Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(6): 107313, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657864

ABSTRACT

Sortilin-related receptor 1 (SORL1) is an intracellular sorting receptor genetically implicated in Alzheimer's disease (AD) that impacts amyloid precursor protein trafficking. The objective of these studies was to test the hypothesis that SORL1 binds tau, modulates its cellular trafficking and impacts the aggregation of cytoplasmic tau induced by pathological forms of tau. Using surface plasmon resonance measurements, we observed high-affinity binding of tau to SORL1 and the vacuolar protein sorting 10 domain of SORL1. Interestingly, unlike LDL receptor-related protein 1, SORL1 binds tau at both pH 7.4 and pH 5.5, revealing its ability to bind tau at endosomal pH. Immunofluorescence studies confirmed that exogenously added tau colocalized with SORL1 in H4 neuroglioma cells, while overexpression of SORL1 in LDL receptor-related protein 1-deficient Chinese hamster ovary (CHO) cells resulted in a marked increase in the internalization of tau, indicating that SORL1 can bind and mediate the internalization of monomeric forms of tau. We further demonstrated that SORL1 mediates tau seeding when tau RD P301S FRET biosensor cells expressing SORL1 were incubated with high molecular weight forms of tau isolated from the brains of patients with AD. Seeding in H4 neuroglioma cells is significantly reduced when SORL1 is knocked down with siRNA. Finally, we demonstrate that the N1358S mutant of SORL1 significantly increases tau seeding when compared to WT SORL1, identifying for the first time a potential mechanism that connects this specific SORL1 mutation to Alzheimer's disease. Together, these studies identify SORL1 as a receptor that contributes to trafficking and seeding of pathogenic tau.


Subject(s)
Cricetulus , LDL-Receptor Related Proteins , Membrane Transport Proteins , tau Proteins , Humans , tau Proteins/metabolism , tau Proteins/genetics , Animals , CHO Cells , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cell Line, Tumor , Protein Binding , Protein Transport
2.
PLoS One ; 18(5): e0285499, 2023.
Article in English | MEDLINE | ID: mdl-37235594

ABSTRACT

Traumatic brain injury (TBI) is associated with cardiovascular mortality in humans. Enhanced sympathetic activity following TBI may contribute to accelerated atherosclerosis. The effect of beta1-adrenergic receptor blockade on atherosclerosis progression induced by TBI was studied in apolipoprotein E deficient mice. Mice were treated with metoprolol or vehicle following TBI or sham operation. Mice treated with metoprolol experienced a reduced heart rate with no difference in blood pressure. Six weeks following TBI, mice were sacrificed for analysis of atherosclerosis. Total surface area and lesion thickness, analyzed at the level of the aortic valve, was found to be increased in mice receiving TBI with vehicle treatment but this effect was ameliorated in TBI mice receiving metoprolol. No effect of metoprolol on atherosclerosis was observed in mice receiving only sham operation. In conclusion, accelerated atherosclerosis following TBI is reduced with beta-adrenergic receptor antagonism. Beta blockers may be useful to reduce vascular risk associated with TBI.


Subject(s)
Atherosclerosis , Brain Injuries, Traumatic , Animals , Mice , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Atherosclerosis/pathology , Blood Pressure , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Disease Models, Animal , Metoprolol/pharmacology , Metoprolol/therapeutic use , Mice, Inbred C57BL
3.
J Neuroinflammation ; 20(1): 56, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36864439

ABSTRACT

BACKGROUND: Noradrenergic neurons in the locus coeruleus (LC) are the primary source of norepinephrine (NE) in the brain and degeneration of these neurons is reported in the early stages of Parkinson's disease (PD), even prior to dopaminergic neuron degeneration in the substantia nigra (SN), which is a hallmark of PD pathology. NE depletion is generally associated with increased PD pathology in neurotoxin-based PD models. The effect of NE depletion in other models of PD-like α-synuclein-based models is largely unexplored. In PD models and in human patients, ß-adrenergic receptors' (AR) signaling is associated with a reduction of neuroinflammation and PD pathology. However, the effect of NE depletion in the brain and the extent of NE and ß-ARs signaling involvement in neuroinflammation, and dopaminergic neuron survival is poorly understood. METHODS: Two mouse models of PD, a 6OHDA neurotoxin-based model and a human α-synuclein (hα-SYN) virus-based model of PD, were used. DSP-4 was used to deplete NE levels in the brain and its effect was confirmed by HPLC with electrochemical detection. A pharmacological approach was used to mechanistically understand the impact of DSP-4 in the hα-SYN model of PD using a norepinephrine transporter (NET) and a ß-AR blocker. Epifluorescence and confocal imaging were used to study changes in microglia activation and T-cell infiltration after ß1-AR and ß2-AR agonist treatment in the hα-SYN virus-based model of PD. RESULTS: Consistent with previous studies, we found that DSP-4 pretreatment increased dopaminergic neuron loss after 6OHDA injection. In contrast, DSP-4 pretreatment protected dopaminergic neurons after hα-SYN overexpression. DSP-4-mediated protection of dopaminergic neurons after hα-SYN overexpression was dependent on ß-AR signaling since using a ß-AR blocker prevented DSP-4-mediated dopaminergic neuron protection in this model of PD. Finally, we found that the ß-2AR agonist, clenbuterol, reduced microglia activation, T-cell infiltration, and dopaminergic neuron degeneration, whereas xamoterol a ß-1AR agonist showed increased neuroinflammation, blood brain barrier permeability (BBB), and dopaminergic neuron degeneration in the context of hα-SYN-mediated neurotoxicity. CONCLUSIONS: Our data demonstrate that the effects of DSP-4 on dopaminergic neuron degeneration are model specific, and suggest that in the context of α-SYN-driven neuropathology, ß2-AR specific agonists may have therapeutic benefit in PD.


Subject(s)
Neurotoxicity Syndromes , Parkinson Disease , Animals , Humans , Mice , alpha-Synuclein , Dopaminergic Neurons , Nerve Degeneration , Neuroinflammatory Diseases , Neurotoxins , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism
4.
J Diabetes Complications ; 36(11): 108318, 2022 11.
Article in English | MEDLINE | ID: mdl-36228562

ABSTRACT

This white paper examines the current challenges for treating ischemic stroke in diabetic patients. The need for a greater understanding of the mechanisms that underlie the relationship between diabetes and the cerebral vascular responses to ischemia is discussed. The critical need to improve the efficacy and safety of thrombolysis is addressed, as is the need for a better characterization the off-target actions of tPA, the only currently approved thrombolytic for the treatment of ischemic stroke.


Subject(s)
Brain Ischemia , Diabetes Mellitus , Ischemic Stroke , Stroke , Humans , Tissue Plasminogen Activator , Brain Ischemia/complications , Brain Ischemia/therapy , Stroke/complications , Stroke/therapy , Diabetes Mellitus/epidemiology , Diabetes Mellitus/therapy , Treatment Outcome
5.
Blood ; 140(4): 388-400, 2022 07 28.
Article in English | MEDLINE | ID: mdl-35576527

ABSTRACT

The current standard of care for moderate to severe ischemic stroke is thrombolytic therapy with tissue plasminogen activator (tPA). Treatment with tPA can significantly improve neurologic outcomes; however, thrombolytic therapy is associated with an increased risk of intracerebral hemorrhage (ICH). The risk of hemorrhage significantly limits the use of thrombolytic therapy, and identifying pathways induced by tPA that increase this risk could provide new therapeutic options to extend thrombolytic therapy to a wider patient population. Here, we investigate the role of protein kinase Cß (PKCß) phosphorylation of the tight junction protein occludin during ischemic stroke and its role in cerebrovascular permeability. We show that activation of this pathway by tPA is associated with an increased risk of ICH. Middle cerebral artery occlusion (MCAO) increased phosphorylation of occludin serine 490 (S490) in the ischemic penumbra in a tPA-dependent manner, as tPA-/- mice were significantly protected from MCAO-induced occludin phosphorylation. Intraventricular injection of tPA in the absence of ischemia was sufficient to induce occludin phosphorylation and vascular permeability in a PKCß-dependent manner. Blocking occludin phosphorylation, either by targeted expression of a non-phosphorylatable form of occludin (S490A) or by pharmacologic inhibition of PKCß, reduced MCAO-induced permeability and improved functional outcome. Furthermore, inhibiting PKCß after MCAO prevented ICH associated with delayed thrombolysis. These results show that PKCß phosphorylation of occludin is a downstream mediator of tPA-induced cerebrovascular permeability and suggest that PKCß inhibitors could improve stroke outcome and prevent ICH associated with delayed thrombolysis, potentially extending the window for thrombolytic therapy in stroke.


