Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
2.
J Neuroinflammation ; 18(1): 289, 2021 Dec 11.
Article in English | MEDLINE | ID: mdl-34895246

ABSTRACT

BACKGROUND: Current therapies targeting several neurotransmitter systems are only able to partially mitigate the symptoms of stress- and trauma-related disorder. Stress and trauma-related disorders lead to a prominent inflammatory response in humans, and in pre-clinical models. However, mechanisms underlying the induction of neuroinflammatory response in PTSD and anxiety disorders are not clearly understood. The present study investigated the mechanism underlying the activation of proinflammatory NLRP3 inflammasome and IL1ß in mouse models of stress. METHODS: We used two mouse models of stress, i.e., mice subjected to physical restraint stress with brief underwater submersion, and predator odor stress. Mice were injected with MCC950, a small molecule specific inhibitor of NLRP3 activation. To pharmacologically inhibit BTK, a specific inhibitor ibrutinib was used. To validate the observation from ibrutinib studies, a separate group of mice was injected with another BTK-specific inhibitor LFM-A13. Seven days after the induction of stress, mice were examined for anxious behavior using open field test (OFT), light-dark test (LDT), and elevated plus maze test (EPM). Following the behavior tests, hippocampus and amygdale were extracted and analyzed for various components of NLRP3-caspase 1-IL1ß pathway. Plasma and peripheral blood mononuclear cells were also used to assess the induction of NLRP3-Caspase 1-IL-1ß pathway in stressed mice. RESULTS: Using two different pre-clinical models of stress, we demonstrate heightened anxious behavior in female mice as compared to their male counterparts. Stressed animals exhibited upregulation of proinflammatory IL1ß, IL-6, Caspase 1 activity and NLRP3 inflammasome activation in brain, which were significantly higher in female mice. Pharmacological inhibition of NLRP3 inflammasome activation led to anxiolysis as well as attenuated neuroinflammatory response. Further, we observed induction of activated Bruton's tyrosine kinase (BTK), an upstream positive-regulator of NLRP3 inflammasome activation, in hippocampus and amygdala of stressed mice. Next, we conducted proof-of-concept pharmacological BTK inhibitor studies with ibrutinib and LFM-A13. In both sets of experiments, we found BTK inhibition led to anxiolysis and attenuated neuroinflammation, as indicated by significant reduction of NLRP3 inflammasome and proinflammatory IL-1ß in hippocampus and amygdala. Analysis of plasma and peripheral blood mononuclear cells indicated peripheral induction of NLRP3-caspase 1-IL1ß pathway in stressed mice. CONCLUSION: Our study identified BTK as a key upstream regulator of neuroinflammation, which drives anxiogenic behavior in mouse model of stress. Further, we demonstrated the sexually divergent activation of BTK, providing a clue to heightened neuroinflammation and anxiogenic response to stress in females as compared to their male counterparts. Our data from the pharmacological inhibition studies suggest BTK as a novel target for the development of potential clinical treatment of PTSD and anxiety disorders. Induction of pBTK and NLRP3 in peripheral blood mononuclear cells of stressed mice suggest the potential effect of stress on systemic inflammation.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Anxiety/enzymology , Disease Models, Animal , Inflammation Mediators/metabolism , Stress, Psychological/enzymology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Amides/pharmacology , Animals , Anxiety/drug therapy , Anxiety/psychology , Female , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitriles/pharmacology , Odorants , Piperidines/pharmacology , Piperidines/therapeutic use , Rats , Restraint, Physical/adverse effects , Stress, Psychological/drug therapy , Stress, Psychological/psychology
3.
Carcinogenesis ; 39(10): 1245-1253, 2018 10 08.
Article in English | MEDLINE | ID: mdl-29982410

ABSTRACT

Breast cancer is a leading cause of cancer-related mortality in women. Triple-negative breast cancer (TNBC; HER2-, ER-/PR-) is an aggressive subtype prone to drug resistance and metastasis, which is characterized by high intratumor microvascular density (iMVD) resulting from angiogenesis. However, the mechanisms contributing to the aggressive phenotypes of TNBC remain elusive. We recently reported that down-regulation of exchange factor directly activated by cyclic AMP (cAMP), also known as EPAC1, leads to a reduction in metastatic properties including proliferation and cell migration in TNBC cell lines. Here, we report that EPAC1 supports TNBC-induced angiogenesis, tumor cell migration and invasiveness as well as pro-metastatic phenotypes in endothelial cells induced through the tumor secretome. Using an approach that integrates proteomics with bioinformatics and gene ontologies, we elucidate that EPAC1 supports a tumor-secreted network of angiogenic, cell adhesion and cell migratory pathways. Using confocal microscopy, we show that signaling molecules involved in focal adhesion, including Paxillin and MENA, are down-regulated in the absence of EPAC1, and electric cell substrate impedance sensing technique confirmed a role for EPAC1 on TNBC-induced endothelial cell permeability. Finally, to provide a translational bridge, we studied iMVD and therapy response using a primary human tumor explant assay, CANscriptTM, which suggests a link between therapy-modulated neovascularization and drug sensitivity. These data provide mechanistic insight into the role of EPAC1 in regulating the tumor microenvironment, iMVD and cancer cell-induced angiogenesis, a dynamic mechanism under drug pressure that may associate to treatment failure.


Subject(s)
Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neovascularization, Pathologic/metabolism , Triple Negative Breast Neoplasms/metabolism , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Cell Movement , Electric Impedance , Humans , Immunohistochemistry , Microvessels/pathology , Signal Transduction , Triple Negative Breast Neoplasms/pathology
4.
Nature ; 485(7399): 512-6, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22622580

ABSTRACT

Human apolipoprotein E has three isoforms: APOE2, APOE3 and APOE4. APOE4 is a major genetic risk factor for Alzheimer's disease and is associated with Down's syndrome dementia and poor neurological outcome after traumatic brain injury and haemorrhage. Neurovascular dysfunction is present in normal APOE4 carriers and individuals with APOE4-associated disorders. In mice, lack of Apoe leads to blood-brain barrier (BBB) breakdown, whereas APOE4 increases BBB susceptibility to injury. How APOE genotype affects brain microcirculation remains elusive. Using different APOE transgenic mice, including mice with ablation and/or inhibition of cyclophilin A (CypA), here we show that expression of APOE4 and lack of murine Apoe, but not APOE2 and APOE3, leads to BBB breakdown by activating a proinflammatory CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes. This, in turn, leads to neuronal uptake of multiple blood-derived neurotoxic proteins, and microvascular and cerebral blood flow reductions. We show that the vascular defects in Apoe-deficient and APOE4-expressing mice precede neuronal dysfunction and can initiate neurodegenerative changes. Astrocyte-secreted APOE3, but not APOE4, suppressed the CypA-nuclear factor-κB-matrix-metalloproteinase-9 pathway in pericytes through a lipoprotein receptor. Our data suggest that CypA is a key target for treating APOE4-mediated neurovascular injury and the resulting neuronal dysfunction and degeneration.


Subject(s)
Apolipoproteins E/metabolism , Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Cyclophilin A/metabolism , Animals , Apolipoprotein E2/deficiency , Apolipoprotein E2/genetics , Apolipoprotein E2/metabolism , Apolipoprotein E3/deficiency , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/deficiency , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Cyclophilin A/antagonists & inhibitors , Cyclophilin A/deficiency , Hippocampus/metabolism , Hippocampus/pathology , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Microcirculation , NF-kappa B/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Pericytes/metabolism
5.
Proc Natl Acad Sci U S A ; 112(8): E881-90, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675483

ABSTRACT

Cerebral amyloid angiopathy (CAA) is characterized by deposition of amyloid ß peptide (Aß) within walls of cerebral arteries and is an important cause of intracerebral hemorrhage, ischemic stroke, and cognitive dysfunction in elderly patients with and without Alzheimer's Disease (AD). NADPH oxidase-derived oxidative stress plays a key role in soluble Aß-induced vessel dysfunction, but the mechanisms by which insoluble Aß in the form of CAA causes cerebrovascular (CV) dysfunction are not clear. Here, we demonstrate evidence that reactive oxygen species (ROS) and, in particular, NADPH oxidase-derived ROS are a key mediator of CAA-induced CV deficits. First, the NADPH oxidase inhibitor, apocynin, and the nonspecific ROS scavenger, tempol, are shown to reduce oxidative stress and improve CV reactivity in aged Tg2576 mice. Second, the observed improvement in CV function is attributed both to a reduction in CAA formation and a decrease in CAA-induced vasomotor impairment. Third, anti-ROS therapy attenuates CAA-related microhemorrhage. A potential mechanism by which ROS contribute to CAA pathogenesis is also identified because apocynin substantially reduces expression levels of ApoE-a factor known to promote CAA formation. In total, these data indicate that ROS are a key contributor to CAA formation, CAA-induced vessel dysfunction, and CAA-related microhemorrhage. Thus, ROS and, in particular, NADPH oxidase-derived ROS are a promising therapeutic target for patients with CAA and AD.


Subject(s)
Aging/pathology , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/physiopathology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Reactive Oxygen Species/metabolism , Vasomotor System/physiopathology , Acetophenones/pharmacology , Animals , Apolipoproteins E/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cerebral Amyloid Angiopathy/complications , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cerebral Hemorrhage/complications , Cricetinae , Cyclic N-Oxides/pharmacology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Oxidative Stress/drug effects , Spin Labels , Vasomotor System/drug effects , Vasomotor System/pathology
6.
Proc Natl Acad Sci U S A ; 110(36): 14771-6, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23959870

ABSTRACT

Whereas amyloid-ß (Aß) accumulates in the brain of normal animals dosed with low levels of copper (Cu), the mechanism is not completely known. Cu could contribute to Aß accumulation by altering its clearance and/or its production. Because Cu homeostasis is altered in transgenic mice overexpressing Aß precursor protein (APP), the objective of this study was to elucidate the mechanism of Cu-induced Aß accumulation in brains of normal mice and then to explore Cu's effects in a mouse model of Alzheimer's disease. In aging mice, accumulation of Cu in brain capillaries was associated with its reduction in low-density lipoprotein receptor-related protein 1 (LRP1), an Aß transporter, and higher brain Aß levels. These effects were reproduced by chronic dosing with low levels of Cu via drinking water without changes in Aß synthesis or degradation. In human brain endothelial cells, Cu, at its normal labile levels, caused LRP1-specific down-regulation by inducing its nitrotyrosination and subsequent proteosomal-dependent degradation due in part to Cu/cellular prion protein/LRP1 interaction. In APP(sw/0) mice, Cu not only down-regulated LRP1 in brain capillaries but also increased Aß production and neuroinflammation because Cu accumulated in brain capillaries and, unlike in control mice, in the parenchyma. Thus, we have demonstrated that Cu's effect on brain Aß homeostasis depends on whether it is accumulated in the capillaries or in the parenchyma. These findings should provide unique insights into preventative and/or therapeutic approaches to control neurotoxic Aß levels in the aging brain.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/drug effects , Copper/pharmacology , Homeostasis/drug effects , Age Factors , Amyloid beta-Peptides/pharmacokinetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/metabolism , Blotting, Western , Brain/blood supply , Brain/metabolism , Capillaries/drug effects , Capillaries/metabolism , Cell Survival/drug effects , Cells, Cultured , Copper/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Iodine Radioisotopes/pharmacokinetics , Low Density Lipoprotein Receptor-Related Protein-1 , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Time Factors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
J Neurosci ; 34(49): 16180-93, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25471560

ABSTRACT

Traumatic brain injury (TBI) is an established risk factor for the early development of dementia, including Alzheimer's disease, and the post-traumatic brain frequently exhibits neurofibrillary tangles comprised of aggregates of the protein tau. We have recently defined a brain-wide network of paravascular channels, termed the "glymphatic" pathway, along which CSF moves into and through the brain parenchyma, facilitating the clearance of interstitial solutes, including amyloid-ß, from the brain. Here we demonstrate in mice that extracellular tau is cleared from the brain along these paravascular pathways. After TBI, glymphatic pathway function was reduced by ∼60%, with this impairment persisting for at least 1 month post injury. Genetic knock-out of the gene encoding the astroglial water channel aquaporin-4, which is importantly involved in paravascular interstitial solute clearance, exacerbated glymphatic pathway dysfunction after TBI and promoted the development of neurofibrillary pathology and neurodegeneration in the post-traumatic brain. These findings suggest that chronic impairment of glymphatic pathway function after TBI may be a key factor that renders the post-traumatic brain vulnerable to tau aggregation and the onset of neurodegeneration.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Cerebrospinal Fluid/metabolism , Extracellular Fluid/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , tau Proteins/metabolism , Animals , Aquaporin 4/genetics , Brain Injuries/complications , Brain Injuries/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurofibrillary Tangles/genetics
8.
J Biol Chem ; 288(21): 15154-66, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23580652

ABSTRACT

Soluble low density lipoprotein receptor-related protein-1 (sLRP1) binds ~70% of amyloid ß-peptide (Aß) in human plasma. In Alzheimer disease (AD) and individuals with mild cognitive impairment converting to AD, plasma sLRP1 levels are reduced and sLRP1 is oxidized, which results in diminished Aß peripheral binding and higher levels of free Aß in plasma. Experimental studies have shown that free circulating Aß re-enters the brain and that sLRP1 and/or its recombinant wild type cluster IV (WT-LRPIV) prevent Aß from entering the brain. Treatment of Alzheimer APPsw(+/0) mice with WT-LRPIV has been shown to reduce brain Aß pathology. In addition to Aß, LRPIV binds multiple ligands. To enhance LRPIV binding for Aß relative to other LRP1 ligands, we generated a library of LRPIV-derived fragments and full-length LRPIV variants with glycine replacing aspartic acid residues 3394, 3556, and 3674 in the calcium binding sites. Compared with WT-LRPIV, a lead LRPIV-D3674G mutant had 1.6- and 2.7-fold higher binding affinity for Aß40 and Aß42 in vitro, respectively, and a lower binding affinity for other LRP1 ligands (e.g. apolipoprotein E2, E3, and E4 (1.3-1.8-fold), tissue plasminogen activator (2.7-fold), matrix metalloproteinase-9 (4.1-fold), and Factor Xa (3.8-fold)). LRPIV-D3674G cleared mouse endogenous brain Aß40 and Aß42 25-27% better than WT-LRPIV. A 3-month subcutaneous treatment of APPsw(+/0) mice with LRPIV-D3674G (40 µg/kg/day) reduced Aß40 and Αß42 levels in the hippocampus, cortex, and cerebrospinal fluid by 60-80% and improved cerebral blood flow responses and hippocampal function at 9 months of age. Thus, LRPIV-D3674G is an efficient new Aß clearance therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Peptide Fragments/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Amino Acid Substitution , Amyloid beta-Peptides/genetics , Animals , CHO Cells , Cerebral Cortex/pathology , Cerebrovascular Circulation/genetics , Cricetinae , Cricetulus , Hippocampus/pathology , Humans , Ligands , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Mutant Strains , Mutation, Missense , Peptide Fragments/genetics , Protein Binding/genetics , Receptors, LDL/genetics , Tumor Suppressor Proteins/genetics
9.
Blood ; 115(23): 4963-72, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20348395

ABSTRACT

The anticoagulant factor protein S (PS) has direct cellular activities. Lack of PS in mice causes lethal coagulopathy, ischemic/thrombotic injuries, vascular dysgenesis, and blood-brain barrier (BBB) disruption with intracerebral hemorrhages. Thus, we hypothesized that PS maintains and/or enhances the BBB integrity. Using a BBB model with human brain endothelial cells, we show PS inhibits time- and dose-dependently (half maximal effective concentration [EC(50)] = 27 +/- 3 nM) oxygen/glucose deprivation-induced BBB breakdown, as demonstrated by measurements of the transmonolayer electrical resistance, permeability of endothelial monolayers to dextran (40 kDa), and rearrangement of F-actin toward the cortical cytoskeletal ring. Using Tyro-3, Axl, and Mer (TAM) receptor, tyrosine kinase silencing through RNA interference, specific N-terminus-blocking antibodies, Tyro3 phosphorylation, and Tyro3-, Axl- and Mer-deficient mouse brain endothelial cells, we show that Tyro3 mediates PS vasculoprotection. After Tyro3 ligation, PS activated sphingosine 1-phosphate receptor (S1P(1)), resulting in Rac1-dependent BBB protection. Using 2-photon in vivo imaging, we show that PS blocks postischemic BBB disruption in Tyro3(+/+), Axl(-/-), and Mer(-/-) mice, but not in Tyro3(-/-) mice or Tyro3(+/+) mice receiving low-dose W146, a S1P(1)-specific antagonist. Our findings indicate that PS protects the BBB integrity via Tyro3 and S1P(1), suggesting potentially novel treatments for neurovascular dysfunction resulting from hypoxic/ischemic BBB damage.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Protein S/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Lysosphingolipid/metabolism , Animals , Brain Ischemia/genetics , Humans , Mice , Mice, Mutant Strains , Models, Biological , Neuropeptides/genetics , Neuropeptides/metabolism , Phosphorylation/genetics , Protein S/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Lysosphingolipid/genetics , Sphingosine-1-Phosphate Receptors , Young Adult , c-Mer Tyrosine Kinase , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Axl Receptor Tyrosine Kinase
10.
Acta Neuropathol Commun ; 9(1): 193, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911575

ABSTRACT

Previous reports indicate a potential role for signal transducer and activator of transcription 3 (STAT3) in amyloid-ß (Aß) processing and neuritic plaque pathogenesis. In the present study, the impact of STAT3 inhibition on cognition, cerebrovascular function, amyloid pathology, oxidative stress, and neuroinflammation was studied using in vitro and in vivo models of Alzheimer's disease (AD)-related pathology. For in vitro experiments, human brain vascular smooth muscle cells (HBVSMC) and human brain microvascular endothelial cells (HBMEC) were used, and these cultured cells were exposed to Aß peptides followed by measurement of activated forms of STAT3 expression and reactive oxygen species (ROS) generation. Further, 6 months old 5XFAD/APOE4 (5XE4) mice and age-matched negative littermates were used for in vivo experiments. These mice were treated with STAT3 specific inhibitor, LLL-12 for 2 months followed by neurobehavioral and histopathological assessment. In vitro experiments showed exposure of cerebrovascular cells to Aß peptides upregulated activated forms of STAT3 and produced STAT3-mediated vascular oxidative stress. 5XE4 mice treated with the STAT3-specific inhibitor (LLL-12) improved cognitive functions and functional connectivity and augmented cerebral blood flow. These functional improvements were associated with a reduction in neuritic plaques, cerebral amyloid angiopathy (CAA), oxidative stress, and neuroinflammation. Reduction in amyloid precursor protein (APP) processing and attenuation of oxidative modification of lipoprotein receptor related protein-1 (LRP-1) were identified as potential underlying mechanisms. These results demonstrate the broad impact of STAT3 on cognitive functions, parenchymal and vascular amyloid pathology and highlight the therapeutic potential of STAT3 specific inhibition for treatment of AD and CAA.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/pharmacology , Anthraquinones/pharmacology , Cerebrovascular Disorders/drug therapy , Cognitive Dysfunction/drug therapy , Nerve Net/diagnostic imaging , Plaque, Amyloid/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology , Animals , Autopsy , Brain , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Transgenic , Microvessels/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , STAT3 Transcription Factor/drug effects
11.
Stroke ; 40(5): 1864-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19057019

ABSTRACT

BACKGROUND AND PURPOSE: Activated protein C (APC), a protease with anticoagulant and cytoprotective activities, protects neurons and endothelium from ischemic injury. Drotrecogin-alfa activated, a hyperanticoagulant form of human recombinant APC, is currently being studied in patients with ischemic stroke. How changes in APC anticoagulant activity influence APC's neuroprotection and risk for bleeding is not clear. METHODS: We used neuronal and brain endothelial cell injury models and middle cerebral artery occlusion in mice to compare efficacy and safety of drotrecogin-alfa activated and human 3K3A-APC, an APC nonanticoagulant mutant. RESULTS: Drotrecogin-alfa activated and 3K3A-APC exhibited 148% and 10% of plasma-derived APC's anticoagulant activity and differ in the carbohydrate content. 3K3A-APC protected mouse neurons from N-methyl-d-aspartate-induced apoptosis and human brain endothelial cell from oxygen-glucose deprivation with 1.8- and 3.1-fold greater efficacy than drotrecogin-alfa activated. Given 5 minutes before transient middle cerebral artery occlusion, 3K3A-APC and drotrecogin-alfa activated (0.5 and 2 mg/kg intravenously) reduced comparably and dose-dependently the infarction lesion up to 85%. 3K3A-APC, but not drotrecogin-alfa activated, improved neurological score dose-dependently (P<0.05). 3K3A-APC did not cause bleeding. In contrast, drotrecogin-alfa activated dose-dependently increased hemoglobin content in postischemic brain. After permanent middle cerebral artery occlusion, 3K3A-APC multidose therapy (1 mg/kg intravenously at 12 hours and 1, 3, 5, and 7 days) improved functional recovery and reduced infarction by 60% with no risk for bleeding, whereas drotrecogin-alfa activated increased hemoglobin deposition in the postischemic brain and showed relatively modest neuroprotection. CONCLUSIONS: Nonanticoagulant 3K3A-APC exhibits greater neuroprotective efficacy with no risk for bleeding compared with drotrecogin-alfa activated, a hyperanticoagulant form of APC.


Subject(s)
Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/epidemiology , Fibrinolytic Agents/pharmacology , Neuroprotective Agents , Protein C/genetics , Protein C/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Excitatory Amino Acid Agonists/pharmacology , Female , Humans , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Inbred C57BL , Mutation/genetics , Mutation/physiology , N-Acetylneuraminic Acid/metabolism , N-Methylaspartate/pharmacology , Neurons/drug effects , Partial Thromboplastin Time , Polysaccharides/metabolism , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Risk
12.
J Neurochem ; 109(1): 116-24, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19166505

ABSTRACT

Activated protein C (APC) is a protease with anticoagulant and cytoprotective activities. APC is neuroprotective in rodent models of stroke. But, an APC variant with reduced anticoagulant activity, 3K3A-APC, compared to wild-type APC shows greater neuroprotection with no risk for bleeding in stroke models. To determine whether 3K3A-APC exhibits species-dependent neuroprotection similar to that as seen with wild-type APC, we studied murine and human recombinant 3K3A-APC mutants which show approximately 80% reduced anticoagulant activity. Murine 3K3A-APC (0.2 mg/kg i.v.) administered at 4 h after embolic stroke improved substantially functional outcome and reduced by 80% the infract volume 7 days after stroke. Human 3K3A-APC was neuroprotective after embolic stroke in mice, but at significantly higher concentrations (i.e. 2 mg/kg i.v.). Species-dependent neuroprotection, i.e. murine > human 3K3A-APC, was confirmed in a mouse model of permanent middle cerebral artery occlusion. Human 3K3A-APC had by fivefold greater cytoprotective activity than murine 3K3A-APC in oxygen-glucose deprivation model in human brain endothelial cells, whereas murine 3K3A-APC was by 2.5-fold more potent than human 3K3A-APC in a mouse model of NMDA-induced neuronal apoptosis. Thus, 3K3A-APC exhibits species-dependent neuroprotection which should be taken into account when designing human trials for ischemic stroke with APC mutants.


Subject(s)
Anticoagulants/metabolism , Mutation , Neuroprotective Agents/metabolism , Protein C/genetics , Protein C/metabolism , Animals , Anticoagulants/therapeutic use , CHO Cells , Cell Line , Cells, Cultured , Cricetinae , Cricetulus , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Pregnancy , Protein C/therapeutic use , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Species Specificity , Stroke/drug therapy , Stroke/metabolism , Stroke/prevention & control
13.
Eur J Neurosci ; 29(6): 1119-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19302148

ABSTRACT

The anticoagulant activated protein C (APC) protects neurons and endothelium via protease activated receptor (PAR)1, PAR3 and endothelial protein C receptor. APC is neuroprotective in stroke models. Bleeding complications may limit the pharmacologic utility of APC. Here, we compared the 3K3A-APC mutant with 80% reduced anticoagulant activity and wild-type (wt)-APC. Murine 3K3A-APC compared with wt-APC protected mouse cortical neurons from N-methyl-D-aspartate-induced apoptosis with twofold greater efficacy and more potently reduced N-methyl-D-aspartate excitotoxic lesions in vivo. Human 3K3A-APC protected human brain endothelial cells (BECs) from oxygen/glucose deprivation with 1.7-fold greater efficacy than wt-APC. 3K3A-APC neuronal protection required PAR1 and PAR3, as shown by using PAR-specific blocking antibodies and PAR1- and PAR3-deficient cells and mice. BEC protection required endothelial protein C receptor and PAR1. In neurons and BECs, 3K3A-APC blocked caspase-9 and -3 activation and induction of p53, and decreased the Bax/Bcl-2 pro-apoptotic ratio. After distal middle cerebral artery occlusion (dMCAO) in mice, murine 3K3A-APC compared with vehicle given 4:00 h after dMCAO improved the functional outcome and reduced the infarction volume by 50% within 3 days. 3K3A-APC compared with wt-APC multi-dosing therapy at 12:00 h, 1, 3, 5 and 7 days after dMCAO significantly improved functional recovery and reduced the infarction volume by 75% and 38%, respectively, within 7 days. The wt-APC, but not 3K3A-APC, significantly increased the risk of intracerebral bleeding as indicated by a 50% increase in hemoglobin levels in the ischemic hemisphere. Thus, 3K3A-APC offers a new approach for safer and more efficacious treatments of neurodegenerative disorders and stroke with APC.


Subject(s)
Anticoagulants/therapeutic use , Mutation/physiology , Neuroprotective Agents/therapeutic use , Protein C/genetics , Protein C/therapeutic use , Analysis of Variance , Animals , Antibodies/pharmacology , Anticoagulants/metabolism , Apoptosis/drug effects , Brain/cytology , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Embryo, Mammalian , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/toxicity , Female , Glucose/deficiency , Hemoglobins/metabolism , Humans , Hypoxia/drug therapy , In Situ Nick-End Labeling/methods , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate/toxicity , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/metabolism , Pregnancy , Protein C/chemistry , Protein C/immunology , Receptors, Proteinase-Activated/genetics , Time Factors , Tumor Suppressor Protein p53/metabolism
14.
Haematologica ; 94(12): 1721-31, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19815836

ABSTRACT

BACKGROUND: The protein C pathway down-regulates thrombin generation and promotes cytoprotection during inflammation and stress. In preclinical studies using models of murine injury (e.g., sepsis and ischemic stroke), murine protein S may be required because of restrictive species specificity. DESIGN AND METHODS: We prepared and characterized recombinant murine protein S using novel coagulation assays, immunoassays, and cell proliferation assays. RESULTS: Purified murine protein S had good anticoagulant co-factor activity for murine activated protein C, but not for human activated protein C, in mouse or rat plasma. In human plasma, murine protein S was a poor co-factor for murine activated protein C and had no anticoagulant effect with human activated protein C, suggesting protein S species specificity for factor V in addition to activated protein C. We estimated that mouse plasma contains 22+/-1 microg/mL protein S and developed assays to measure activated protein C co-factor activity of the protein S in murine plasma. Activated protein C-independent anticoagulant activity of murine protein S was demonstrable and quantifiable in mouse plasma, and this activity was enhanced by exogenous murine protein S. Murine protein S promoted the proliferation of mouse and human smooth muscle cells. The potency of murine protein S was higher for mouse cells than for human cells and similarly, human protein S was more potent for human cells than for mouse cells. CONCLUSIONS: The spectrum of bioactivities of recombinant murine protein S with mouse plasma and smooth muscle cells is similar to that of human protein S. However, in vitro and in vivo studies of the protein C pathway in murine disease models are more appropriately performed using murine protein S. This study extends previous observations regarding the remarkable species specificity of protein S to the mouse.


Subject(s)
Anticoagulants/pharmacology , Mitogens/pharmacology , Protein S/metabolism , Protein S/pharmacology , Animals , Blood Coagulation/drug effects , Cattle , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Half-Life , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Partial Thromboplastin Time , Protein C/metabolism , Protein C/pharmacology , Protein S/genetics , Rats , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Species Specificity
16.
Ann Clin Transl Neurol ; 4(12): 865-876, 2017 12.
Article in English | MEDLINE | ID: mdl-29296615

ABSTRACT

Objective: Delayed cerebral ischemia (DCI) is an independent risk factor for poor outcome after aneurysmal subarachnoid hemorrhage (SAH) and is multifactorial in etiology. While prior studies have suggested a role for matrix metalloproteinase-9 (MMP-9) in early brain injury after SAH, its contribution to the pathophysiology of DCI is unclear. Methods: In the first experiment, wild-type (WT) and MMP-9-/- mice were subjected to sham or endovascular perforation SAH surgery. In separate experiments, WT and MMP-9-/-mice were administered vehicle or minocycline either pre- or post-SAH. All mice underwent assessment of multiple components of DCI including vasospasm, neurobehavioral function, and microvessel thrombosis. In another experiment, rabbits were subjected to sham or cisterna magna injection SAH surgery, and administered vehicle or minocycline followed by vasospasm assessment. Results: MMP-9 expression and activity was increased after SAH. Genetic (MMP-9-/- mice) and pharmacological (pre-SAH minocycline administration) inhibition of MMP-9 resulted in decreased vasospasm and neurobehavioral deficits. A therapeutically feasible strategy of post-SAH administration of minocycline resulted in attenuation of multiple components of DCI. Minocycline administration to MMP-9-/- mice did not yield additional protection. Consistent with experiments in mice, both pre- and post-SAH administration of minocycline attenuated SAH-induced vasospasm in rabbits. Interpretation: MMP-9 is a key player in the pathogenesis of DCI. The consistent attenuation of multiple components of DCI with both pre- and post-SAH administration of minocycline across different species and experimental models of SAH, combined with the excellent safety profile of minocycline in humans suggest that a clinical trial in SAH patients is warranted.

17.
Sci Rep ; 7(1): 9126, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28831103

ABSTRACT

Various techniques have been developed to study changes in the cerebral vasculature in numerous neuropathological processes including subarachnoid hemorrhage (SAH). One of the most widely employed techniques uses India ink-gelatin casting, which presents numerous challenges due to its high viscosity, rapid solidification, and its impact on immunohistochemical analysis. To overcome these limitations, we developed a novel technique for assessing cerebral vasospasm using cerebrovascular perfusion with ROX, SE (5-Carboxy-X-Rhodamine, Succinimidyl Ester), a fluorescent labeling dye. We found that ROX SE perfusion achieves excellent delineation of the cerebral vasculature, was qualitatively and quantitatively superior to India ink-gelatin casting for the assessment of cerebral vasospasm, permits outstanding immunohistochemical examination of non-vasospasm components of secondary brain injury, and is a more efficient and cost-effective experimental technique. ROX SE perfusion is therefore a novel and highly useful technique for studying cerebrovascular pathology following experimental SAH.


Subject(s)
Optical Imaging , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/physiopathology , Vasospasm, Intracranial/diagnosis , Vasospasm, Intracranial/etiology , Animals , Biomarkers , Blood-Brain Barrier/metabolism , Costs and Cost Analysis , Immunohistochemistry , Male , Mice , Observer Variation , Optical Imaging/economics , Optical Imaging/methods , Staining and Labeling
18.
Mol Neurodegener ; 11: 9, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26801396

ABSTRACT

BACKGROUND: Substantial evidence suggests that amyloid-ß (Aß) species induce oxidative stress and cerebrovascular (CV) dysfunction in Alzheimer's disease (AD), potentially contributing to the progressive dementia of this disease. The upstream molecular pathways governing this process, however, are poorly understood. In this report, we examine the role of heparan sulfate proteoglycans (HSPG) in Aß-induced vascular smooth muscle cell (VSMC) dysfunction in vitro. RESULTS: Our results demonstrate that pharmacological depletion of HSPG (by enzymatic degradation with active, but not heat-inactivated, heparinase) in primary human cerebral and transformed rat VSMC mitigates Aß(1-40⁻) and Aß(1-42⁻)induced oxidative stress. This inhibitory effect is specific for HSPG depletion and does not occur with pharmacological depletion of other glycosaminoglycan (GAG) family members. We also found that Aß(1-40) (but not Aß(1-42)) causes a hypercontractile phenotype in transformed rat cerebral VSMC that likely results from a HSPG-mediated augmentation in intracellular Ca(2+) activity, as both Aß(1-40⁻)induced VSMC hypercontractility and increased Ca(2+) influx are inhibited by pharmacological HSPG depletion. Moreover, chelation of extracellular Ca(2+) with ethylene glycol tetraacetic acid (EGTA) does not prevent the production of Aß(1-40⁻) or Aß(1-42⁻)mediated reactive oxygen species (ROS), suggesting that Aß-induced ROS and VSMC hypercontractility occur through different molecular pathways. CONCLUSIONS: Taken together, our data indicate that HSPG are critical mediators of Aß-induced oxidative stress and Aß(1-40⁻)induced VSMC dysfunction.


Subject(s)
Amyloid beta-Peptides/metabolism , Heparan Sulfate Proteoglycans/metabolism , Muscle, Smooth, Vascular/metabolism , Oxidative Stress/physiology , Peptide Fragments/metabolism , Reactive Oxygen Species/metabolism , Alzheimer Disease/metabolism , Cell Line , Cells, Cultured , Humans
19.
Jpn J Infect Dis ; 58(1): 31-3, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15728988

ABSTRACT

Yersinia enterocolitica biotype 1A isolates are increasingly being associated with diarrhea. However, the mechanism of their pathogenicity is not well understood. In the present study interaction of Y. enterocolitica isolates with CHO cells, HEp-2 cells and J774 mouse macrophages was studied. Y. enterocolitica biotype 1A strains of clinical origin invaded CHO and HEp-2 cells to a significantly higher degree than non-clinical isolates. However, among non-clinical isolates, Y. enterocolitica strains of swine origin showed significantly more invasion in CHO and HEp-2 cells than water isolates. Y. enterocolitica isolates from clinical samples exhibited a greater level of survival in macrophages than isolates from non-clinical sources. It may be construed that Y. enterocolitica biotype 1A isolates of clinical and swine origin have higher virulence potential than those from other sources.


Subject(s)
Yersinia enterocolitica/classification , Yersinia enterocolitica/physiology , Animals , Cell Line , Cells/microbiology , Cricetinae , Humans , Mice , Yersinia enterocolitica/pathogenicity
20.
J Med Microbiol ; 53(Pt 11): 1065-1068, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15496381

ABSTRACT

Two hundred and fifty nine isolates of Yersinia enterocolitica and related species were examined for the production of heat-stable enterotoxin (Yersinia stable toxin; YST) as well as for the prevalence of enterotoxin genes, viz. ystA, ystB and ystC. Under the conventional conditions used for the production of Y. enterocolitica enterotoxin, i.e. in tryptic soy broth (TSB) supplemented with yeast extract at 28 degrees C for 48 h, 77.7 % of clinical isolates and 62.3 % of swine isolates showed enterotoxigenicity in infant mice. All isolates that produced enterotoxin at 28 degrees C also showed enterotoxic activity at 37 degrees C after 48 h incubation under an alkaline pH of 7.5, the pH present in the ileum. All Yersinia intermedia and Yersinia frederiksenii isolates were negative for enterotoxin production. All clinical isolates and 96.3 % of Y. enterocolitica isolates from swine hybridized with a probe for ystB, which indicated that the ystB gene was most prevalent in Y. enterocolitica biotype 1A strains. None of the Y. enterocolitica isolates showed hybridization with oligonucleotide probes for ystA or ystC. The study indicated that YST-b was the major contributor to diarrhoea produced by biotype 1A strains of Y. enterocolitica.


Subject(s)
Bacterial Toxins/biosynthesis , Bacterial Toxins/genetics , Enterotoxins/biosynthesis , Enterotoxins/genetics , Genes, Bacterial , Yersinia enterocolitica/genetics , Yersinia enterocolitica/metabolism , Animals , Culture Media/chemistry , Feces/microbiology , Humans , Hydrogen-Ion Concentration , Mice , Molecular Epidemiology , Nucleic Acid Hybridization , Pharynx/microbiology , Swine/microbiology , Temperature , Time Factors , Water Microbiology , Yersinia Infections/microbiology , Yersinia enterocolitica/classification , Yersinia enterocolitica/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL