Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Indian J Med Res ; 148(Suppl): S151-S155, 2018 12.
Article in English | MEDLINE | ID: mdl-30964093

ABSTRACT

Background & objectives: Polycystic ovary syndrome (PCOS) is an endocrinopathy warranting lifelong individualized management by lifestyle and pharmacological agents mainly oral contraceptive pills (OCPs). This study was aimed to report the impact of six-month OCP use on plasminogen activator inhibitor-1 (PAI-1) and factor VIII (FVIII) in women with PCOS. Methods: PCOS women diagnosed on the basis of Rotterdam 2003 criteria, either treated with OCPs (ethinyl estradiol-0.03 mg, levonorgestrel-0.15 mg) for a period of six months (n=40) or drug-naïve (n=42), were enrolled in this study. Blood was drawn to estimate glucose, insulin levels and lipid profile. Chemiluminescence immunoassays were used to measure hormones (LH, FSH, PRL, T4). Plasma levels of PAI-I and FVIII were measured by commercially available kits. Results: Menstrual regularity, Ferriman-Gallwey score and serum total testosterone significantly improved in the OCP group compared to drug-naïve group (P<0.01). No significant difference was observed in PAI-1 levels of the two groups; however, significant decrease in FVIII levels was observed in OCP group as compared to drug-naïve group. PAI-1 levels of OCP group correlated positively with blood glucose two hours, triglycerides and insulin two hours, while FVIII levels of OCP group correlated negatively with fasting insulin and homoeostatic model assessment-insulin resistance. Interpretation & conclusions: OCPs use has differential effect on pro-coagulant markers among women with PCOS. Well-designed, long-term, prospective, large-scale studies are prerequisite to elucidate the efficacy and safety of OCP in the treatment of PCOS.


Subject(s)
Contraceptives, Oral/administration & dosage , Factor VIII/administration & dosage , Plasminogen Activator Inhibitor 1/administration & dosage , Polycystic Ovary Syndrome/drug therapy , Adult , Blood Glucose/drug effects , Contraceptives, Oral/chemistry , Contraceptives, Oral, Combined/administration & dosage , Factor VIII/chemistry , Female , Humans , Insulin Resistance/genetics , Metformin/administration & dosage , Pilot Projects , Plasminogen Activator Inhibitor 1/chemistry , Polycystic Ovary Syndrome/physiopathology
2.
Biomed Res Int ; 2015: 392862, 2015.
Article in English | MEDLINE | ID: mdl-25883959

ABSTRACT

PAI-1 prevents lysis of blood clot by inhibiting the urokinase and tPA induced conversion of plasminogen to plasmin. VLHL PAI-1 protein mutant was created to extend half-life over 700 hours. The objective of this paper was to test VLHL PAI-1 effects on bleeding during partial nephrectomy in mice. All animals had a left partial nephrectomy after intravenous infusion of saline or tPA. The animals were divided into four groups. Group 1 was infused with saline and kidney was exposed to saline too; Group 2 was infused with saline and kidney was exposed to PAI-1. Group 3 was infused with tPA and kidney was exposed to saline, while Group 4 was infused with tPA and kidney was exposed to PAI-1. Preweighed gauze containing PAI-1 or saline was then applied to the kidney for 30 minutes. The gauze was afterward weighed and blood loss was measured by subtracting the preweight of gauze from the final weight. We have observed a statistically significant (P ≤ 0.05) reduction of bleeding in PAI-1-treated group in comparison to saline and tPA-treated groups. Based on these results we propose that VLHL PAI-1 can be used therapeutically in limiting the flow of blood from renal wounds.


Subject(s)
Hemorrhage/drug therapy , Kidney/drug effects , Nephrectomy/methods , Plasminogen Activator Inhibitor 1/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Animals , Bandages , Blood Coagulation , Hemostasis , Humans , Kidney/surgery , Male , Mice , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/genetics , Thrombelastography
3.
J Autoimmun ; 52: 36-43, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24423642

ABSTRACT

Tissue plasminogen activator (tPA), a component of the PA/plasmin system, is elevated in inflammatory areas and plays a role in inflammatory neurological disorders. In the present study we explored the involvement of tPA and the potential immunomodulatory activity of tPA in experimental autoimmune myasthenia gravis (EAMG). Mice deficient in tPA (tPA(-/-)) present with a markedly more severe disease than wild type EAMG mice. In an attempt to treat EAMG with an 18aa peptide derived from the PA system inhibitor (PAI-1), designed to tether out the endogenous inhibitor, a significant suppression of disease severity was demonstrated. The more severe disease in tPA(-/-) mice was accompanied by a higher level of anti-AChR antibodies and increased expression of B-cell markers. In view of the essential role of B-cell activating factor (BAFF) in B-cell maturation, the expression of BAFF family components was tested. An increase in BAFF and BAFF receptor was observed in EAMG tPA(-/-) mice, whereas BCMA expression was reduced, consistent with the increased level of pathogenic antibodies and the more severe disease. Given the importance of T regulatory cells (Tregs) in EAMG, they were evaluated and their number was reduced in tPA(-/-) mice, in which EAMG was aggravated, whereas following PAI-1dp treatment, Tregs were replenished and the disease was ameliorated. The results show the involvement of tPA in EAMG, implying a protective role for tPA in EAMG pathogenesis. The amelioration of EAMG by PAI-1dp treatment suggests that the PA system may be considered a potential site for therapeutic intervention in neuroimmune diseases.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/blood , T-Lymphocytes, Regulatory/immunology , Tissue Plasminogen Activator/metabolism , Animals , Autoantibodies/blood , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , Disease Progression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , Peptide Fragments/administration & dosage , Plasminogen Activator Inhibitor 1/administration & dosage , Receptors, Cholinergic/immunology , T-Lymphocytes, Regulatory/drug effects , Tissue Plasminogen Activator/genetics , Up-Regulation
4.
Stroke ; 44(9): 2623-2627, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23881953

ABSTRACT

BACKGROUND AND PURPOSE: Plasminogen activator inhibitor-I (PAI-1), a ≈50-kDa serine protease inhibitor, markedly reduces the extravascular toxicity of tissue-type plasminogen activator in experimental hypoxic-ischemic (HI) brain injury of newborns. However, the current treatment with PAI-1 requires intracerebroventricle injection to cross the blood-brain barrier, which is an invasive procedure of limited clinical potential. Thus, we tested whether intranasal administration of PAI-1 can bypass blood-brain barrier and mitigate neonatal HI brain injury. METHODS: Rat pups were subjected to HI, with or without lipopolysaccharide pre-exposure, followed by intranasal delivery of a stable-mutant form of PAI-1 (CPAI). RESULTS: Immunoblotting showed that CPAI sequentially entered the olfactory bulbs and cerebral cortex after intranasal delivery and reduced ≈75% of brain atrophy in HI or lipopolysaccharide-sensitized HI injury. Mechanistically, CPAI attenuated HI-induced plasminogen activators and lipopolysaccharide/HI-induced nuclear factor-κB signaling, neuroinflammation, and blood-brain barrier permeability. CONCLUSIONS: Intranasal delivery of CPAI is an effective treatment of experimental HI brain injury of newborns. Clinical application of this experimental therapy merits further investigation.


Subject(s)
Hypoxia-Ischemia, Brain/drug therapy , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Inactivators/administration & dosage , Plasminogen Inactivators/therapeutic use , Administration, Intranasal , Animals , Animals, Newborn , Atrophy , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Disease Models, Animal , Drug Administration Schedule , Female , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Male , Plasminogen Activator Inhibitor 1/therapeutic use , Rats , Rats, Wistar , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/therapeutic use
5.
Chronobiol Int ; 29(10): 1383-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23130962

ABSTRACT

Circadian rhythm interactions of hemostatic factors can modify tissue plasminogen activator (tPA) effects. We assess the relationship of the time frame of intravenous tPA administration with the outcome of patients with acute ischemic stroke (AIS). We studied 135 consecutive patients with AIS and transcranial duplex documented middle cerebral artery (MCA) occlusion treated with intravenous tPA. Complete recanalization was defined as total improvement on thrombolysis in brain ischemia (TIBI) grades 2 h after tPA infusion. Clinical response was evaluated by the modified Rankin scale at 90 days. We determined plasminogen activator inhibitor-1 (PAI-1) levels in 33 patients with available plasma samples before treatment. Our results are follows: 92 (68.1%) patients were treated in the diurnal (9:00-21:00) and 43 (31.8%) in the nocturnal period (21:00-9:00). Complete recanalization was recorded in 52/135 (38.5%) patients. Both the rate of complete recanalization (45.6% vs. 23.2%; p = .01) and good clinical outcome (64.1% vs. 44.2%; p = .02) were significantly higher in the group of diurnal tPA administration compared with those treated in the nocturnal period. The adjusted odds ratio (OR) of diurnal tPA treatment for complete MCA recanalization was 2.37 (95% confidence interval [CI], 1.02-5.52; p = .045). Diurnal tPA infusion significantly improved the overall distribution of scores on the modified Rankin scale, as compared with nocturnal treatment (OR, 2.07; 95% CI, 1.16-4.64 by ordinal regression analysis). Low PAI-1 levels were associated with complete recanalization but did not significantly differ between the two time frames. In conclusion, diurnal administration of tPA is associated with complete MCA recanalization and better functional outcome at 90 days in patients with AIS.


Subject(s)
Brain Ischemia/drug therapy , Circadian Rhythm/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Plasminogen Activator Inhibitor 1/therapeutic use , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Aged , Aged, 80 and over , Circadian Rhythm/physiology , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Plasminogen Activator Inhibitor 1/administration & dosage , Time Factors , Tissue Plasminogen Activator/administration & dosage , Treatment Outcome
6.
Brain ; 135(Pt 11): 3251-64, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22822039

ABSTRACT

The neurovascular unit provides a dynamic interface between the circulation and central nervous system. Disruption of neurovascular integrity occurs in numerous brain pathologies including neurotrauma and ischaemic stroke. Tissue plasminogen activator is a serine protease that converts plasminogen to plasmin, a protease that dissolves blood clots. Besides its role in fibrinolysis, tissue plasminogen activator is abundantly expressed in the brain where it mediates extracellular proteolysis. However, proteolytically active tissue plasminogen activator also promotes neurovascular disruption after ischaemic stroke; the molecular mechanisms of this process are still unclear. Tissue plasminogen activator is naturally inhibited by serine protease inhibitors (serpins): plasminogen activator inhibitor-1, neuroserpin or protease nexin-1 that results in the formation of serpin:protease complexes. Proteases and serpin:protease complexes are cleared through high-affinity binding to low-density lipoprotein receptors, but their binding to these receptors can also transmit extracellular signals across the plasma membrane. The matrix metalloproteinases are the second major proteolytic system in the mammalian brain, and like tissue plasminogen activators are pivotal to neurological function but can also degrade structures of the neurovascular unit after injury. Herein, we show that tissue plasminogen activator potentiates neurovascular damage in a dose-dependent manner in a mouse model of neurotrauma. Surprisingly, inhibition of activity following administration of plasminogen activator inhibitor-1 significantly increased cerebrovascular permeability. This led to our finding that formation of complexes between tissue plasminogen activator and plasminogen activator inhibitor-1 in the brain parenchyma facilitates post-traumatic cerebrovascular damage. We demonstrate that following trauma, the complex binds to low-density lipoprotein receptors, triggering the induction of matrix metalloproteinase-3. Accordingly, pharmacological inhibition of matrix metalloproteinase-3 attenuates neurovascular permeability and improves neurological function in injured mice. Our results are clinically relevant, because concentrations of tissue plasminogen activator: plasminogen activator inhibitor-1 complex and matrix metalloproteinase-3 are significantly elevated in cerebrospinal fluid of trauma patients and correlate with neurological outcome. In a separate study, we found that matrix metalloproteinase-3 and albumin, a marker of cerebrovascular damage, were significantly increased in brain tissue of patients with neurotrauma. Perturbation of neurovascular homeostasis causing oedema, inflammation and cell death is an important cause of acute and long-term neurological dysfunction after trauma. A role for the tissue plasminogen activator-matrix metalloproteinase axis in promoting neurovascular disruption after neurotrauma has not been described thus far. Targeting tissue plasminogen activator: plasminogen activator inhibitor-1 complex signalling or downstream matrix metalloproteinase-3 induction may provide viable therapeutic strategies to reduce cerebrovascular permeability after neurotrauma.


Subject(s)
Brain Injuries/physiopathology , Capillary Permeability/physiology , Plasminogen Activator Inhibitor 1/physiology , Tissue Plasminogen Activator/physiology , Adult , Aged , Aged, 80 and over , Albumins/metabolism , Animals , Brain/blood supply , Brain/metabolism , Brain Injuries/cerebrospinal fluid , Brain Injuries/drug therapy , Brain Injuries/metabolism , Capillary Permeability/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Injections, Intraventricular , Male , Matrix Metalloproteinase Inhibitors/therapeutic use , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/metabolism , Recovery of Function/physiology , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism
7.
Neurol Res ; 33(9): 983-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22081002

ABSTRACT

OBJECTIVE: The thrombolytic treatment of stroke is limited by a narrow therapeutic time window and is associated with significant adverse side effects. To improve this situation, the modulation of tissue-type plasminogen activator (tPA) activity by a synthetic plasminogen activator inhibitor-1-derived 18-mer peptide (THR-18) was examined in two models of stroke in rats. METHODS: In the first model (thromboembolic), stroke was induced by intra-carotid injection of micro-clots to rats, and tPA (6 mg/kg) was intravenously infused for 30 minutes with or without THR-18 (1 mg/kg) at 4 hours post-induction. In the second model [transient middle cerebral artery occlusion (tMCAO)], stroke was induced for 2 hours by a transient mechanical occlusion. tPA and/or THR-18 (0.02, 0.1, and 1 mg/kg) were intravenously infused for 60 minutes at the time of reperfusion. RESULTS: In the thromboembolic model, cerebral blood flow, measured before and up to 5.5 hours post-induction, revealed that tPA administration caused reperfusion of flow at 30 minutes post-infusion. Later on, an additional increase in reperfusion was seen in the tPA+THR-18 group, and not with tPA alone. In both models, the frequency of intracranial hemorrhage in the tPA-treated group was found to be significantly higher than the control, and this tPA effect was attenuated by THR-18. In the thromboembolic study, infarct size and brain edema were similar in the control and tPA-treated rats. However, the combination of tPA and THR-18 caused a statistically significant reduction in both parameters (infarct size 17.8 versus 25.0%, brain edema 5 versus 8%, tPA+THR-18 versus control, respectively). In the tMCAO mechanical model, infarct size and brain edema were both increased by tPA treatment as compared to the control group, and this increase was markedly diminished by THR-18 co-administration. Neurobehavioral assessment of the tMCAO animals performed at 72 hours post-stroke induction revealed significant improvements (P<0.05-0.01) in neuroscores in all groups of animals treated with peptide-tPA, as compared to the tPA monotherapy group. A significant (P<0.05) improvement in the neurological outcome was also seen in the THR-18 monoterapy group, as compared to the control animals, thus demonstrating a clear neuroprotective effect by the peptide on its own. DISCUSSION: The results support the use of THR-18 together with tPA in the thrombolytic therapy of stroke, in order to achieve better patency, less tPA-induced damage, and possibly a widening of tPA therapeutic time window.


Subject(s)
Fibrinolytic Agents/administration & dosage , Neuroprotective Agents/administration & dosage , Plasminogen Activator Inhibitor 1/administration & dosage , Stroke/drug therapy , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/administration & dosage , Animals , Brain Edema/prevention & control , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Intracranial Hemorrhages/prevention & control , Male , Peptides/administration & dosage , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects
8.
Chem Pharm Bull (Tokyo) ; 59(2): 215-24, 2011.
Article in English | MEDLINE | ID: mdl-21297302

ABSTRACT

Novel anthranilic acid derivatives having substituted N-acyl side chains were designed and synthesized for evaluation as plasminogen activator inhibitor-1 (PAI-1) inhibitors. Compounds with a 4-diphenylmethyl-1-piperazinyl moiety on the acyl side chains in general exhibited potent in vitro PAI-1 inhibitory activity and good pharmacokinetic profiles after oral administration in rats. Compound 16f (TM5275) was identified as a promising candidate for further pharmacological evaluation.


Subject(s)
Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/chemistry , ortho-Aminobenzoates/administration & dosage , ortho-Aminobenzoates/chemistry , Administration, Oral , Animals , Humans , Male , Plasminogen Activator Inhibitor 1/pharmacokinetics , Rats , Rats, Wistar , Structure-Activity Relationship , ortho-Aminobenzoates/pharmacokinetics
9.
Int J Mol Med ; 26(4): 501-4, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818488

ABSTRACT

Civilian and military trauma patients consist of a disproportional number of young people, causing a considerable burden to society in terms of disability and premature death. Hemorrhage is a leading cause of mortality in this group of patients and the novel methods to reduce bleeding would be welcomed. Management of bleeding following major trauma includes hemostatic agents that offer effective clotting. However a very limited number of agents control secondary bleeding triggered by lysis of the clot. Fibrinolysis depends on the balance between tissue plasminogen activator (tPA), activating plasminogen to plasmin initiating fibrinolysis, and plasminogen activator inhibitor type 1 (PAI-1) inhibiting tPA and preventing lysis. The drugs available on the market that prevent the activation of plasminogen have been used successfully, but have some side effects and limited efficacy for the control of localized bleeding in the surgical setting. Inhibitors of tPA, initiator of clot fibrinolysis, have not yet found their way into the clinical arena. Plasminogen activator inhibitor-1, the major specific inhibitor of tPA, can be used to limit fibrinolysis. Unfortunately, PAI-1 has a short half-life of approximately 2 h and is rapidly converted to the latent form. A recombinant PAI-1 with very long half-life developed in our laboratory (a two-point mutant, VLHL PAI-1, half-life over 700 h) has clinical potential as an agent to promote hemostasis in several scenarios including surgical injury, trauma, and PAI-1 deficiency. Here we report testing of VLHL PAI-1 as a potent inactivator of fibrinolysis reducing total blood loss while applied systemically or topically in experimental animals. The very long half-life of VLHL PAI-1 may provide an advantage in the important physiological mechanism to protect clots from premature dissolution, when applied topically or systemically to prevent excessive bleeding in the surgical and trauma setting and possibly in PAI-1 deficient patients.


Subject(s)
Antifibrinolytic Agents/administration & dosage , Antifibrinolytic Agents/therapeutic use , Fibrinolysis/drug effects , Hemorrhage/drug therapy , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/therapeutic use , Administration, Topical , Animals , Hemostatics , Humans , Mice , Mice, Inbred C57BL , Mutation , Plasminogen Activator Inhibitor 1/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
10.
J Neurosci ; 29(27): 8669-74, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587273

ABSTRACT

Disruption of the integrity of the blood-brain barrier (BBB) is an important mechanism of cerebrovascular diseases, including neonatal cerebral hypoxia-ischemia (HI). Although both tissue-type plasminogen activator (tPA) and matrix metalloproteinase-9 (MMP-9) can produce BBB damage, their relationship in neonatal cerebral HI is unclear. Here we use a rodent model to test whether the plasminogen activator (PA) system is critical for MMP-9 activation and HI-induced brain injury in newborns. To test this hypothesis, we examined the therapeutic effect of intracerebroventricular injection of plasminogen activator inhibitor-1 (PAI-1) in rat pups subjected to unilateral carotid artery occlusion and systemic hypoxia. We found that the injection of PAI-1 greatly reduced the activity of both tPA and urokinase-type plasminogen activator after HI. It also blocked HI-induced MMP-9 activation and BBB permeability at 24 h of recovery. Furthermore, magnetic resonance imaging and histological analysis showed the PAI-1 treatment reduced brain edema, axonal degeneration, and cortical cell death at 24-48 h of recovery. Finally, the PAI-1 therapy provided a dose-dependent decrease of brain tissue loss at 7 d of recovery, with the therapeutic window at 4 h after the HI insult. Together, these results suggest that the brain PA system plays a pivotal role in neonatal cerebral HI and may be a promising therapeutic target in infants suffering hypoxic-ischemic encephalopathy.


Subject(s)
Hypoxia-Ischemia, Brain/prevention & control , Hypoxia-Ischemia, Brain/physiopathology , Plasminogen Activator Inhibitor 1/administration & dosage , Animals , Animals, Newborn , Dose-Response Relationship, Drug , Humans , Hypoxia-Ischemia, Brain/enzymology , Injections, Intraventricular , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Protease Inhibitors/administration & dosage , Rats , Rats, Wistar , Serine Proteinase Inhibitors/administration & dosage
11.
J Urol ; 181(1): 336-42, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19010488

ABSTRACT

PURPOSE: The potent effects of PAI-1 on tumorigenesis and angiogenesis in various experimental models are complex, complicated and at times contradictory. We determined the therapeutic potential of PAI-1 for inhibiting bladder tumor invasion under conditions that closely mimic the clinical setting. MATERIALS AND METHODS: An orthotopic rat bladder tumor model was established by implanting AY-27 rat transitional carcinoma cells into the bladder lumen of syngeneic Fischer F344 rats. Three weeks after implantation 1 microM PAI-1 was administrated directly into the bladder lumen twice weekly for 2 weeks. Two days after the final treatment tumor size, total bladder weight, tumor stage and angiogenesis were assessed. To assess the uPA axis the levels of active and total uPA, and active and total PAI-1 in tumor extracts were determined 0, 2, 24 and 48 hours after intravesical PAI-1 administration. RESULTS: Intravesical PAI-1 bound and inactivated its molecular target, tumor uPA. There was significant inhibition of bladder tumor progression, as manifested by 53%, 37% and 57% reductions in tumor size, total bladder weight and angiogenesis, respectively. Only 22% of PAI-1 treated tumors invaded muscle vs 79% in controls. No PAI-1 toxicity was detected. CONCLUSIONS: To our knowledge this study is the first to demonstrate that intravesical treatment with PAI-1 significantly inhibits tumor progression in an in vivo model of bladder cancer. Further clinical development is warranted for using PAI-1 directly or in combination with current standards, such as bacillus Calmette-Guerin or interferon.


Subject(s)
Plasminogen Activator Inhibitor 1/administration & dosage , Serine Proteinase Inhibitors/administration & dosage , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/prevention & control , Administration, Intravesical , Animals , Disease Progression , Neoplasm Invasiveness , Rats , Rats, Inbred F344
13.
J Immunol ; 177(10): 7233-41, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17082641

ABSTRACT

Recruitment of neutrophils to the lung is a sentinel event in acute lung inflammation. Identifying mechanisms that regulate neutrophil recruitment to the lung may result in strategies to limit lung damage and improve clinical outcomes. Recently, the renin angiotensin system (RAS) has been shown to regulate neutrophil influx in acute inflammatory models of cardiac, neurologic, and gastrointestinal disease. As a role for the RAS in LPS-induced acute lung inflammation has not been described, we undertook this study to examine the possibility that the RAS regulates neutrophil recruitment to the lung after LPS exposure. Pretreatment of mice with the angiotensin-converting enzyme (ACE) inhibitor enalapril, but not the anti-hypertensive hydralazine, decreased pulmonary neutrophil recruitment after exposure to LPS. We hypothesize that inhibition of LPS-induced neutrophil accumulation to the lung with enalapril occurred through both an increase in bradykinin, and a decrease in angiotensin II (ATII), mediated signaling. Bradykinin receptor blockade reversed the inhibitory effect of enalapril on neutrophil recruitment. Similarly, pretreatment with bradykinin receptor agonists inhibited IL-8-induced neutrophil chemotaxis and LPS-induced neutrophil recruitment to the lung. Inhibition of ATII-mediated signaling, with the ATII receptor 1a inhibitor losartan, decreased LPS-induced pulmonary neutrophil recruitment, and this was suggested to occur through decreased PAI-1 levels. LPS-induced PAI-1 levels were diminished in animals pretreated with losartan and in those deficient for the ATII receptor 1a. Taken together, these results suggest that ACE regulates LPS-induced pulmonary neutrophil recruitment via modulation of both bradykinin- and ATII-mediated pathways, each regulating neutrophil recruitment by separate, but distinct, mechanisms.


Subject(s)
Angiotensin II/physiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Bradykinin/physiology , Lipopolysaccharides/antagonists & inhibitors , Lung/enzymology , Lung/pathology , Signal Transduction/immunology , Angiotensin II/antagonists & inhibitors , Angiotensin-Converting Enzyme Inhibitors/metabolism , Animals , Bradykinin/agonists , Cell Migration Inhibition , Chemokine CXCL1 , Chemokine CXCL2 , Chemokines/antagonists & inhibitors , Chemokines/metabolism , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/metabolism , Chemotaxis, Leukocyte , Down-Regulation/immunology , Female , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Losartan/pharmacology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Bradykinin/agonists , Signal Transduction/drug effects
14.
J Neurochem ; 93(5): 1272-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15948318

ABSTRACT

The extracellular serine protease tissue plasminogen activator (tPA) that converts plasminogen into plasmin is abundantly expressed throughout the central nervous system. We have recently demonstrated that the tPA-plasmin system participates in the rewarding and locomotor-stimulating effects of morphine by acutely regulating morphine-induced dopamine release in the nucleus accumbens (NAc). In the present study, we examined the effects of microinjections of plasminogen activator inhibitor-1 (PAI-1), tPA or plasmin into the NAc on morphine-induced dopamine release, hyperlocomotion and anti-nociceptive effects in ICR mice. A single morphine treatment resulted in an increase in protein levels of PAI-1 in the NAc. Microinjection of PAI-1 into the NAc dose-dependently reduced morphine-induced dopamine release and hyperlocomotion. In contrast, microinjection of tPA into the NAc significantly potentiated morphine-induced dopamine release and hyperlocomotion without affecting basal levels. Furthermore, microinjection of plasmin enhanced morphine-induced dopamine release, but did not modify the hyperlocomotion induced by morphine. The intracerebroventricular injection of PAI-1, tPA and plasmin at high doses had no effect on the anti-nociceptive effects of morphine. These results suggest that the tPA-plasmin system is involved in the regulation of morphine-induced dopamine release and dopamine-dependent behaviors but not the anti-nociceptive effects of morphine.


Subject(s)
Dopamine/metabolism , Fibrinolysin/metabolism , Morphine/pharmacology , Motor Activity/drug effects , Narcotics/pharmacology , Nociceptors/drug effects , Tissue Plasminogen Activator/metabolism , Animals , Fibrinolysin/administration & dosage , Fibrinolysin/pharmacology , Injections, Intraventricular , Male , Mice , Mice, Inbred ICR , Nucleus Accumbens/metabolism , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/pharmacology , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/pharmacology , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/pharmacology
15.
Circulation ; 111(17): 2241-9, 2005 May 03.
Article in English | MEDLINE | ID: mdl-15851587

ABSTRACT

BACKGROUND: Accumulating evidence demonstrates a critical involvement of tissue-type plasminogen activator (tPA) in pathological and physiological brain conditions. Determining whether and how vascular tPA can cross the blood-brain barrier (BBB) to enter the brain is thus important, not only during stroke but also in physiological conditions. METHODS AND RESULTS: In the present work, we provide evidence in vivo that intravenous injection of tPA increases NMDA-induced striatal lesion in the absence of BBB leakage. Accordingly, we show that tPA crosses the BBB both after excitotoxic lesion and in control conditions. Indeed, vascular injected tPA can be detected within the brain parenchyma and in the cerebrospinal fluid. By using an in vitro model of BBB, we have confirmed that tPA can cross the intact BBB. Its passage was blocked at 4 degrees C, was saturable, and was independent of its proteolytic activity. We have shown that tPA crosses the BBB by transcytosis, mediated by a member of the LDL receptor-related protein family. CONCLUSIONS: We demonstrate that blood-derived tPA can reach the brain parenchyma without alteration of the BBB. The molecular mechanism of the passage of tPA from blood to brain described here could represent an interesting target to improve thrombolysis in stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Tissue Plasminogen Activator/adverse effects , Tissue Plasminogen Activator/pharmacokinetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cold Temperature , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Male , N-Methylaspartate/administration & dosage , Neurotoxicity Syndromes/etiology , Plasminogen Activator Inhibitor 1/administration & dosage , Protein Transport , Rats , Rats, Sprague-Dawley
16.
Invest Ophthalmol Vis Sci ; 44(6): 2791-7, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12766088

ABSTRACT

PURPOSE: To explain the conflicting reports about the influence of plasminogen activator inhibitor type (PAI-1) on pathologic angiogenesis, such as occurs during the exudative form of age-related macular degeneration. METHODS: The expression of PAI-1 mRNA was analyzed in human and murine choroidal neovascularization (CNV) by RT-PCR. The influences of increasing doses of recombinant PAI-1 were evaluated by daily intraperitoneal injections in PAI-1(-/-) and wild-type animals with a model of laser-induced CNV. The double mechanism of action of PAI-1 (proteolytic activity inhibition versus vitronectin binding) was explored by immunohistochemical localization of fibrinogen/fibrin and by injection of recombinant PAI-1 protein defective for vitronectin binding or with adenoviral vectors bearing a mutated binding-deficient PAI-1 gene. RESULTS: PAI-1 expression was present in human CNV and strongly induced in the course of experimental subretinal neovascularization. Daily injections of recombinant PAI-1 proteins in control and PAI-1(-/-) animals demonstrated that PAI-1 could exhibit both pro- and antiangiogenic effects, dependent on the dose. PAI-1 mutants defective for vitronectin binding were used to show that PAI-1 promotes choroidal pathologic angiogenesis merely through its antiproteolytic activity. CONCLUSIONS: These observations may help to reconcile reports with opposite results regarding the effects of PAI-1 on angiogenesis and certainly warn against uncontrolled use of PAI-1-modulating drugs in clinical trials.


Subject(s)
Choroidal Neovascularization/physiopathology , Plasminogen Activator Inhibitor 1/administration & dosage , Serine Proteinase Inhibitors/administration & dosage , Adenoviridae/genetics , Animals , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fluorescent Antibody Technique, Indirect , Gene Transfer Techniques , Genetic Vectors , Humans , Immunoenzyme Techniques , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA, Messenger/metabolism , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/metabolism
17.
Circulation ; 104(16): 1972-1, 2001 Oct 16.
Article in English | MEDLINE | ID: mdl-11602503

ABSTRACT

BACKGROUND: Elevated plasma levels of plasminogen activator inhibitor type 1 (PAI-1) are associated with myocardial infarction, atherosclerosis, and restenosis. PAI-1 is increased in atherosclerotic arteries and failed vein grafts. No experimental data, however, support a causal relationship between elevated PAI-1 expression and vascular lesions. Paradoxically, data generated in PAI-1 knockout mice suggest that PAI-1 might decrease lesion formation after arterial injury and that PAI-1 gene transfer might prevent restenosis. METHODS AND RESULTS: Using the rat carotid balloon injury model and a PAI-1-expressing adenoviral vector, we tested whether elevated arterial PAI-1 expression would alter neointima formation. Compared with control-transduced arteries, neointima formation in PAI-1-transduced arteries was initially retarded. By 14 days, however, the intimas of PAI-1-transduced arteries were significantly larger than intimas of control-transduced arteries (1.6+/-0.1x10(5) versus 1.2+/-0.1x10(5) micrometer(2), n=18 to 19, P<0.03). PAI-1 expression in individual arteries correlated with increased cell proliferation at 4 and 8 days after injury (R=0.6, P<0.02 and P<0.006). PAI-1 expression also correlated with fibrin(ogen) accumulation (R=0.77, P<0.001), and fibrin(ogen) accumulation correlated strongly with proliferation (R=0.86, P<0.00001). CONCLUSIONS: Increased expression of PAI-1 in the artery wall promotes neointima growth after balloon injury. Therefore, despite encouraging data generated in other animal models, PAI-1 is not a promising agent for gene therapy to prevent restenosis. Moreover, our data associate elevated PAI-1 expression with fibrin(ogen) accumulation and increased cell proliferation. These data suggest a mechanism to explain the association between elevated PAI-1 expression and the progression of arterial disease.


Subject(s)
Angioplasty, Balloon , Carotid Artery Diseases/pathology , Plasminogen Activator Inhibitor 1/administration & dosage , Tunica Intima/drug effects , Tunica Intima/pathology , Adenoviridae/genetics , Angioplasty, Balloon/adverse effects , Animals , Carotid Artery Diseases/etiology , Carotid Artery Diseases/prevention & control , Cell Division/drug effects , Disease Models, Animal , Disease Progression , Fibrin/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Immunohistochemistry , Plasminogen Activator Inhibitor 1/genetics , Rats , Transduction, Genetic , Tunica Intima/metabolism , Tunica Media/drug effects , Tunica Media/pathology
18.
Br J Surg ; 88(2): 286-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11167882

ABSTRACT

BACKGROUND: Adhesion formation is a common cause of complications following surgery. A reduction in peritoneal fibrinolytic capacity during operation is a key mechanism in the early formation of adhesions. An increase in the main inhibitor of fibrinolysis, plasminogen activator inhibitor type 1 (PAI-1), is a major factor in the loss of fibrinolytic activity. The aim of this study was to investigate if inhibition of PAI-1 could reduce the formation of adhesions after surgery. METHODS: Mice (n = 53) were subjected to a standard surgical procedure in order to induce adhesion formation to the abdominal side wall. At the conclusion of the operation, fragments for antigen binding of polyclonal rabbit antibody against PAI-1 (PRAP-1) were injected intraperitoneally, at two different concentrations. Control animals received an equal volume of the vehicle (saline). One week after operation adhesion formation was quantified. RESULTS: Both doses of PRAP-1 significantly reduced adhesion formation compared with the saline control (P = 0.003 and P = 0.002). There were no signs of bleeding in the postoperative period or at reoperation. CONCLUSION: The present observations lend further support to the hypothesis of a pivotal role of fibrinolysis in the early formation of adhesions, and open up new possibilities for adhesion reduction by inhibiting PAI-1.


Subject(s)
Peritoneal Cavity/surgery , Plasminogen Activator Inhibitor 1/administration & dosage , Tissue Adhesions/prevention & control , Animals , Female , Mice
19.
Blood ; 90(7): 2738-46, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9326241

ABSTRACT

Uveal melanoma is the most common intraocular malignancy in adults and results in the death of 50% of the patients. Plasminogen activators (PA) are believed to facilitate tumor metastasis by promoting invasion of tissue barriers. The present study explored the possibility of preventing the metastasis of intraocular melanomas by disrupting plasminogen activator function through gene transfer. A replication-deficient adenovirus vector was used for the in vivo transfer of plasminogen activator inhibitor type 1 (PAI-1) cDNA. Intraocular injection of an adenovirus vector (AdCMV-PAI-1) expressing plasminogen activator inhibitor-1 resulted in: (1) the transduction of more than 95% of human and murine uveal melanoma cells in the eyes of nude mice; (2) a 50% reduction in the number of animals developing liver metastases; and (3) a 78% reduction in the metastatic tumor burden in animals that eventually developed metastases. In other experiments intravenous injections of AdCMV-PAI-1 resulted in transduction of normal liver cells and culminated in a sharp reduction in the incidence of metastases and a significant prolongation of host survival. The results support the feasibility of disruption of PA function through gene transfer as a therapeutic strategy for preventing metastases and prolonging host survival.


Subject(s)
Adenoviruses, Human/genetics , Defective Viruses/genetics , Genetic Therapy , Genetic Vectors/genetics , Melanoma, Experimental/therapy , Melanoma/therapy , Neoplasm Metastasis/prevention & control , Plasminogen Activator Inhibitor 1/therapeutic use , Uveal Neoplasms/therapy , Adult , Animals , Evaluation Studies as Topic , Humans , Liver Neoplasms, Experimental/prevention & control , Liver Neoplasms, Experimental/secondary , Liver Neoplasms, Experimental/therapy , Melanoma/pathology , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Transplantation , Plasminogen Activator Inhibitor 1/administration & dosage , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activators/antagonists & inhibitors , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Transfection , Transplantation, Heterologous , Uveal Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...