Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Public Health ; 215: 124-130, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36725155

ABSTRACT

OBJECTIVE: To summarise the evidence on the impacts of gambling-related advertising that could lead to gambling-related harm, including impacts on vulnerable individuals and inequalities in the distribution of harms. STUDY DESIGN: An umbrella review of studies investigating the impact of gambling advertising. METHODS: A review was undertaken of systematic reviews of qualitative, quantitative and mixed method studies reporting outcomes associated with gambling advertising and marketing. The search strategy included database searches (Web of Science, PsycInfo) and website searches. The quality of the included reviews was determined using A MeaSurement Tool to Assess systematic Reviews 2. RESULTS: 1024 papers were identified by database searches. Eight systematic reviews, including 74 unique studies, met inclusion criteria. Included studies, using quantitative and qualitative methods, consistently support the existence of a causal relationship between exposure to advertising of gambling products/brands and more positive attitudes to gambling, greater intentions to gamble and increased gambling activity at both individual and population level. There is evidence of a 'dose-response' effect; greater advertising exposure increases participation which leads to a greater risk of harm. There was more evidence for the impact on children and young people and for those already at risk from current gambling activity with those most vulnerable more likely to be influenced. CONCLUSION: Gambling advertising restrictions could reduce overall harm and mitigate the impact of advertising on gambling-related inequalities. Public health harm prevention strategies should include policies which limit exposure to advertising, particularly among children and vulnerable groups.


Subject(s)
Advertising , Gambling , Adolescent , Child , Humans , Gambling/prevention & control , Marketing , Policy , Systematic Reviews as Topic
2.
Br J Ophthalmol ; 90(2): 175-80, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16424529

ABSTRACT

AIMS: To prospectively study the evolution of possible high risk features of conjunctival filtration blebs like avascularity, transconjunctival oozing (TCO), and leaks after mitomycin C (MMC) enhanced glaucoma surgery. METHODS: Single observer, 2 year prospective study on bleb characteristics of 125 eyes of 125 consecutive patients who had MMC augmented glaucoma surgery with initially successful filtration. MMC (0.2 mg/ml for 2 minutes in most cases) was applied on the area of the scleral flap before dissection. Glaucoma surgeries included were trabeculectomy, deep sclerectomy, and combined procedures. A dry fluorescein strip was applied on the avascular part of the bleb and observed for aqueous egress with flow (point leak, PL) or without (TCO). RESULTS: The mean time (95% CI) for observing bleb avascularity, TCO, and bleb leaks were 106 days (69 to 143), 208 days (155 to 261), and 609 days (559 to 659), respectively. Bleb leaks were observed in 17 eyes (13.6%)-15 (24.6%) in the trabeculectomy group and two (3.1%) in the deep sclerectomy group (p = 0.003). Kaplan-Meier survival analyses showed that the probability of observing bleb avascularity at sixth, 12th, and 24th month after surgery was 56%, 71%, and 73%, respectively. In eyes with avascular blebs, the probability of developing TCO and leaks was 77% and 1% at 6 months, 81% and 12% at 12 months, and 95% and 26% at 24 months, respectively. Cox's regression analyses and log rank tests showed that eyes with larger avascular blebs (>4 mm) were more likely to develop TCO (hazard ratio 3.77, p = 0.001) and delayed bleb leaks were more likely to be seen in eyes of the trabeculectomy group rather than the deep sclerectomy group (hazard ratio 0.06, p = 0.0006). CONCLUSIONS: MMC application over the area of scleral flap dissection during glaucoma surgery is associated with a high incidence of bleb avascularity, TCO, and delayed bleb leaks. Most eyes developed bleb avascularity within the first year after surgery. TCO will eventually be seen in all eyes with avascular blebs and the incidence of leaks gradually increases with time. This study shows that patients with eyes undergoing glaucoma surgery with MMC and avascular blebs should be monitored indefinitely.


Subject(s)
Blister/chemically induced , Conjunctival Diseases/chemically induced , Glaucoma/surgery , Mitomycin/adverse effects , Aged , Blister/pathology , Conjunctiva/blood supply , Conjunctival Diseases/pathology , Female , Fluorouracil/administration & dosage , Glaucoma/pathology , Humans , Intraocular Pressure/drug effects , Male , Postoperative Complications , Prospective Studies , Sclera/surgery , Survival Analysis , Time Factors , Trabeculectomy/methods , Treatment Outcome , Wound Healing/drug effects
3.
Br J Ophthalmol ; 89(6): 694-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923504