Subject(s)
Ischemic Stroke , Stroke , Animals , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/etiology , Fibrinolytic Agents/therapeutic use , Humans , Infarction, Middle Cerebral Artery/drug therapy , Mice , Occludin/genetics , Occludin/metabolism , Phosphorylation , Stroke/complications , Stroke/etiology , Thrombolytic Therapy/adverse effects , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/metabolism
6.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768908

ABSTRACT

Ischemic stroke is the most common type of stroke and thrombolytic therapy is the only approved treatment. However, current thrombolytic therapy with tissue plasminogen activator (tPA) is often hampered by the increased risk of hemorrhage. Plasmin, a direct fibrinolytic, has a significantly superior hemostatic safety profile; however, if injected intravenously it becomes rapidly inactivated by anti-plasmin. Nanoformulations have been shown to increase drug stability and half-life and hence could be applied to increase the plasmin therapeutic efficacy. Here in this paper, we report a novel heparin and arginine-based plasmin nanoformulation that exhibits increased plasmin stability and efficacy. In vitro studies revealed significant plasmin stability in the presence of anti-plasmin and efficient fibrinolytic activity. In addition, these particles showed no significant toxicity or oxidative stress effects in human brain microvascular endothelial cells, and no significant blood brain barrier permeability. Further, in a mouse photothrombotic stroke model, plasmin nanoparticles exhibited significant efficacy in reducing stroke volume without overt intracerebral hemorrhage (ICH) compared to free plasmin treatment. The study shows the potential of a plasmin nanoformulation in ischemic stroke therapy.


Subject(s)
Arginine/chemistry , Fibrinolysin/administration & dosage , Heparin/chemistry , Ischemic Stroke/therapy , Nanoparticles/administration & dosage , Thrombolytic Therapy/methods , Animals , Blood-Brain Barrier , Fibrinolysin/chemistry , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/chemistry , Humans , Infarction, Middle Cerebral Artery/complications , Ischemic Stroke/etiology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Male , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry
7.
J Biol Chem ; 294(5): 1464-1477, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30510136

ABSTRACT

Plasminogen activator inhibitor type-1 (PAI-1) is a serine protease inhibitor (serpin) implicated in numerous pathological processes, including coronary heart disease, arterial and venous thrombosis, and chronic fibrotic diseases. These associations have made PAI-1 an attractive pharmaceutical target. However, the complexity of the serpin inhibitory mechanism, the inherent metastability of serpins, and the high-affinity association of PAI-1 with vitronectin in vivo have made it difficult to identify pharmacologically effective small-molecule inhibitors. Moreover, the majority of current small-molecule PAI-1 inhibitors are poor pharmaceutical candidates. To this end and to find leads that can be efficiently applied to in vivo settings, we developed a dual-reporter high-throughput screen (HTS) that reduced the rate of nonspecific and promiscuous hits and identified leads that inhibit human PAI-1 in the high-protein environments present in vivo Using this system, we screened >152,000 pure compounds and 27,000 natural product extracts (NPEs), reducing the apparent hit rate by almost 10-fold compared with previous screening approaches. Furthermore, screening in a high-protein environment permitted the identification of compounds that retained activity in both ex vivo plasma and in vivo Following lead identification, subsequent medicinal chemistry and structure-activity relationship (SAR) studies identified a lead clinical candidate, MDI-2268, having excellent pharmacokinetics, potent activity against vitronectin-bound PAI-1 in vivo, and efficacy in a murine model of venous thrombosis. This rigorous HTS approach eliminates promiscuous candidate leads, significantly accelerates the process of identifying PAI-1 inhibitors that can be rapidly deployed in vivo, and has enabled identification of a potent lead compound.


Subject(s)
Calorimetry/methods , Fluorescence , High-Throughput Screening Assays , Plasminogen Activator Inhibitor 1/chemistry , Serine Proteinase Inhibitors/chemistry , Serpins/metabolism , Small Molecule Libraries/pharmacology , Animals , Genes, Reporter , Humans , Mice , Plasminogen Activator Inhibitor 1/metabolism , Rats , Serine Proteinase Inhibitors/metabolism
8.
Front Cell Neurosci ; 9: 385, 2015.
Article in English | MEDLINE | ID: mdl-26500491

ABSTRACT

Current therapies for Traumatic brain injury (TBI) focus on stabilizing individuals and on preventing further damage from the secondary consequences of TBI. A major complication of TBI is cerebral edema, which can be caused by the loss of blood brain barrier (BBB) integrity. Recent studies in several CNS pathologies have shown that activation of latent platelet derived growth factor-CC (PDGF-CC) within the brain can promote BBB permeability through PDGF receptor α (PDGFRα) signaling, and that blocking this pathway improves outcomes. In this study we examine the efficacy for the treatment of TBI of an FDA approved antagonist of the PDGFRα, Imatinib. Using a murine model we show that Imatinib treatment, begun 45 min after TBI and given twice daily for 5 days, significantly reduces BBB dysfunction. This is associated with significantly reduced lesion size 24 h, 7 days, and 21 days after TBI, reduced cerebral edema, determined from apparent diffusion co-efficient (ADC) measurements, and with the preservation of cognitive function. Finally, analysis of cerebrospinal fluid (CSF) from human TBI patients suggests a possible correlation between high PDGF-CC levels and increased injury severity. Thus, our data suggests a novel strategy for the treatment of TBI with an existing FDA approved antagonist of the PDGFRα.

9.
Ann Clin Transl Neurol ; 2(7): 722-38, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26273685

ABSTRACT

OBJECTIVE: A growing body of evidence suggests that increased blood-brain barrier (BBB) permeability can contribute to the development of seizures. The protease tissue plasminogen activator (tPA) has been shown to promote BBB permeability and susceptibility to seizures. In this study, we examined the pathway regulated by tPA in seizures. METHODS: An experimental model of kainate-induced seizures was used in genetically modified mice, including mice deficient in tPA (tPA (-/-) ), its inhibitor neuroserpin (Nsp (-/-) ), or both (Nsp:tPA (-/-) ), and in mice conditionally deficient in the platelet-derived growth factor receptor alpha (PDGFRα). RESULTS: Compared to wild-type (WT) mice, Nsp (-/-) mice have significantly reduced latency to seizure onset and generalization; whereas tPA (-/-) mice have the opposite phenotype, as do Nsp:tPA (-/-) mice. Furthermore, interventions that maintain BBB integrity delay seizure propagation, whereas osmotic disruption of the BBB in seizure-resistant tPA (-/-) mice dramatically reduces the time to seizure onset and accelerates seizure progression. The phenotypic differences in seizure progression between WT, tPA (-/-) , and Nsp (-/-) mice are also observed in electroencephalogram recordings in vivo, but absent in ex vivo electrophysiological recordings where regulation of the BBB is no longer necessary to maintain the extracellular environment. Finally, we demonstrate that these effects on seizure progression are mediated through signaling by PDGFRα on perivascular astrocytes. INTERPRETATION: Together, these data identify a specific molecular pathway involving tPA-mediated PDGFRα signaling in perivascular astrocytes that regulates seizure progression through control of the BBB. Inhibition of PDGFRα signaling and maintenance of BBB integrity might therefore offer a novel clinical approach for managing seizures.

11.
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
13.
Am J Pathol ; 180(3): 1136-1144, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22230248

ABSTRACT

Platelet-derived growth factors (PDGFs) and their tyrosine kinase receptors (PDGFRs) are known to play important roles during development of the lungs, central nervous system (CNS), and skeleton and in several diseases. PDGF-C is a ligand for the tyrosine kinase receptor PDGFRα. Mutations in the gene encoding PDGF-C have been linked to clefts of the lip and/or palate in humans, and ablation of PDGF-C in 129/Sv background mice results in death during the perinatal period. In this study, we report that ablation of PDGF-C in C57BL/6 mice results in a milder phenotype than in 129/Sv mice, and we present a phenotypic characterization of PDGF-C deficiency in the adult murine CNS. Multiple congenital defects were observed in the CNS of PDGF-C-null C57BL/6 mice, including cerebral vascular abnormalities with abnormal vascular smooth muscle cell coverage. In vivo imaging of mice deficient in PDGF-C also revealed cerebral ventricular abnormalities, such as asymmetry of the lateral ventricles and hypoplasia of the septum, reminiscent of cavum septum pellucidum in humans. We further noted that PDGF-C-deficient mice displayed a distorted ependymal lining of the lateral ventricles, and we found evidence of misplaced neurons in the ventricular lining. We conclude that PDGF-C plays a critical role in the development of normal cerebral ventricles and neuroependymal integrity as well as in normal cerebral vascularization.


Subject(s)
Cerebral Ventricles/abnormalities , Ependyma/abnormalities , Intracranial Arteriovenous Malformations/etiology , Lymphokines/deficiency , Platelet-Derived Growth Factor/deficiency , Animals , Lymphokines/physiology , Mice , Mice, Inbred C57BL , Platelet-Derived Growth Factor/physiology , Receptor, Platelet-Derived Growth Factor alpha/metabolism
14.
J Am Heart Assoc ; 1(5): e002584, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23316294

ABSTRACT

BACKGROUND: Mineralocorticoid receptor (MR) antagonists have protective effects in the brain during experimental ischemic stroke, and we have previously demonstrated a key role for myeloid MR during stroke pathogenesis. In this study, we explore both model- and sex-specific actions of myeloid MR during ischemic stroke. METHODS AND RESULTS: The MR antagonist eplerenone significantly reduced the infarct size in male (control, 99.5 mm(3); eplerenone, 74.2 mm(3); n=8 to 12 per group) but not female (control, 84.0 mm(3); eplerenone, 83.7 mm(3); n=6 to 7 per group) mice after transient (90-minute) middle cerebral artery occlusion. In contrast to MR antagonism, genetic ablation of myeloid MR in female mice significantly reduced infarct size (myeloid MR knockout, 9.4 mm(3) [5.4 to 36.6]; control, 66.0 mm(3) [50.0 to 81.4]; n=6 per group) after transient middle cerebral artery occlusion. This was accompanied by reductions in inflammatory gene expression and improvement in neurological function. In contrast to ischemia-reperfusion, myeloid MR-knockout mice were not protected from permanent middle cerebral artery occlusion. The infarct size and inflammatory response after permanent occlusion showed no evidence of protection by myeloid MR knockout in photothrombotic and intraluminal filament models of permanent occlusion. CONCLUSIONS: These studies demonstrate that MR antagonism is protective in male but not female mice during transient middle cerebral artery occlusion, whereas genetic ablation of myeloid MR is protective in both male and female mice. They also highlight important mechanistic differences in the role of myeloid cells in different models of stroke and confirm that specific myeloid phenotypes play key roles in stroke protection.


Subject(s)
Brain Ischemia/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Mineralocorticoid Receptor Antagonists/pharmacology , Receptors, Mineralocorticoid/metabolism , Spironolactone/analogs & derivatives , Stroke/drug therapy , Animals , Brain Ischemia/physiopathology , Disease Models, Animal , Eplerenone , Female , Gene Expression Profiling , Immunohistochemistry , Infarction, Middle Cerebral Artery/physiopathology , Male , Mice , Mice, Knockout , Spironolactone/pharmacology , Stroke/physiopathology
15.
J Biol Chem ; 285(11): 7892-902, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20061381

ABSTRACT

Plasminogen activator inhibitor type 1, (PAI-1) the primary inhibitor of the tissue-type (tPA) and urokinase-type (uPA) plasminogen activators, has been implicated in a wide range of pathological processes, making it an attractive target for pharmacologic inhibition. Currently available small-molecule inhibitors of PAI-1 bind with relatively low affinity and do not inactivate PAI-1 in the presence of its cofactor, vitronectin. To search for novel PAI-1 inhibitors with improved potencies and new mechanisms of action, we screened a library selected to provide a range of biological activities and structural diversity. Five potential PAI-1 inhibitors were identified, and all were polyphenolic compounds including two related, naturally occurring plant polyphenols that were structurally similar to compounds previously shown to provide cardiovascular benefit in vivo. Unique second generation compounds were synthesized and characterized, and several showed IC(50) values for PAI-1 between 10 and 200 nm. This represents an enhanced potency of 10-1000-fold over previously reported PAI-1 inactivators. Inhibition of PAI-1 by these compounds was reversible, and their primary mechanism of action was to block the initial association of PAI-1 with a protease. Consistent with this mechanism and in contrast to previously described PAI-1 inactivators, these compounds inactivate PAI-1 in the presence of vitronectin. Two of the compounds showed efficacy in ex vivo plasma and one blocked PAI-1 activity in vivo in mice. These data describe a novel family of high affinity PAI-1-inactivating compounds with improved characteristics and in vivo efficacy, and suggest that the known cardiovascular benefits of dietary polyphenols may derive in part from their inactivation of PAI-1.


Subject(s)
Drug Design , Flavonoids/pharmacology , Phenols/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Protease Inhibitors/pharmacology , Serpins/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Flavonoids/chemical synthesis , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenols/chemical synthesis , Polyphenols , Protease Inhibitors/chemical synthesis , Recombinant Proteins/metabolism , Serpin E2 , Surface Plasmon Resonance , Vitronectin/metabolism
16.
J Clin Invest ; 119(5): 1136-49, 2009 May.
Article in English | MEDLINE | ID: mdl-19381014

ABSTRACT

Leukocyte and platelet accumulation at sites of cerebral ischemia exacerbate cerebral damage. The ectoenzyme CD39 on the plasmalemma of endothelial cells metabolizes ADP to suppress platelet accumulation in the ischemic brain. However, the role of leukocyte surface CD39 in regulating monocyte and neutrophil trafficking in this setting is not known. Here we have demonstrated in mice what we believe to be a novel mechanism by which CD39 on monocytes and neutrophils regulates their own sequestration into ischemic cerebral tissue, by catabolizing nucleotides released by injured cells, thereby inhibiting their chemotaxis, adhesion, and transmigration. Bone marrow reconstitution and provision of an apyrase, an enzyme that hydrolyzes nucleoside tri- and diphosphates, each normalized ischemic leukosequestration and cerebral infarction in CD39-deficient mice. Leukocytes purified from Cd39-/- mice had a markedly diminished capacity to phosphohydrolyze adenine nucleotides and regulate platelet reactivity, suggesting that leukocyte ectoapyrases modulate the ambient vascular nucleotide milieu. Dissipation of ATP by CD39 reduced P2X7 receptor stimulation and thereby suppressed baseline leukocyte alphaMbeta2-integrin expression. As alphaMbeta2-integrin blockade reversed the postischemic, inflammatory phenotype of Cd39-/- mice, these data suggest that phosphohydrolytic activity on the leukocyte surface suppresses cell-cell interactions that would otherwise promote thrombosis or inflammation. These studies indicate that CD39 on both endothelial cells and leukocytes reduces inflammatory cell trafficking and platelet reactivity, with a consequent reduction in tissue injury following cerebral ischemic challenge.


Subject(s)
Antigens, CD/physiology , Apyrase/physiology , Cell Movement/physiology , Leukocytes/cytology , 5'-Nucleotidase/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Apyrase/pharmacology , Bone Marrow Transplantation , Brain Ischemia/genetics , Brain Ischemia/pathology , Cell Line, Tumor , Cell Movement/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Leukocytes/drug effects , Leukocytes/metabolism , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Platelet Activation/physiology , Purinergic P2 Receptor Antagonists , RNA Interference , Receptors, Purinergic P2/genetics , Transplantation Chimera/physiology
17.
Nat Med ; 14(7): 731-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18568034

ABSTRACT

Thrombolytic treatment of ischemic stroke with tissue plasminogen activator (tPA) is markedly limited owing to concerns about hemorrhagic complications and the requirement that tPA be administered within 3 h of symptoms. Here we report that tPA activation of latent platelet-derived growth factor-CC (PDGF-CC) may explain these limitations. Intraventricular injection of tPA or active PDGF-CC, in the absence of ischemia, leads to significant increases in cerebrovascular permeability. In contrast, co-injection of neutralizing antibodies to PDGF-CC with tPA blocks this increased permeability, indicating that PDGF-CC is a downstream substrate of tPA within the neurovascular unit. These effects are mediated through activation of PDGF-alpha receptors (PDGFR-alpha) on perivascular astrocytes, and treatment of mice with the PDGFR-alpha antagonist imatinib after ischemic stroke reduces both cerebrovascular permeability and hemorrhagic complications associated with late administration of thrombolytic tPA. These data demonstrate that PDGF signaling regulates blood-brain barrier permeability and suggest potential new strategies for stroke treatment.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Lymphokines/metabolism , Platelet-Derived Growth Factor/metabolism , Stroke/drug therapy , Tissue Plasminogen Activator/metabolism , Animals , Benzamides , Blood-Brain Barrier/pathology , Brain/blood supply , Brain/ultrastructure , Fibrinolytic Agents/metabolism , Imatinib Mesylate , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors
18.
J Biol Chem ; 282(21): 15679-89, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17403662

ABSTRACT

The serine proteinase inhibitor, plasminogen activator inhibitor type-1 (PAI-1), binds to the adhesion protein vitronectin with high affinity at a site that is located directly adjacent to the vitronectin RGD integrin binding sequence. The binding of PAI-1 to vitronectin sterically blocks integrin access to this site and completely inhibits the binding of purified integrins to vitronectin; however, its inhibition of endothelial and smooth muscle cell adhesion to vitronectin is at most 50-75%. Because PAI-1 binds vitronectin with approximately 10-100-fold higher affinity than purified integrins, we have analyzed the mechanism whereby these cells are able to overcome this obstacle. Our studies exclude proteolytic removal of PAI-1 from vitronectin as the mechanism, and show instead that cell adhesion in the presence of PAI-1 is dependent on integrin-cytoskeleton engagement. Disrupting endothelial or smooth muscle cell actin polymerization and/or focal adhesion assembly reduces cell adhesion to vitronectin in the presence of PAI-1 to levels similar to that observed for the binding of purified integrins to vitronectin. Furthermore, endothelial cell, but not smooth muscle cell adhesion to vitronectin in the presence of PAI-1 requires both polymerized microtubules and actin, further demonstrating the importance of the cytoskeleton for integrin-mediated adhesion. Finally, we show that cell adhesion in the presence of PAI-1 leads to colocalization of PAI-1 with the integrins alphavbeta3 and alphavbeta5 at the cell-matrix interface.


Subject(s)
Cytoskeleton/metabolism , Endothelial Cells/metabolism , Integrin alphaVbeta3/metabolism , Integrins/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, Vitronectin/metabolism , Vitronectin/metabolism , Actins/metabolism , Animals , Cell Adhesion/physiology , Endothelial Cells/cytology , Humans , Myocytes, Smooth Muscle/cytology , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding
19.
EMBO J ; 25(9): 1860-70, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16601674

ABSTRACT

Migration of activated macrophages is essential for resolution of acute inflammation and the initiation of adaptive immunity. Here, we show that efficient macrophage migration in inflammatory environment depends on Mac-1 recognition of a binary complex consisting of fibrin within the provisional matrix and the protease tPA (tissue-type plasminogen activator). Subsequent neutralization of tPA by its inhibitor PAI-1 enhances binding of the integrin-protease-inhibitor complex to the endocytic receptor LRP (lipoprotein receptor-related protein), triggering a switch from cell adhesion to cell detachment. Genetic inactivation of Mac-1, tPA, PAI-1 or LRP but not the protease uPA abrogates macrophage migration. The defective macrophage migration in PAI-1-deficient mice can be restored by wild-type but not by a mutant PAI-1 that does not interact with LRP. In vitro analysis shows that tPA promotes Mac-1-mediated adhesion, whereas PAI-1 and LRP facilitate its transition to cell retraction. Our results emphasize the importance of ordered transitions both temporally and spatially between individual steps of cell migration, and support a model where efficient migration of inflammatory macrophages depends on cooperation of three physiologically prominent systems (integrins, coagulation and fibrinolysis, and endocytosis).


Subject(s)
Cell Movement/immunology , LDL-Receptor Related Proteins/metabolism , Macrophage-1 Antigen/metabolism , Macrophages/immunology , Plasminogen Activator Inhibitor 1/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Endocytosis , Fibrinogen/metabolism , Humans , Immunoprecipitation , Integrases/metabolism , LDL-Receptor Related Proteins/analysis , LDL-Receptor Related Proteins/genetics , Macrophage-1 Antigen/analysis , Macrophage-1 Antigen/genetics , Macrophages/chemistry , Macrophages/metabolism , Mice , Mice, Knockout , Plasminogen Activator Inhibitor 1/genetics , Tissue Plasminogen Activator/antagonists & inhibitors , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL