generate matrix, which in turn may also regulate HSCs function during liver fibrosis. We hypothesized that HSCs may endocytose matrix proteins to sense and respond to changes in microenvironment. Primary human HSCs, LX2, or mouse embryonic fibroblasts (MEFs) [wild-type; c-abl(-/-); or Yes, Src, and Fyn knockout mice (YSF(-/-))] were incubated with fluorescent-labeled collagen or gelatin. Fluorescence-activated cell sorting analysis and confocal microscopy were used for measuring cellular internalization of matrix proteins. Targeted PCR array and quantitative real-time PCR were used to evaluate gene expression changes. HSCs and LX2 cells endocytose collagens in a concentration- and time-dependent manner. Endocytosed collagen colocalized with Dextran 10K, a marker of macropinocytosis, and 5-ethylisopropyl amiloride, an inhibitor of macropinocytosis, reduced collagen internalization by 46%. Cytochalasin D and ML7 blocked collagen internalization by 47% and 45%, respectively, indicating that actin and myosin are critical for collagen endocytosis. Wortmannin and AKT inhibitor blocked collagen internalization by 70% and 89%, respectively, indicating that matrix macropinocytosis requires phosphoinositide-3-kinase (PI3K)/AKT signaling. Overexpression of dominant-negative dynamin-2 K44A blocked matrix internalization by 77%, indicating a role for dynamin-2 in matrix macropinocytosis. Whereas c-abl(-/-) MEF showed impaired matrix endocytosis, YSF(-/-) MEF surprisingly showed increased matrix endocytosis. It was also associated with complex gene regulations that related with matrix dynamics, including increased matrix metalloproteinase 9 (MMP-9) mRNA levels and zymographic activity. HSCs endocytose matrix proteins through macropinocytosis that requires a signaling network composed of PI3K/AKT, dynamin-2, and c-abl. Interaction with extracellular matrix regulates matrix dynamics through modulating multiple gene expressions including MMP-9.


Subject(s)
Collagen/metabolism , Endocytosis , Extracellular Matrix/metabolism , Hepatic Stellate Cells/metabolism , Actins/metabolism , Animals , Cells, Cultured , Cellular Microenvironment , Dynamin II/metabolism , Fibrosis , Gene Expression Regulation , Genes, abl , Genes, src , Hepatic Stellate Cells/pathology , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinase/metabolism , Pinocytosis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fyn/deficiency , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-yes/deficiency , Proto-Oncogene Proteins c-yes/genetics , Time Factors
9.
Clin Liver Dis (Hoboken) ; 2(2): 80-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-30992830
10.
Circ Res ; 104(3): 337-45, 2009 Feb 13.
Article in English | MEDLINE | ID: mdl-19122176

ABSTRACT

Plaque vascularity has been implicated in its growth and stability. However, there is a paucity of information regarding the origin of plaque vasculature and the role of vasa vasorum in plaque growth. To inhibit growth of vasa vasorum in atherogenic mice and assess its effect on plaque growth, we used a truncated plasminogen activator inhibitor (PAI)-1 protein, rPAI-1(23), that has significant antiangiogenic activity. Female LDLR(-/-)ApoB-48-deficient mice fed Paigen's diet without cholate for 20 weeks received rPAI-1(23) treatment (n=21) for the last 6 weeks. Plaque size and vasa vasorum density were compared to 2 controls: mice fed Paigen's diet and treated with saline for the last 6 weeks (n=16) and mice fed Paigen's diet until the onset of treatment (n=14). The rPAI-1(23) treatment significantly reduced plaque area and plaque cholesterol in the descending aorta and plaque area in the innominate artery. Measurements of reconstructed confocal microscopy images of vasa vasorum demonstrate that rPAI-1(23) treatment decreased vasa vasorum area and length, which was supported by microCT images. Confocal images provide evidence for vascularized plaque in the saline-treated group but not in rPAI-1(23)-treated mice. The increased vessel density in saline-treated mice is attributable, in part, to upregulated fibroblast growth factor-2 expression, which is inhibited by rPAI-1(23). In conclusion, rPAI-1(23) inhibits growth of vasa vasorum, as well as vessels within the adjacent plaque and vessel wall, through inhibition of fibroblast growth factor-2, leading to reduced plaque growth in atherogenic female LDLR(-/-)ApoB-48-deficient mice.


Subject(s)
Angiogenesis Inhibitors/physiology , Atherosclerosis/prevention & control , Plasminogen Activator Inhibitor 1/physiology , Vasa Vasorum/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Apolipoprotein B-48/genetics , Arteries/pathology , Atherosclerosis/pathology , Female , Fibroblast Growth Factor 2/genetics , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Microscopy, Confocal , Peptide Fragments/pharmacology , Peptide Fragments/physiology , Plasminogen Activator Inhibitor 1/pharmacology , Receptors, LDL/genetics , Recombinant Proteins/pharmacology , Vasa Vasorum/growth & development , Vasa Vasorum/pathology , Vascular Endothelial Growth Factor A/genetics
11.
Arthritis Rheum ; 56(10): 3448-58, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17907150

ABSTRACT

OBJECTIVE: Systemic sclerosis (SSc; scleroderma) is a systemic connective tissue disease with an extensive vascular component that includes aberrant microvasculature and impaired wound healing. The aim of this study was to investigate the presence of antiangiogenic factors in patients with SSc. METHODS: Plasma samples were obtained from 30 patients with SSc and from 10 control patients without SSc. The samples were analyzed for the ability of plasma to affect endothelial cell migration and vascular structure formation and for the presence of antiangiogenic activity. RESULTS: Exposure of normal human microvascular dermal endothelial cells to plasma from patients with SSc resulted in decreased cell migration (mean +/- SEM 52 +/- 5%) and tube formation (34 +/- 6%) compared with that in plasma from control patients (P < 0.001 for both). SSc plasma contained 2.9-fold more plasminogen kringle 1-3 fragments (angiostatin) than that in control plasma. The addition of angiostatin to control plasma resulted in inhibition of endothelial cell migration and proliferation similar to that observed in SSc plasma. In vitro studies demonstrated that granzyme B and other proteases contained in T cell granule content cleave plasminogen and plasmin into angiostatin fragments. CONCLUSION: Plasminogen conformation in patients with SSc enables granzyme B and granule content protease to limit the proangiogenic effects of plasmin and increase the levels of antiangiogenic angiostatin. This increase in angiostatin production may account for some of the vascular defects observed in patients with SSc.


Subject(s)
Angiogenesis Inhibitors/metabolism , Endothelial Cells/metabolism , Scleroderma, Systemic/physiopathology , Adult , Angiogenesis Inhibitors/blood , Cell Movement/physiology , Female , Granzymes/metabolism , Humans , In Vitro Techniques , Male , Middle Aged , Scleroderma, Systemic/blood , Wound Healing/physiology
12.
J Biol Chem ; 281(44): 33336-44, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16950776

ABSTRACT

Many angiogenesis inhibitors are breakdown products of endogenous extracellular matrix proteins. Plasmin and matrix metalloproteinase-3 generate breakdown products of matrix-bound plasminogen activator inhibitor-1 (PAI-1). We produced a truncated form of PAI-1, rPAI-1(23), that possesses significant anti-angiogenic activity and stimulates high levels of apoptosis in quiescent arterial endothelial cells. Quiescent endothelial cells are less susceptible to apoptosis than angiogenic endothelial cells. The present study was designed to determine the mechanism of the rPAI-1(23) effects in bovine aortic endothelial cells. Apoptosis was measured in annexin V and caspase 3 assays. Expression of death and survival signaling molecules were examined by Western blot and kinase activity. Fibroblast growth factor 2 (FGF2) functions were analyzed in angiogenesis assays. The early response to rPAI-1(23) was an increase in annexin V-positive cells and phosphorylated (p) JNK isoform expression followed by an increase in p-Akt and p-c-Jun expression. Caspase 3 was activated at 4 h, whereas p-Akt was reduced to control levels. By 6 h of rPAI-1(23) treatment cell number was reduced by 35%, and p-c-Jun and p-JNK were degraded by proteasomes. Confocal microscopic images showed increased amounts of FGF2 in the extracellular matrix. However, rPAI-1(23) blocked FGF2 signaling through FGF receptor 1 and syndecan-4, inhibiting cell migration, tubulogenesis, and proliferation. Exogenous FGF2 stimulation could not reverse these effects. We conclude that rPAI-1(23) stimulation of apoptosis in BAEC triggers a cascade of death versus survival events that includes release of FGF2. The rPAI-1(23) anti-angiogenic activity inhibits FGF2 pro-angiogenic functions by blocking FGF2 signaling through FGF receptor 1 and syndecan-4 and downstream effectors p-Akt, p-JNK, and p-c-Jun.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Fibroblast Growth Factor 2/metabolism , Peptide Fragments/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Animals , Apoptosis/drug effects , Cattle , Cells, Cultured , Chickens , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/pharmacology , Signal Transduction
13.
Arterioscler Thromb Vasc Biol ; 26(3): 520-6, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16397137

ABSTRACT

OBJECTIVE: To assess the importance of genetic background for collateral artery development. METHODS AND RESULTS: C57BL/6, BALB/c and 129S2/Sv mice were studied after femoral artery ligation by laser Doppler imaging, visible light oximetry, time-of-flight-magnetic resonance imaging, and treadmill testing; C57BL/6 and BALB/c also underwent electron paramagnetic resonance (EPR) oximetry, x-ray angiography, and histology. C57BL/6 had the least initial distal ischemia and most complete recovery. BALB/c had the most severe initial ischemia and poorest recovery. BALB/c had some vasodilatory reserve in their ligated limbs not seen in the other strains at 3 weeks. By in vivo TOF-magnetic resonance angiography, C57BL/6 had larger preexistent and developed collaterals. By x-ray angiography, C57BL/6 had a higher collateral-dependent filling score and number of visible collaterals immediately after femoral ligation and a higher number of visible collaterals at 1 week but not at 4 weeks. EPR oximetry and histology revealed hypoxia and tissue damage in regions of collateral growth of BALB/c but not C57BL/6 mice. In C57BL/6 BrdUrd uptake in the thigh was limited to larger vessels and isolated perivascular cells. Proliferative activity in collateral arterioles was similar in both strains. CONCLUSIONS: Genetic differences in preexistent collateral vasculature can profoundly affect outcome and milieu for compensatory collateral artery growth after femoral artery occlusion.


Subject(s)
Disease Models, Animal , Ischemia/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Physiologic/genetics , Animals , Electron Spin Resonance Spectroscopy , Femoral Artery , Hindlimb/blood supply , Hyperemia/genetics , Hyperemia/pathology , Hyperemia/physiopathology , Ischemia/pathology , Ischemia/physiopathology , Ligation , Magnetic Resonance Angiography , Male , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Organ Size , Oximetry , Oxyhemoglobins/metabolism , Species Specificity
14.
Proc Natl Acad Sci U S A ; 102(31): 10999-1004, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16043715

ABSTRACT

The genetic loss of endothelial-derived nitric oxide synthase (eNOS) in mice impairs vascular endothelial growth factor (VEGF) and ischemia-initiated blood flow recovery resulting in critical limb ischemia. This result may occur through impaired arteriogenesis, angiogenesis, or mobilization of stem and progenitor cells. Here, we show that after ischemic challenge, eNOS knockout mice [eNOS (-/-)] have defects in arteriogenesis and functional blood flow reserve after muscle stimulation and pericyte recruitment, but no impairment in endothelial progenitor cell recruitment. More importantly, the defects in blood flow recovery, clinical manifestations of ischemia, ischemic reserve capacity, and pericyte recruitment into the growing neovasculature can be rescued by local intramuscular delivery of an adenovirus encoding a constitutively active allele of eNOS, eNOS S1179D, but not a control virus. Collectively, our data suggest that endogenous eNOS-derived NO exerts direct effects in preserving blood flow, thereby promoting arteriogenesis, angiogenesis, and mural cell recruitment to immature angiogenic sprouts.


Subject(s)
Ischemia/enzymology , Nitric Oxide Synthase/physiology , Animals , Extremities/blood supply , Gene Expression , Gene Transfer Techniques , Ischemia/pathology , Ischemia/physiopathology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/blood supply , Neovascularization, Pathologic , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Pericytes/pathology , Regional Blood Flow
15.
Am J Physiol Cell Physiol ; 287(4): C1012-22, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15201140

ABSTRACT

The transforming growth factor (TGF) family of secretory polypeptides comprises signaling proteins involved in numerous physiological processes, including vascular development and vessel wall integrity. Both pro- and anti-angiogenic effects of TGF-beta1 have also been documented. To study the intracellular mechanisms involved in capillary tube morphogenesis, endothelial cell aggregates were cultured in a fibrin matrix. It was found that the pattern of capillary tubes formed in a fibrin matrix was altered in response to TGF-beta1 treatment such that the capillary-like structures displayed a bipolarized pattern. In contrast, in untreated control and fibroblast growth factor-2-treated cells, the pattern of capillary tubes formed was random. TGF-beta1 also downregulated urokinase-type plasminogen activator (uPA) activity while upregulating PA inhibitor (PAI)-1 and thrombospondin (TSP)1 gene expression. To investigate the signaling cascade mediating the phenotypic changes observed, pharmacological inhibitors of p38 MAPK, Sp1 transcription factor, c-Jun NH(2)-terminal kinase (JNK), and the cytokine TNF-alpha were used. The p38 MAPK inhibitor SB203580 reversed the TGF-beta1-dependent inhibition of uPA activity but not its morphogenetic effect. In contrast, the DNA intercalator WP631 and TNF-alpha counteracted the TGF-beta1-induced morphogenetic effect while the JNK inhibitor SP600125 effectively inhibited capillary tube formation. These results indicate that the TGF-beta1-induced capillary tube pattern is independent of the p38 MAPK-activated PAI-1 and TSP1 expression, but the mechanism involves Sp1-dependent transcriptional regulation. The results also raise the possibility that the JNK pathway, which controls convergent extension in Xenopus, may be involved in vessel wall patterning in mammalian systems.


Subject(s)
Capillaries/metabolism , Endothelial Cells/metabolism , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/metabolism , Neovascularization, Physiologic , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , Animals , Blotting, Northern , Blotting, Western , Body Patterning , Capillaries/drug effects , Cattle , Cell Aggregation , Cell Movement , Cells, Cultured , Endothelial Cells/drug effects , Fibrin , Image Processing, Computer-Assisted , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinases/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thrombospondin 1/metabolism , Transforming Growth Factor beta1 , p38 Mitogen-Activated Protein Kinases
16.
J Biol Chem ; 277(50): 49077-89, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12381729

ABSTRACT

We have made deletions of the porcine plasminogen activator inhibitor-1 (PAI-1) gene to obtain recombinant truncated PAI-1 proteins to examine functions of the PAI-1 isoforms. We previously reported that one recombinant truncated protein, rPAI-1(23), induces the formation of angiostatin by cleaving plasmin. The rPAI-1(23) protein is also able to bind urokinase plasminogen activator and plasminogen and then reduce the amount of plasmin that is formed. We have now prepared three different truncated rPAI-1 proteins and demonstrate that PAI-1 conformations control the release of heparin-binding vascular endothelial growth factor (VEGF) isoforms. The rPAI-1(23) isoform can regulate the functional activity of heparan sulfate-binding VEGF-A isoforms by blocking the activation of VEGF from heparan sulfate. The rPAI-1(23) conformation induced extensive apoptosis in cultured endothelial cells and thus reduced the number of proliferating cells. The rPAI-1(23) isoform inhibited migration of VEGF-stimulated sprouting from chick aortic rings by 65%, thus displaying a role in anti-angiogenic mechanisms. This insight into anti-angiogenic functions related to PAI-1 conformational changes could provide potential intervention points in angiogenesis associated with atherosclerotic plaques and cancer.


Subject(s)
Endothelial Growth Factors/antagonists & inhibitors , Heparin/metabolism , Lymphokines/antagonists & inhibitors , Plasminogen Activator Inhibitor 1/physiology , Animals , Apoptosis , Base Sequence , Cattle , Cells, Cultured , Chick Embryo , Culture Media , DNA Primers , Endothelial Growth Factors/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Plasminogen/metabolism , Plasminogen Activator Inhibitor 1/chemistry , Protein Binding , Swine , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...