ABSTRACT

AIMS: To compare outcomes of phacoemulsification combined with trabeculectomy (PT) or deep sclerectomy (PDS) with intraoperative mitomycin C (MMC) application. METHODS: Non-randomised, consecutive, retrospective comparative study. 97 eyes of 97 patients (59 PDS, 38 PT) undergoing combined surgery with intraoperative MMC (0.1-0.4 mg/ml for 1-3 minutes) were identified for inclusion in the study. RESULTS: The probability of maintaining intraocular pressure (IOP) below 19 mm Hg and 15 mm Hg, with a 30% drop from preoperative IOP and without additional medication, 1 year after surgery were 77.6% (95% CI: 67 to 90) and 71.5% (60 to 85) for the PDS group and 89.5% (80 to 99) and 89.5 (80 to 99) for the PT group, respectively, and these differences were not statistically significant (p>0.05, log rank test). After excluding ocular co-morbidity no differences were observed in the improvement of visual acuity between the two groups. There were no major differences in the complication rates except that delayed bleb leaks were seen in seven eyes (18.4%) of the PT group (p = 0.004). CONCLUSION: In this study, no statistically significant difference was found in the IOP and visual outcomes between PDS and PT. A significantly higher frequency of late bleb leaks after PT was observed.


Subject(s)
Glaucoma/surgery , Mitomycin/therapeutic use , Phacoemulsification , Sclera/surgery , Trabeculectomy , Aged , Cataract/complications , Combined Modality Therapy , Female , Glaucoma/complications , Humans , Intraocular Pressure , Intraoperative Care/methods , Male , Nucleic Acid Synthesis Inhibitors/therapeutic use , Phacoemulsification/adverse effects , Postoperative Care/methods , Retrospective Studies , Survival Analysis , Trabeculectomy/adverse effects , Treatment Outcome , Visual Acuity
4.
Arterioscler Thromb Vasc Biol ; 21(6): 949-54, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11397702

ABSTRACT

Cultured arterial smooth muscle cells (SMCs) with distinct phenotypic features have been described by several laboratories; however, it is not presently known whether this phenotypic heterogeneity can be maintained within an in vivo environment. To answer this question, we have seeded into the intima of denuded rat carotid artery 2 SMC populations with well-established distinct biological features, ie, spindle-shaped, not growing in the absence of serum, and well differentiated versus epithelioid, growing in the absence of serum, and relatively undifferentiated, derived from the aortic media of newborn rats (aged 4 days) and old rats (aged >18 months), respectively. We show that these 2 populations maintain their distinct biochemical features (ie, expression of alpha-smooth muscle actin, smooth muscle myosin heavy chains, and cellular retinol binding protein-1) in the in vivo environment. The old rat media-derived SMCs continue to produce cellular retinol binding protein-1 but little alpha-smooth muscle actin and smooth muscle myosin heavy chains, whereas the newborn rat media-derived SMCs continue to express alpha-smooth muscle actin and smooth muscle myosin heavy chains but no cellular retinol binding protein-1. Our results reinforce the notion of arterial SMC phenotypic heterogeneity and suggest that in our model, heterogeneity is controlled genetically and not by the local environment.


Subject(s)
Arteries/cytology , Carotid Artery Injuries/surgery , Muscle, Smooth, Vascular/transplantation , Actins/metabolism , Animals , Animals, Newborn , Arteriosclerosis/metabolism , Arteriosclerosis/surgery , Carotid Artery Injuries/metabolism , Cells, Cultured , Male , Muscle, Smooth, Vascular/metabolism , Myosin Heavy Chains/metabolism , Phenotype , Rats , Rats, Inbred F344 , Retinol-Binding Proteins/metabolism , Retinol-Binding Proteins, Cellular
5.
Circ Res ; 86(6): 676-83, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10747004

ABSTRACT

Decorin is an extracellular matrix (ECM) proteoglycan that may modify vascular smooth muscle cell (SMC) function by altering the response to growth factors and the accumulation of ECM proteins during vascular injury. To investigate these possibilities in vivo, decorin was overexpressed at the site of arterial injury by cell-mediated gene transfer. Fischer rat SMCs were transduced in vitro with a retroviral construct that contained the bovine decorin gene and were subsequently seeded into injured rat carotid arteries. A species-specific antibody to bovine decorin and polymerase chain reaction primers were used to detect bovine decorin and distinguish it from endogenous rat decorin. Immunohistochemical and Northern analyses of rat carotid arteries revealed only low levels of rat decorin expression up to 8 weeks after balloon injury. However, after cell-mediated transfer of bovine decorin, strong expression of bovine decorin was verified by immunohistochemistry and reverse transcriptase-polymerase chain reaction. Four weeks after injury, the intimal area in vessels seeded with bovine decorin-overexpressing SMCs was significantly reduced by 35+/-4% (mean+/-SEM, n=9; P<0.01). Decorin overexpression also induced a higher intimal nuclear density and decreased volume of ECM. Specifically, immunostaining for versican and fibronectin was markedly reduced. In contrast, immunostaining for collagen type I was increased, and electron microscopy confirmed that collagen accumulation was altered. Bromodeoxyuridine labeling indicated that intimal SMC proliferation was not affected by the expression of bovine decorin. In summary, we demonstrate that gene transfer of the ECM proteoglycan, decorin, into the injured arterial wall reduces intimal ECM volume and alters the composition of the ECM.


Subject(s)
Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Catheterization/adverse effects , Proteoglycans/metabolism , Tunica Intima/metabolism , Tunica Intima/pathology , Animals , Cattle , Cells, Cultured , Decorin , Extracellular Matrix/metabolism , Extracellular Matrix Proteins , Gene Transfer Techniques , Muscle, Smooth, Vascular/metabolism , Proteoglycans/genetics , Rats , Rats, Inbred F344
6.
J Clin Invest ; 102(7): 1413-20, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9769334

ABSTRACT

Although matrix metalloproteinases (MMPs) are expressed in abundance in arterial aneurysms, their contribution to arterial wall degeneration, dilation, and rupture has not been determined. We investigated MMP function in a rat model of aneurysm associated with arterial dilation, elastin loss, medial invasion by mononuclear inflammatory cells, and MMP upregulation. Rupture was correlated with increased gelatinase B (MMP-9) and activated gelatinase A (MMP-2). Syngeneic rat smooth muscle cells retrovirally transfected with tissue inhibitor of matrix metalloproteinases (TIMP)-1 cDNA (LTSN) or with the vector alone as a control (LXSN) were seeded onto the luminal surface of the vessels. The seeding of LTSN cells resulted in TIMP-1 local overexpression. The seeding with LTSN cells, but not LXSN cells, decreased MMP-9, activated MMP-2 and 28-kD caseinase and elastase activity, preserved elastin in the media, and prevented aneurysmal degeneration and rupture. We conclude that MMP overexpression is responsible for aneurysmal degeneration and rupture in this rat model and that local pharmacological blockade might be a reasonable strategy for controlling the formation of aneurysms in humans.


Subject(s)
Aorta, Abdominal/physiology , Aortic Aneurysm, Abdominal/physiopathology , Aortic Rupture/physiopathology , Muscle, Smooth, Vascular/physiology , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/transplantation , Aortic Rupture/prevention & control , Collagenases/metabolism , Desmosine/analysis , Elastin/analysis , Gelatinases/metabolism , Guinea Pigs , Male , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Metalloendopeptidases/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/transplantation , Rats , Rats, Inbred F344 , Recombinant Proteins/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-3/biosynthesis , Transfection , Transplantation, Heterologous
7.
Circulation ; 98(3): 249-55, 1998 Jul 21.
Article in English | MEDLINE | ID: mdl-9697825

ABSTRACT

BACKGROUND: Arterial aneurysms exhibit a loss of elastin and an increase in the plasminogen activators urokinase plasminogen activator (u-PA) and tissue plasminogen activator (t-PA). Because u-PA, t-PA, and plasmin have a limited proteolytic activity against elastin, the role of plasminogen activators in the aneurysmal disease is unclear. To investigate this question, we overexpressed plasminogen activator inhibitor-1 (PAI-1), an inhibitor of t-PA and u-PA, in a rat model of aortic aneurysm. METHODS AND RESULTS: Guinea pig-to-rat aortic xenografts were seeded with syngeneic Fischer 344 rat smooth muscle cells retrovirally transduced with the rat PAI-1 gene (LPSN group) or the vector alone (LXSN group). Some grafts were not seeded with cells (NO group). Western blots showed increased PAI-1 in grafts from the LPSN group compared with LXSN and NO groups. All grafts in the NO group (n=8) and 40% in the LXSN group ruptured between days 4 and 14. At 4 weeks in the LXSN group, the remaining unruptured grafts (n=6) were aneurysmal (diameter increase > or =100%), whereas in the LPSN group (n=6) none of the grafts had ruptured or were aneurysmal. Elastin was preserved in the LPSN group. t-PA, the major PA expressed in the model, was decreased in the LPSN group compared with the other groups, as determined by zymography. Quantitative zymography showed decreased levels of two matrix metalloproteinases (MMPs), a 28-kD caseinase, and activated MMP-9 in the LPSN group. CONCLUSIONS: The blockade of plasminogen activators prevents formation of aneurysms and arterial rupture by inhibiting MMP activation.


Subject(s)
Aortic Aneurysm/prevention & control , Aortic Rupture/prevention & control , Plasminogen Activator Inhibitor 1/metabolism , Animals , Aorta/transplantation , Aortic Aneurysm/etiology , Desmosine/metabolism , Elastin/metabolism , Guinea Pigs , Immunization , Male , Metalloendopeptidases/metabolism , Plasminogen Activators/metabolism , Rats , Rats, Inbred F344 , Time Factors , Tissue Plasminogen Activator/metabolism , Transplantation, Heterologous
8.
Circ Res ; 82(8): 862-70, 1998 May 04.
Article in English | MEDLINE | ID: mdl-9576106

ABSTRACT

Endothelial cells in normal blood vessels might prevent the unscheduled proliferation of smooth muscle cells (SMCs) by the expression of cell migration and growth inhibitors. NO, a potent vasodilator, generated by endothelium-specific constitutive NO synthase (ecNOS) might be such an inhibitor. To test this hypothesis, we overexpressed human ecNOS in syngeneic rat arterial SMCs using retrovirus-mediated gene transfer. Compared with SMCs transduced with vector alone (LXSN SMCs), DNA synthesis and cell proliferation were inhibited in the ecNOS-expressing SMCs (LCNSN SMCs). Basal and stimulated (by the calcium ionophore A23187) secretion of NO and intracellular cGMP were increased in LCNSN SMCs. Nomega-Nitro-L-arginine (L-NA), an inhibitor of NO synthesis, enhanced the proliferation of LCNSN SMCs but had no effect on LXSN SMCs. LCNSN SMCs seeded onto the luminal surface of balloon-injured rat carotid arteries inhibited neointimal formation by 37% and induced marked dilatation (3-fold increase in vessel diameter) at 2 weeks compared with LXSN SMC-seeded arteries. Orally administered L-NA blocked these changes. Phosphorylation of vasodilator-stimulated phosphoprotein, which is regulated in part by NO, was elevated in LCNSN SMCs and in LCNSN SMC-seeded arteries. This study demonstrates that NO generation by ecNOS inhibits SMC proliferation in vitro and modulates vascular tone locally in vivo.


Subject(s)
Angioplasty, Balloon , Carotid Arteries/physiology , Carotid Artery Injuries , Endothelium, Vascular/enzymology , Muscle, Smooth, Vascular/physiology , Nitric Oxide Synthase/biosynthesis , Animals , Aorta , Calcimycin/pharmacology , Carotid Arteries/cytology , Cell Adhesion Molecules/metabolism , Cell Division , Cells, Cultured , Cyclic GMP/metabolism , Gene Transfer Techniques , Humans , Immunohistochemistry , Isoenzymes/biosynthesis , Kinetics , Male , Microfilament Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/injuries , Nitric Oxide Synthase Type III , Nitroarginine/pharmacology , Phosphoproteins/metabolism , Rats , Rats, Inbred F344 , Recombinant Proteins/biosynthesis , Retroviridae , Transfection
9.
Arterioscler Thromb Vasc Biol ; 18(5): 803-7, 1998 May.
Article in English | MEDLINE | ID: mdl-9598840

ABSTRACT

We have recently demonstrated that the blockade of matrix metalloproteinases by local overexpression of the intrinsic inhibitor tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) reduces intimal hyperplasia. We now report a major change in the elastin content of the intima of rat carotid arteries seeded with TIMP-1-overexpressing smooth muscle cells. To understand the mechanism responsible for elastin accumulation, synthesis and degradation of elastin in TIMP-1 and control cell-seeded rats were measured. There were no differences in elastin mRNA or elastin synthesis, as documented by 14[C]proline incorporation between TIMP-1 and control cell-seeded arteries. In contrast, there was an increase in cross-linked elastin in the TIMP-1 group. In addition, in TIMP-1 and control rats, an elastase activity of approximately 28 kD was detected by elastin zymography and was decreased in TIMP-1 cell-seeded vessels. The 28 kD elastolytic activity was inhibited by exogenously added TIMP-1 and EDTA but not by PMSF, suggesting that it was a metalloelastase. Therefore, we have demonstrated that a shift of the proteolytic balance toward protease inhibition by TIMP-1 overexpression does not change elastin synthesis but rather changes posttranslational processing, resulting in increased elastin accumulation.


Subject(s)
Carotid Arteries/metabolism , Elastin/biosynthesis , Metalloendopeptidases/metabolism , Protease Inhibitors/metabolism , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Animals , Carotid Arteries/pathology , Carotid Artery Injuries , Elastin/genetics , Pancreatic Elastase/metabolism , Papio , RNA, Messenger , Rats , Rats, Inbred F344
10.
J Surg Res ; 70(2): 178-86, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9245569

ABSTRACT

UNLABELLED: Arterial smooth muscle cell (SMC) proliferation is an important factor in the development of atherosclerotic plaques and restenotic lesions following arterial reconstruction. Basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF), and thrombin are known to induce SMC proliferation and migration in vitro and in vivo. In cultured cells the proliferative responses to these mitogens depend on the activation of the p42/p44 mitogen-activated protein kinases (MAPKs), whereas the role of these kinases in vivo has yet to be established. We tested whether MAPK activity is induced following vessel injury and whether activity is dependent on the release of bFGF, PDGF, and thrombin. Following balloon injury of the left carotid of male Sprague-Dawley rats, arteries were removed and analyzed with respect to MAPK activity, BrdU-labeled nuclei, and/or luminal, medial, and intimal areas. MAPK activity is induced in the rat carotid artery following balloon-catheter injury with a maximum activation at 30 min with a return to just above baseline at 11 hr after injury. Intravenous administration of heparin or neutralizing antibodies to bFGF or PDGF prior to injury reduced SMC proliferation and neointimal lesional formation but did not affect the early induction of MAPK activity. Administration of a tissue factor inhibitor or thrombin inhibitor also did not affect MAPK activity, although it impaired the initiation of the coagulation cascade. IN CONCLUSION: (1) MAPK is activated in a time-dependent manner in response to injury; (2) the antiproliferative effect of heparin in vivo is not mediated through the inhibition of MAPK activity induced 30 min after injury; (3) the activation of MAPK after 30 min is not dependent on PDGF, bFGF, or thrombin following vessel injury in the rat.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carotid Artery Injuries , Catheterization/adverse effects , Protein-Tyrosine Kinases/metabolism , Animals , Blood Coagulation/drug effects , Carotid Arteries/enzymology , Enzyme Activation , Fibroblast Growth Factors/physiology , Heparin/pharmacology , Hirudins/pharmacology , Male , Mitogen-Activated Protein Kinase 1 , Platelet-Derived Growth Factor/physiology , Rats , Rats, Sprague-Dawley
11.
J Biol Chem ; 271(42): 25928-35, 1996 Oct 18.
Article in English | MEDLINE | ID: mdl-8824227

ABSTRACT

Heparin is a complex glycosaminoglycan that inhibits vascular smooth muscle cell (SMC) growth in vitro and in vivo. To define the mechanism by which heparin exerts its antiproliferative effects, we asked whether heparin interferes with the activity of intracellular protein kinase C (PKC). The membrane-associated intracellular PKC activity increased following stimulation of cultured rat SMCs with fetal calf serum and was suppressed by heparin in a time- and dose-dependent manner. Heparin acted through a selective inhibition of the PKC-alpha since preincubation of the cells with a 20-mer phosphorothioate PKC-alpha antisense oligodeoxynucleotide (ODN) eliminated the heparin effect. In vivo, following balloon injury of the rat carotid artery, particulate fraction PKC content increased with a time course and to an extent comparable with the observed changes in vitro. Heparin, administered at the time of injury or shortly thereafter, inhibited the activity of the particulate PKC and suppressed the in situ phosphorylation of an 80-kDa myristoylated alanine-rich protein kinase C substrate (MARCKS), a substrate of PKC. The topical application of the phosphorothioate antisense ODN selectively suppressed the expression of the PKC-alpha isoenzyme in vivo but did not affect injury-induced myointimal proliferation. Topical application of the ODN also eliminated the antiproliferative activity of heparin. These results therefore suggest that heparin might block SMC proliferation by interfering with the PKC pathway through a selective direct inhibition of the PKC-alpha isoenzyme.


Subject(s)
Heparin/pharmacology , Isoenzymes/metabolism , Muscle, Smooth, Vascular/cytology , Protein Kinase C/metabolism , Animals , Carotid Arteries/cytology , Carotid Arteries/drug effects , Cell Division/drug effects , Down-Regulation/drug effects , Enzyme Activation , Heparin Lyase , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Male , Muscle, Smooth, Vascular/drug effects , Oligonucleotides, Antisense/pharmacology , Polysaccharide-Lyases/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-alpha , Rats , Rats, Sprague-Dawley
12.
Circ Res ; 79(4): 812-20, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8831505

ABSTRACT

Arterial smooth muscle cells (SMCs) are in a quiescent growth state under normal physiological conditions, but they can be stimulated to proliferate and migrate from one tissue compartment to another if the vessel is injured. This response might require a selective and focal increase in tissue degradation, which might be mediated through the increased production of matrix metalloproteinases (MMPs). Blockade of MMP activity might therefore inhibit the SMC response to injury. To test this hypothesis, we developed clones of rat SMCs that overexpress baboon tissue inhibitor of matrix metalloproteinase-I (TIMP-1), using retrovirally mediated gene transfer, and characterized the functional capacity of these cells in vitro and in vivo. SMCs transduced with the TIMP-1 vector (LTSN) grew more slowly and also migrated through a gel matrix in a Boyden chamber assay more slowly than the vector alone (LXSN) cells. The conditioned medium from LTSN cells completely inhibited the platelet-derived growth factor-BB-induced migration of normal SMCs across a matrix-coated filter, while the LXSN cell conditioned medium had no effect. The inhibitor activity in the LTSN conditioned medium could be neutralized with an antibody to TIMP-1. In vivo, local overexpression of TIMP-1 using LTSN cells implanted onto balloon-injured rat carotid artery inhibited intimal hyperplasia. Neutralizing antibodies against TIMP-1 suppressed the effect of LTSN cell seeding on intimal thickening. These data support the conclusion that the process of SMC activation leading to a thickened intima is dependent on MMP activity and that TIMP-1 could be utilized to inhibit this process.


Subject(s)
Glycoproteins/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Cell Division/drug effects , Cell Movement/drug effects , Clone Cells , Culture Media, Conditioned/pharmacology , Gene Transfer Techniques , Glycoproteins/genetics , Male , Muscle, Smooth, Vascular/cytology , Rats , Rats, Inbred F344 , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tissue Inhibitor of Metalloproteinases
13.
Biotechniques ; 20(4): 694-701, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8800691

ABSTRACT

The traditional method of antibody (Ab) generation requires repeated injections of antigen (Ag). We have developed an alternative method that allows an investigator to generate a polyclonal antiserum with only a cDNA in hand. We cloned a cDNA encoding the coding frame for baboon tissue inhibitor of matrix metalloproteinase-1 (TIMP-1). Fischer rat arterial smooth muscle cells (SMC) transduced with the baboon TIMP-1 using a replication-defective retrovirus were propagated in culture. TIMP-1 overexpressing rat SMC were seeded into de-endothelialized rat carotid arteries. Three weeks after cell seeding in the rat, the presence of Ab to the baboon TIMP-1 was detected by dot blot and enzyme-linked immunosorbent assay in 5 of 6 of the animals. The major portion of the Ab generated against baboon TIMP-1 during the 12-month monitoring period after the cell seeding was identified as belonging to the IgG1 subtype. More interestingly, the titer of the Ab kept rising throughout an 8-month monitoring period. Among the salient features of this Ab are its capacity to block TIMP-1 activity and its utility for detecting TIMP-1 by immunohistochemistry. These results demonstrate that Ab against a secreted protein can be obtained in response to continuous expression of the cDNA by vascular SMC. Purified Ag is not required.


Subject(s)
Genetic Vectors , Glycoproteins/immunology , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/immunology , Retroviridae/genetics , Animals , Antibodies/chemistry , Antibody Formation , Arteries/chemistry , Base Sequence , Blotting, Northern , Enzyme-Linked Immunosorbent Assay , Glycoproteins/genetics , Glycoproteins/metabolism , Immunoblotting , Immunohistochemistry , Male , Molecular Sequence Data , Polymerase Chain Reaction , Protease Inhibitors/immunology , Rats , Rats, Inbred F344 , Replication Origin/genetics , Tissue Inhibitor of Metalloproteinases , Transduction, Genetic
14.
Proc Natl Acad Sci U S A ; 92(17): 8055-8, 1995 Aug 15.
Article in English | MEDLINE | ID: mdl-7644537

ABSTRACT

The injection of recombinant erythropoietin (Epo) is now widely used for long-term treatment of anemia associated with chronic renal failure, cancer, and human immunodeficiency virus infections. The ability to deliver this hormone by gene therapy rather than by repeated injections could provide substantial clinical and economic benefits. As a preliminary approach, we investigated in rats the expression and biological effects of transplanting autologous vascular smooth muscle cells transduced with a retroviral vector encoding rat Epo cDNA. Vector-derived Epo secretion caused increases in reticulocytes, with peak levels of 7.8-9.6% around day 10 after implantation. The initial elevation in reticulocytes was followed by clinically significant increases in hematocrit and hemoglobin for up to 11 weeks. Ten control and treated animals showed mean hematocrits of 44.9 +/- 0.4% and 58.7 +/- 3.1%, respectively (P < 0.001), and hemoglobin values of 15.6 +/- 0.1 g/dl and 19.8 +/- 0.9 g/dl, respectively (P < 0.001). There were no significant differences between control and treated animals in the number of white blood cells and platelets. Kidney and to a lesser extent liver are specific organs that synthesize Epo in response to tissue oxygenation. In the treated animals, endogenous Epo mRNA was largely down regulated in kidney and absent from liver. These results indicate that vascular smooth muscle cells can be genetically modified to provide treatment of anemias due to Epo deficiency and suggest that this cell type may be targeted in the treatment of other diseases requiring systemic therapeutic protein delivery.


Subject(s)
Erythropoietin/biosynthesis , Gene Expression , Genetic Therapy , Kidney/metabolism , Liver/metabolism , Reticulocytes/physiology , 3T3 Cells , Animals , Base Sequence , Cell Line , DNA Primers , Genetic Vectors , Hematocrit , Hemoglobins/metabolism , Kinetics , Mice , Molecular Sequence Data , Moloney murine leukemia virus , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Time Factors , Transduction, Genetic
15.
J Vasc Surg ; 20(2): 209-17, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8040944

ABSTRACT

PURPOSE: Although matrix metalloproteinase (MMP) expression has been correlated with proliferation and migration of various tumor cells, the relation between MMP expression and smooth muscle cell (SMC) proliferation and migration has not been established. METHODS: We measured MMP expression (gelatin, casein, and elastin zymography) by vascular wall cells in balloon-injured carotid artery during the period of medial SMC proliferation, migration of SMC from the media to the intima, and subsequent intimal SMC proliferation. RESULTS: The 72 and 64-kd gelatinases (presumably 72 kd type IV collagenase or MMP 2) were constitutively expressed in normal carotid arteries, and the activated (59 and 54 kd) forms of this enzyme were increased at 5 days when SMCs start to migrate. A 92 kd gelatinase (presumably 92 kd type IV collagenase or MMP 9) was increased at 24 hours, when SMCs entered the growth cycle, and decreased thereafter. A low-molecular-weight metalloproteinase with elastolytic activity was present in the adventitia, and the activity was increased at 5 days after surgery. CONCLUSIONS: These results suggest that the 72 kd and 92 kd gelatinases may be involved in basement membrane and matrix degradation in the media in relation to SMC proliferation and migration, whereas the low-molecular-weight metalloproteinase may have a role in elastin turnover in the adventitia.


Subject(s)
Carotid Artery Injuries , Catheterization/adverse effects , Metalloendopeptidases/metabolism , Muscle, Smooth, Vascular/enzymology , Animals , Carotid Arteries/cytology , Carotid Arteries/enzymology , Caseins/metabolism , Cell Division , Elastin/metabolism , Gelatin/metabolism , Male , Muscle, Smooth, Vascular/cytology , Rats , Rats, Sprague-Dawley , Time Factors
16.
J Clin Invest ; 93(2): 644-51, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8113400

ABSTRACT

Cultured vascular smooth muscle cells (SMCs) containing retrovirally introduced genes are a potential vehicle for gene replacement therapy. Because the cultured SMCs are selected for their ability to proliferate in vitro, it is possible that the SMCs might be permanently altered and lose their capacity to respond to growth-suppressing conditions after being seeded back into blood vessels. To investigate this possibility we measured SMC proliferation and intimal thickening in balloon-injured Fischer 344 rat carotid arteries seeded with SMCs stained with the fluorescent marker 1,1'-dioctadecyl-3,3,3',3'-tetramethylindo-carbocyanine perchlorate (DiI) and infected with replication-defective retrovirus expressing human adenosine deaminase or human placental alkaline phosphatase. The majority of the seeded SMCs remained in the intima while a few of the cells appeared to migrate into the first layer of the media. Intimal SMC proliferation returned to background levels (< 0.1% thymidine labeling index) by 28 d. At late times (1 and 12 mo) the morphological appearance of the intima was the same for balloon-injured arteries with or without seeded SMC, except that the seeded arteries continued to express human adenosine deaminase or alkaline phosphatase. These results support the conclusion that cultured SMC infected with a replication-defective virus containing human adenosine deaminase or alkaline phosphatase are not phenotypically altered and do not become transformed. After seeding onto the surface of an injured artery, they stop replicating but continue to express the introduced human genes even over the long term.


Subject(s)
Adenosine Deaminase/genetics , Carotid Arteries/physiology , Carotid Artery Injuries , Muscle, Smooth, Vascular/physiology , Retroviridae , Transfection/methods , Adenosine Deaminase/analysis , Adenosine Deaminase/biosynthesis , Alkaline Phosphatase/analysis , Animals , Carbocyanines , Carotid Arteries/ultrastructure , Catheterization/adverse effects , Cells, Cultured , Fluorescent Dyes , Genetic Therapy/methods , Genetic Vectors , Humans , Male , Microscopy, Electron, Scanning , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/ultrastructure , Rats , Rats, Inbred F344
17.
J Vasc Res ; 30(3): 161-8, 1993.
Article in English | MEDLINE | ID: mdl-8518334

ABSTRACT

The effect of heparin on the amount of membrane-associated heparan sulfate proteoglycan (HSPG) was examined biochemically in balloon-injured rat carotid arteries. Heparin (Choay 1772) was administered at the rate of 1 mg/kg/h.i.v. by an osmotic infusion pump for 10 days. Arterial tissues were incubated ex vivo with Na2(35)SO4 and 3H-glucosamine for 6 or 24 h. The arteries from heparin-treated rats were also incubated with heparin (100 micrograms/ml) during the radiolabeling. Membrane-associated proteoglycans were extracted with 4 M guanidine-HCl and isolated by a series of chromatographic steps using Sephadex G-50, DEAE-Sephacel and Octyl-Sepharose CL-4B. Membrane-associated proteoglycans were eluted with 0.8% Triton X-100 from Octyl-Sepharose columns. Heparin treatment significantly increased membrane-associated HSPG by 64% in the arteries incubated for 24 h, while heparin hardly increased the HSPG in 6 h incubated arteries. Since the 24-hour data seem to reflect a combination of biosynthesis and degradation of proteoglycans while the 6-hour data primarily reflect biosynthesis, these results suggest that inhibition of degradation of membrane-associated HSPG is involved in the mechanism of heparin action. This speculation is supported by the observation that membrane-associated HSPG was elevated in balloon-injured arteries by treatment with protease inhibitors.


Subject(s)
Carotid Arteries/metabolism , Carotid Artery Injuries , Catheterization , Heparin/pharmacology , Heparitin Sulfate/metabolism , Proteoglycans/metabolism , Animals , Cell Membrane/metabolism , Chromatography, Agarose , Heparan Sulfate Proteoglycans , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
18.
Haemostasis ; 23 Suppl 1: 177-82, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8495866

ABSTRACT

Heparin, an inhibitor of vascular smooth muscle cell proliferation and migration, affects a number of other cell functions. These effects include inhibition of growth factor binding, deposition of matrix proteins and gene expression. Various mechanisms have been proposed and, yet, how heparin works as an inhibitor remains unclear. We have postulated that heparin inhibits smooth muscle cell growth and migration by suppressing the expression of matrix-degrading enzymes such as plasminogen activators and interstitial collagenase. The molecular mechanism of heparin's inhibitory action on these proteases is currently under investigation.


Subject(s)
Heparin/pharmacology , Models, Biological , Muscle, Smooth, Vascular/drug effects , Animals , Base Sequence , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Collagenases/biosynthesis , Depression, Chemical , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase Inhibitors , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Oncogenes/drug effects , Plasminogen Activators/antagonists & inhibitors , Plasminogen Activators/biosynthesis , Primates , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/metabolism , Rats
19.
Arterioscler Thromb ; 12(12): 1488-95, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1450180

ABSTRACT

Intimal thickening in response to vascular injury is inhibited in animals previously subjected to hypophysectomy. We have investigated the nature and cell kinetics of this effect in a balloon catheter model of injury to the rat carotid artery. The ability of injury to stimulate [3H]thymidine labeling 48 hours after injury was almost completely eliminated in hypophysectomized (hypox) compared with control animals (0.1% versus 32.1%). Total DNA content of the developing neointima 14 days after injury was only 30% of the values found in ballooned carotid arteries of normal rats. If hypox rats were treated with recombinant human growth hormone, the proliferative response was not restored. There are two possible general explanations for the reduction of proliferative response in hypox animals: 1) that smooth muscle cells in the hypox animals have lost the ability to respond to the stimulus of injury or 2) that the ability of the smooth muscle cells to respond has not been reduced by prior hypophysectomy, but that the response itself requires the presence of pituitary-dependent factors. Transplantation experiments were performed in vivo to distinguish between these possibilities. Carotid arteries in inbred Lewis rats were excised 1 hour after balloon injury to give platelets the opportunity to adhere. These vessels were then transplanted from hypox into control animals and vice versa. At 48 hours, proliferation of smooth muscle cells in "control-to-hypox" transplants was 0.3% compared with 14.3% in "control-to-control" transplants, whereas vessels from hypox rats increased their indices to 4.8% if transplanted into control animals.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Muscle, Smooth, Vascular/cytology , Pituitary Gland/physiology , Animals , Carotid Arteries/transplantation , Cell Division , Cells, Cultured , Growth Hormone/pharmacology , Hypophysectomy , Insulin-Like Growth Factor I/physiology , Male , Platelet-Derived Growth Factor/analysis , Rats , Rats, Sprague-Dawley
20.
Circ Res ; 70(6): 1128-36, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1374298

ABSTRACT

Smooth muscle cells (SMCs) in balloon-injured rat carotid artery express tissue-type plasminogen activator (t-PA) at a time when they are migrating from the media to the intima. Since heparin inhibits SMC migration and intimal thickening, we have examined the possibility that heparin might also inhibit t-PA expression. Heparin (nonanticoagulant fraction; molecular weight, approximately 6,000) was administered by continuous intravenous infusion (1.0 mg/kg per hour) to Sprague-Dawley rats subjected to balloon injury of the left common carotid artery. At various times up to 14 days after injury, plasminogen activator expression was analyzed by zymography, plasmin generation, enzyme-linked immunosorbent assay, Northern blotting, and in situ hybridization. This dose of heparin inhibited SMC accumulation at 14 days by 60%. Both urokinase plasminogen activator (u-PA) and t-PA activity increased in injured arteries and reached a maximum at 7 days. Heparin treatment decreased t-PA, but not u-PA, activity. Total t-PA protein was decreased by treatment with heparin but not chondroitin sulfate, and the decrease in t-PA protein was associated with decreased t-PA mRNA in the media. These results in the injured rat carotid artery agree with our earlier observations that heparin inhibits t-PA gene expression in cultured baboon aortic SMCs. They also provide support for the hypothesis that heparin interferes with the expression of certain proteases required for SMC migration and proliferation.


Subject(s)
Carotid Artery Injuries , Heparin/pharmacology , Muscle, Smooth, Vascular/physiology , Tissue Plasminogen Activator/analysis , Animals , Autoradiography , Blotting, Northern , DNA/analysis , DNA Probes , Electrophoresis , Enzyme-Linked Immunosorbent Assay , Glycosaminoglycans/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , RNA/analysis , Rats , Rats, Inbred Strains , Sodium Chloride/pharmacology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...