Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Cell Immunol ; 371: 104451, 2022 01.
Article in English | MEDLINE | ID: mdl-34781155

ABSTRACT

The COVID-19 pandemic has once again brought to the forefront the existence of a tight link between the coagulation/fibrinolytic system and the immunologic processes. Tissue-type plasminogen activator (tPA) is a serine protease with a key role in fibrinolysis by converting plasminogen into plasmin that can finally degrade fibrin clots. tPA is released in the blood by endothelial cells and hepatocytes but is also produced by various types of immune cells including T cells and monocytes. Beyond its role on hemostasis, tPA is also a potent modulator of inflammation and is involved in the regulation of several inflammatory diseases. Here, after a brief description of tPA structure, we review its new functions in adaptive immunity focusing on T cells and antigen presenting cells. We intend to synthesize the recent knowledge on proteolysis- and receptor-mediated effects of tPA on immune response in physiological and pathological context.


Subject(s)
Blood Coagulation/immunology , COVID-19/immunology , Fibrinolysis/immunology , Immunity/immunology , SARS-CoV-2/immunology , Tissue Plasminogen Activator/immunology , Antigen-Presenting Cells/immunology , COVID-19/epidemiology , COVID-19/virology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Humans , Models, Immunological , Pandemics , SARS-CoV-2/physiology , T-Lymphocytes/immunology , Tissue Plasminogen Activator/metabolism
2.
Cell Signal ; 75: 109761, 2020 11.
Article in English | MEDLINE | ID: mdl-32861744

ABSTRACT

A fine-tuned activation and deactivation of proteases and their inhibitors are involved in the execution of the inflammatory response. The zymogen/proenzyme plasminogen is converted to the serine protease plasmin, a key fibrinolytic factor by plasminogen activators including tissue-type plasminogen activator (tPA). Plasmin is part of an intricate protease network controlling proteins of initial hemostasis/coagulation, fibrinolytic and complement system. Activation of these protease cascades is required to mount a proper inflammatory response. Although best known for its ability to dissolve clots and cleave fibrin, recent studies point to the importance of fibrin-independent functions of plasmin during acute inflammation and inflammation resolution. In this review, we provide an up-to-date overview of the current knowledge of the enzymatic and cytokine-like effects of tPA and describe the role of tPA and plasminogen receptors in the regulation of the inflammatory response with emphasis on the cytokine storm syndrome such as observed during coronavirus disease 2019 or macrophage activation syndrome. We discuss tPA as a modulator of Toll like receptor signaling, plasmin as an activator of NFkB signaling, and summarize recent studies on the role of plasminogen receptors as controllers of the macrophage conversion into the M2 type and as mediators of efferocytosis during inflammation resolution.


Subject(s)
Inflammation/immunology , Plasminogen/immunology , Animals , Blood Coagulation , COVID-19 , Complement Activation , Coronavirus Infections/blood , Coronavirus Infections/complications , Coronavirus Infections/immunology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/complications , Cytokine Release Syndrome/immunology , Cytokines/immunology , Humans , Immune System/immunology , Inflammation/blood , Inflammation/complications , Low Density Lipoprotein Receptor-Related Protein-1/immunology , NF-kappa B/immunology , Pandemics , Pneumonia, Viral/blood , Pneumonia, Viral/complications , Pneumonia, Viral/immunology , Tissue Plasminogen Activator/immunology
3.
Cell Immunol ; 348: 104040, 2020 02.
Article in English | MEDLINE | ID: mdl-31955841

ABSTRACT

Tissue plasminogen activator (tPA), a component of the plasminogen activator (PA) system, is elevated in inflammatory neurological disorders. In the present study, we explored the immunomodulatory activity of tPA in experimental autoimmune encephalomyelitis (EAE). The EAE was treated with two catalytic inactive tPA variant proteins: S(481)A and S(481)A + KHRR(296-299)AAAA. EAE-induced tPA-/- mice presented with markedly more severe disease than wt EAE mice. Further, treatment with tPA variants, demonstrated a significant suppression of disease severity in tPA-/- and wt mice. Immunological evaluation showed that specific T-cell reactivity was markedly reduced in the tPA-/- animals, as indicated by decreased T-cell reactivity and reduction in T-regulatory cells. The current findings indicate that tPA plays a role in the pathogenesis of EAE. Moreover, successful amelioration of EAE was achieved by administration of tPA variant proteins. This might mean that these proteins have potential for the immunomodulation of neuroinflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Tissue Plasminogen Activator/immunology , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Front Immunol ; 10: 897, 2019.
Article in English | MEDLINE | ID: mdl-31110506

ABSTRACT

East Coast Fever (ECF), caused by the tick-borne apicomplexan parasite Theileria parva, is a leading cause of morbidity and mortality in cattle of sub-Saharan Africa. The infection and treatment method (ITM) is currently the only vaccine available to control T. parva. Although ITM elicits levels of protection, its widespread adoption is limited by costs, laborious production process, and antibiotic co-treatment requirement, necessitating the development of a more sustainable vaccine. To this end, efforts have been concentrated in the identification of new T. parva vaccine antigens and in the development of suitable platforms for antigen expression. In this study, we investigated the molecular and antigenic properties of T. parva antigen Tp9 expressed by mammalian cells. Data indicate that Tp9 contains a signal peptide that is weakly functional in mammalian cells. Thus, Tp9 secretion from mammalian cells increased 10-fold after the native signal peptide was replaced with the human tissue plasminogen activator signal peptide (tPA). Sera from all T. parva-immune cattle recognized this recombinant, secreted Tp9. Additionally, PBMC from ITM-immunized cattle produced significant (p < 0.05) amounts of IFNγ following ex vivo exposure to Tp9, but this response varied between cattle of different MHC class I and class II genotypes. In addition, depletion experiments demonstrated that IFNγ to Tp9 was primarily produced by CD4+ T cells. Molecular analysis demonstrated that Tp9 presents a signal peptide that is weakly functional in mammalian cells, suggesting that it remains within lymphocytes during infection. Tp9 secretion from mammalian cells was substantially increased when the tPA secretion signal sequence was substituted for the native secretion signal sequence. Using full-length, recombinant Tp9 secreted from mammalian cells, we demonstrated that T. parva-immune cattle develop both humoral and cellular immune responses to this antigen. Collectively, these results provide rationale for further evaluation of Tp9 as a component of a T. parva subunit vaccine.


Subject(s)
Antigens, Protozoan/immunology , Mammals/immunology , Theileria parva/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cattle , Cell Line , Dogs , HEK293 Cells , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Madin Darby Canine Kidney Cells , Major Histocompatibility Complex/immunology , Protozoan Vaccines/immunology , Theileriasis/immunology , Tissue Plasminogen Activator/immunology , Vaccines, Subunit/immunology
5.
Front Immunol ; 10: 591, 2019.
Article in English | MEDLINE | ID: mdl-30972077

ABSTRACT

Introduction: Acute ischemic stroke (AIS) is a potent trigger of immunosuppression, resulting in increased infection risk. While thrombolytic therapy with tissue-type plasminogen activator (t-PA) is still the only pharmacological treatment for AIS, plasmin, the effector protease, has been reported to suppress dendritic cells (DCs), known for their potent antigen-presenting capacity. Accordingly, in the major group of thrombolyzed AIS patients who fail to reanalyze (>60%), t-PA might trigger unintended and potentially harmful immunosuppressive consequences instead of beneficial reperfusion. To test this hypothesis, we performed an exploratory study to investigate the immunomodulatory properties of t-PA treatment in a mouse model of ischemic stroke. Methods: C57Bl/6J wild-type mice and plasminogen-deficient (plg-/-) mice were subjected to middle cerebral artery occlusion (MCAo) for 60 min followed by mouse t-PA treatment (0.9 mg/kg) at reperfusion. Behavioral testing was performed 23 h after occlusion, pursued by determination of blood counts and plasma cytokines at 24 h. Spleens and cervical lymph nodes (cLN) were also harvested and characterized by flow cytometry. Results: MCAo resulted in profound attenuation of immune activation, as anticipated. t-PA treatment not only worsened neurological deficit, but further reduced lymphocyte and monocyte counts in blood, enhanced plasma levels of both IL-10 and TNFα and decreased various conventional DC subsets in the spleen and cLN, consistent with enhanced immunosuppression and systemic inflammation after stroke. Many of these effects were abolished in plg-/- mice, suggesting plasmin as a key mediator of t-PA-induced immunosuppression. Conclusion: t-PA, via plasmin generation, may weaken the immune response post-stroke, potentially enhancing infection risk and impairing neurological recovery. Due to the large number of comparisons performed in this study, additional pre-clinical work is required to confirm these significant possibilities. Future studies will also need to ascertain the functional implications of t-PA-mediated immunosuppression for thrombolyzed AIS patients, particularly for those with failed recanalization.


Subject(s)
Fibrinolysin/immunology , Stroke/pathology , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/immunology , Tissue Plasminogen Activator/therapeutic use , Animals , Cytokines/blood , Disease Models, Animal , Immunomodulation/immunology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Cerebral Artery/pathology , Plasminogen/genetics
6.
Front Immunol ; 9: 2183, 2018.
Article in English | MEDLINE | ID: mdl-30333824

ABSTRACT

Multiple pathways have been proposed to generate bradykinin (BK)-related peptides from blood. We applied various forms of activation to fresh blood obtained from 10 healthy subjects or 10 patients with hereditary angioedema (HAE-1 or -2 only) to investigate kinin formation. An enzyme immunoassay for BK was applied to extracts of citrated blood incubated at 37°C under gentle agitation for 0-2 h in the presence of activators and/or inhibitory agents. Biologically active kinins in extracts were corroborated by c-Fos accumulation in HEK 293a cells that express either recombinant human B2 or B1 receptors (B2R, B1R). Biological evidence of HAE diagnostic and blood cell activation was also obtained. The angiotensin converting enzyme inhibitor enalaprilat, without any effect per se, increased immunoreactive BK (iBK) concentration under active stimulation of blood. Tissue kallikrein (KLK-1) and Kontact-APTT, a particulate material that activates the contact system, rapidly (5 min) and intensely (>100 ng/mL) induced similar iBK generation in the blood of control or HAE subjects. Tissue plasminogen activator (tPA) slowly (≥1 h) induced iBK generation in control blood, but more rapidly and intensely so in that of HAE patients. Effects of biotechnological inhibitors indicate that tPA recruits factor XIIa (FXIIa) and plasma kallikrein to generate iBK. KLK-1, independent of the contact system, is the only stimulus leading to an inconsistent B1R stimulation. Stimulating neutrophils or platelets did not generate iBK. In the HAE patients observed during remission, iBK formation capability coupled to B2R stimulation appears largely intact. However, a selective hypersensitivity to tPA in the blood of HAE patients suggests a role of plasmin-activated FXIIa in the development of attacks. Proposed pathways of kinin formation dependent on blood cell activation were not corroborated.


Subject(s)
Angioedemas, Hereditary , Bradykinin , Factor XIIa , Tissue Kallikreins , Tissue Plasminogen Activator , Adolescent , Adult , Aged , Angioedemas, Hereditary/blood , Angioedemas, Hereditary/immunology , Angioedemas, Hereditary/pathology , Blood Platelets/immunology , Blood Platelets/metabolism , Blood Platelets/pathology , Bradykinin/blood , Bradykinin/immunology , Factor XIIa/immunology , Factor XIIa/metabolism , Female , HEK293 Cells , Humans , Male , Middle Aged , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Tissue Kallikreins/blood , Tissue Kallikreins/immunology , Tissue Plasminogen Activator/blood , Tissue Plasminogen Activator/immunology
7.
Vet Microbiol ; 225: 40-47, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30322531

ABSTRACT

Duck circovirus (DuCV) is an immunosuppressive pathogen that causes a huge economic loss in the avian industry. Efficient vaccination has become a necessary strategy for preventing DuCV infection in the breeding industry. Three DNA vaccines encoding the Capsid (Cap) protein of DuCV were developed in this study, which were based on the eukaryotic vector pcDNA3.1 containing (i) the full length of Cap gene, pcDNA3.1-Cap, (ii) the Cap gene with a deletion of its nuclear localization signal (NLS) peptide encoding sequence, pcDNA3.1-CapΔNLS, and (iii) the Cap gene without NLS but harboring a fragment encoding the secretory signal peptide of tissue plasminogen activator (tPA), pcDNA3.1-tPA-CapΔNLS. Production of Cap protein-derived antigens from these three DNA vaccines was confirmed in vitro. The deletion of the NLS coding sequence of the Cap gene changed the subcellular location of the Capsid protein from the nucleus to the cytoplasm. Secretion of the Cap protein was observed in pcDNA3.1-tPA-CapΔNLS-transfected cells. The immunogenicity of these three DNA vaccines was assessed in vivo by measuring Cap-specific antibody and related cytokine levels. The results demonstrated that all these vaccines could induce a significant, specific immune response to protect ducks from DuCV challenge. Notably, higher titers of Cap-specific antibody were produced in ducks vaccinated with pcDNA3.1-tPA-CapΔNLS, which provided the highest protective efficacy at a rate of 90% in the challenge experiment. Taken together, DNA vaccines expressing the DuCV Cap protein show promising immunogenicity, which can be enhanced by replacing the NLS of the Cap protein with a secretory signal peptide of tPA.


Subject(s)
Capsid Proteins/immunology , Circoviridae Infections/veterinary , Circovirus/genetics , Ducks/immunology , Poultry Diseases/prevention & control , Vaccines, DNA/immunology , Animals , Capsid Proteins/administration & dosage , Capsid Proteins/genetics , Capsid Proteins/metabolism , Circoviridae Infections/immunology , Circoviridae Infections/prevention & control , Circoviridae Infections/virology , Circovirus/chemistry , Enzyme-Linked Immunosorbent Assay , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Immunogenicity, Vaccine , Nuclear Localization Signals/deficiency , Nuclear Localization Signals/genetics , Poultry Diseases/immunology , Poultry Diseases/virology , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/immunology , Vaccines, DNA/administration & dosage
8.
Appl Microbiol Biotechnol ; 102(17): 7499-7507, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29961099

ABSTRACT

Devastating outbreaks of porcine epidemic diarrhea (PED) started in China in late 2010 and rapidly spread to North America and Asia causing severe diarrhea and high mortality in neonatal piglets, indicating that a new generation of vaccine against porcine epidemic diarrhea virus (PEDV) is urgently needed. In the present study, to mimic the native spike (S) glycoprotein, a stable cell line producing the trimeric ectodomain of S glycoprotein of the PEDV Pintung-52 (PEDV-PT) strain was successfully established by incorporating T4 bacteriophage foldon sequence of fibritin trimerization domains at the C-terminal end and replacing the signal peptide of S protein with the tissue plasminogen activator signal peptide sequence at the N terminal end. The trimeric structure, bio-reactivity to PEDV-specific antibodies, and the N-glycosylation level of the recombinant S protein were characterized. To induce systemic and mucosal immunity, conventional 5-week-old piglets were immunized with the trimeric S glycoprotein combined with the B subunit of Escherichia coli heat-labile enterotoxin (LTB) by the intramuscular (IM) route. As compared with the control group, all piglets in the S protein-LTB immunized (IM PEDV S-LTB) group generated systemic PEDV S-specific IgG and neutralizing antibody in blood but a low level of fecal PEDV-specific IgA and limited protection against challenge of PEDV-PT strain. Our results suggest that the recombinant PEDV trimeric S glycoprotein could be a potential subunit vaccine candidate against PEDV, but IM immunization with LTB as the adjuvant provided insufficient protection. The development of a vaccine regimen for inducing mucosal immunity is an important task for generating a successful subunit vaccine against PEDVs.


Subject(s)
Porcine epidemic diarrhea virus/immunology , Spike Glycoprotein, Coronavirus/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/blood , Cell Line , Enterotoxins/immunology , Hot Temperature , Swine , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Vaccines/administration & dosage
9.
Thromb Haemost ; 118(1): 42-53, 2018 01.
Article in English | MEDLINE | ID: mdl-29304524

ABSTRACT

Acute ischaemic stroke, myocardial infarction and pulmonary embolism are the main causes of mortality and morbidity worldwide. Thrombolysis by intravenous injection of recombinant tissue plasminogen activator (rtPA) remains the most common non-interventional treatment to recanalize occluded vessels. However, this procedure is limited by significant drawbacks, including high doses and bleeding complications. Recent studies showed that fucoidan targets the intraluminal thrombus in vivo. We have developed a chimaera covalently linking fucoidan, able to target platelets within the thrombus, to dilysine, able to non-covalently bind rtPA. We hypothesize that this construct should vectorize rtPA to the thrombus, thus increasing its fibrinolytic efficacy and avoiding its deleterious effects. In vitro, rtPA mixed to dilysine fucoidan (DLF) shows a greater fibrinolytic effect than rtPA alone, both on platelet-rich thrombus and in whole blood. In vivo, occluded mesenteric vessels, carotid artery and vena cava were more efficiently recanalized by DLF complexed to rtPA than by rtPA alone. This study thus provides evidence that DLF may be a promising therapeutic tool to fight against acute thrombosis by enhancing rtPA fibrinolytic efficiency.


Subject(s)
Lysine/chemistry , Polysaccharides/chemistry , Thrombosis/immunology , Tissue Plasminogen Activator/immunology , Amines/chemistry , Animals , Carotid Arteries/pathology , Dipeptides/chemistry , Disease Models, Animal , Fibrinogen/chemistry , Fibrinolysis , Fucose/chemistry , Glucose/chemistry , Humans , Intravital Microscopy , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Serum Albumin/chemistry , Sulfates/chemistry , Thrombelastography , Thrombosis/metabolism , Tissue Plasminogen Activator/chemistry , Venous Thrombosis/immunology
10.
Am J Cardiol ; 118(3): 311-8, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27289291

ABSTRACT

It is unknown whether inflammatory/hemostatic biomarkers are associated with coronary artery calcium (CAC) progression. Our purpose was to evaluate the associations of baseline levels of C-reactive protein, fibrinogen, plasminogen activator inhibitor-1 (PAI-1), tissue plasminogen activator antigen, and circulating factor VII with CAC progression in healthy midlife women. Inflammatory/hemostatic biomarkers were measured at baseline. CAC was quantified by computed tomography scans at baseline and after 2.3 ± 0.5 years of follow-up. Significant CAC progression was defined as present if (1) follow-up CAC Agatston score was >0 if baseline CAC score = 0; (2) annualized change in CAC score was ≥10 if baseline CAC score >0 to <100; and (3) annualized percent change in CAC score was ≥10% if baseline CAC score ≥100. Extent of CAC progression was defined as [log(CAC(follow-up)+25) - log(CAC(baseline)+25)]/year. Logistic and linear regression models were used as appropriate, and the final models were adjusted for baseline CAC score, age, study site, race/ethnicity, menopausal status, sociodemographics, traditional cardiovascular disease (CVD) risk factors, family history of CVD, and CVD medication use. The study included 252 women (baseline age 51.2 ± 2.6 years; 67.5% white; 56.4% premenopausal or early perimenopausal). In final models, only log(PAI-1) was associated with presence of CAC progression (odds ratio 1.91, 95% CI 1.24 to 2.93; per 1 log unit increase in PAI-1; p = 0.003). In addition, higher log(PAI-1) was marginally associated with greater extent of CAC progression (p = 0.06). In conclusion, PAI-1 is associated with the presence of CAC progression in middle-aged women. Targeting PAI-1 may decrease atherogenesis beyond conventional CVD risk factors.


Subject(s)
Calcinosis/metabolism , Coronary Artery Disease/metabolism , Adult , Antigens/immunology , Biomarkers/metabolism , C-Reactive Protein/metabolism , Calcinosis/diagnostic imaging , Calcinosis/immunology , Cohort Studies , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/immunology , Coronary Vessels/diagnostic imaging , Disease Progression , Factor VII/metabolism , Female , Fibrinogen/metabolism , Humans , Linear Models , Logistic Models , Middle Aged , Plasminogen Activator Inhibitor 1/metabolism , Prospective Studies , Tissue Plasminogen Activator/immunology , Tomography, X-Ray Computed
11.
Biochim Biophys Acta ; 1862(3): 395-402, 2016 03.
Article in English | MEDLINE | ID: mdl-26493446

ABSTRACT

The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Subject(s)
Blood-Brain Barrier/immunology , Central Nervous System Diseases/immunology , Inflammation/immunology , Microglia/immunology , Plasminogen/immunology , Animals , Blood-Brain Barrier/pathology , Central Nervous System Diseases/pathology , Fibrin/immunology , Fibrinolysin/immunology , Humans , Inflammation/pathology , Leukocytes/immunology , Leukocytes/pathology , Microglia/pathology , Tissue Plasminogen Activator/immunology
12.
J Cardiovasc Pharmacol Ther ; 21(4): 372-80, 2016 07.
Article in English | MEDLINE | ID: mdl-26657322

ABSTRACT

BACKGROUND: Thrombolysis is an effective treatment strategy for prosthetic valve thrombosis (PVT). Recombinant tissue-type plasminogen activator (rt-PA) is widely used as a thrombolytic agent. Infusion of rt-PA may trigger the production of anti-tissue plasminogen activator (tPA) antibodies (ATAs). We aimed to evaluate the possible relationship between ATA levels and PVT formation, and the role of baseline ATA levels on outcomes of thrombolytic therapy in patients with PVT. METHODS: This prospective, single-center cohort study included 28 patients with PVT undergoing thrombolysis and 31 controls with normal prostheses. Plasma samples were collected from patients with PVT at baseline and at 15th, 30th, 90th, and 180th days after thrombolysis and from controls at baseline only. The ATA levels were assessed in human plasma by an enzyme-linked immunosorbent assay. RESULTS: Baseline ATA-immunoglobulin (Ig) G and IgM were significantly higher in patients with PVT than in controls. The levels of IgM and IgG peaked at 15th and 30th days after rt-PA infusion, respectively. Subtherapeutic international normalized ratio and baseline ATA-IgM were independent predictors of PVT. Thrombolysis failed in 6 patients (21%) in whom baseline IgM levels were significantly higher than successfully lysed patients. Rethrombosis occurred in 9 patients (32%) in whom baseline IgG levels were significantly higher than those without rethrombosis. There was a moderate positive correlation between baseline and 15th-day IgM levels and the dose of rt-PA needed for successful lysis. CONCLUSION: The ATA levels tended to be higher in patients with PVT at the time of initial diagnosis compared to controls without PVT. In addition, such patients with PVT and high ATA levels may be at high risk for failed thrombolysis or rethrombosis.


Subject(s)
Antibodies/blood , Fibrinolytic Agents/immunology , Heart Valve Diseases/drug therapy , Heart Valve Prosthesis Implantation/adverse effects , Heart Valve Prosthesis Implantation/instrumentation , Heart Valve Prosthesis , Immunoglobulin G/blood , Immunoglobulin M/blood , Thrombolytic Therapy/methods , Thrombosis/drug therapy , Tissue Plasminogen Activator/immunology , Adult , Aged , Biomarkers/blood , Case-Control Studies , Female , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/adverse effects , Heart Valve Diseases/diagnosis , Heart Valve Diseases/etiology , Humans , Male , Middle Aged , Pilot Projects , Prospective Studies , Recurrence , Risk Factors , Thrombolytic Therapy/adverse effects , Thrombosis/diagnosis , Thrombosis/etiology , Time Factors , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/adverse effects , Treatment Outcome , Young Adult
14.
J Cereb Blood Flow Metab ; 34(5): 802-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24473480

ABSTRACT

The effect of recombinant human tissue plasminogen activator (rtPA) on neuroinflammation after stroke remains largely unknown. Here, we tested the effect of rtPA on expression of cellular adhesion molecules, chemokines, and cytokines, and compared those with levels of inflammatory cell recruitment, brain injury, and mortality over 3 days after transient middle cerebral artery occlusion (MCAO) in mice. Mortality was dramatically increased after rtPA treatment compared with saline treatment during the first day of reperfusion. Among the animals that survived, rtPA significantly increased CCL3 expression, microglia recruitment, and cerebral infarction 6 hours after MCAO. In contrast, the extent of neutrophils and macrophages infiltration in the brain was similar in both saline- and rtPA-treated animals. Recombinant human tissue plasminogen activator induced Il1b and Tnf expression, 6 and 72 hours after MCAO, respectively, and dramatically reduced interleukin 6 (IL-6) level 24 hours after reperfusion. A dose response study confirmed the effect of rtPA on CCL3 and Il1b expressions. The effect was similar at the doses of 1 and 10 mg/kg. In conclusion, we report for the first time that rtPA amplified microglia recruitment early after stroke in association with a rapid CCL3 production. This early response may take part in the higher susceptibility of rtPA-treated animals to reperfusion injury.


Subject(s)
Chemokine CCL3/immunology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/immunology , Tissue Plasminogen Activator/adverse effects , Tissue Plasminogen Activator/immunology , Animals , Brain/blood supply , Brain/drug effects , Brain/pathology , Chemokine CCL3/genetics , Gene Expression Regulation/drug effects , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Inflammation/chemically induced , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Male , Mice , Microglia/drug effects , Microglia/immunology , RNA, Messenger/genetics , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
15.
Atherosclerosis ; 231(1): 54-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24125410

ABSTRACT

OBJECTIVE: To evaluate whether higher circulating levels of complement proteins C3 and C4 are associated with menopausal status and with hemostatic/thrombus formation markers (circulating factor VII (factor VIIc), fibrinogen, plasminogen activator inhibitor-1 (PAI-1) and tissue plasminogen activator antigen (tPA-ag)) in a sample of midlife women. METHODS AND RESULTS: A total of 100 women (50 late peri-/postmenopausal and 50 pre-/early peri- menopausal women) from the Study of Women's Health Across the Nation (SWAN) Pittsburgh site were included in the present analysis. Factor VIIc and PAI-1 were log transformed. Linear regression was used for analysis. The mean age of the study participants was 50.5 ± 2.6 years with 73% were Caucasian and 27% were African American. C3 but not C4 was significantly higher in postmenopausal women compared to premenopausal women (P value = 0.03), adjusting for age, race and BMI. In final model (adjusting for age, race, BMI and menopausal status), C3 was associated with higher levels of log PAI-1 (P value = 0.0009) and tPA-ag (P value = 0.0003), while C4 was associated with higher levels of log factor VIIc (P value = 0.04) and fibrinogen (P value = 0.005). CONCLUSIONS: These data suggest that C3 and C4 may be related to blood clots via their associations with hemostatic markers and that C3 is related to menopausal status. Complement proteins C3 and C4 could be possible pathways by which postmenopausal women are at higher risk of atherosclerosis and cardiovascular related events. It is important to replicate these findings in a larger sample size.


Subject(s)
Complement C3/analysis , Complement C4/analysis , Hemostasis/physiology , Menopause/blood , Premenopause/blood , Black or African American , Cross-Sectional Studies , Factor VII/analysis , Female , Fibrinogen/analysis , Humans , Longitudinal Studies , Middle Aged , Plasminogen Activator Inhibitor 1/blood , Tissue Plasminogen Activator/blood , Tissue Plasminogen Activator/immunology , White People , Women's Health
16.
PLoS One ; 8(5): e63118, 2013.
Article in English | MEDLINE | ID: mdl-23658802

ABSTRACT

The only approved pharmacological treatment for ischemic stroke is intravenous administration of plasminogen activator (tPA) to re-canalize the occluded cerebral vessel. Not only reperfusion but also tPA itself can induce an inflammatory response. Microglia are the innate immune cells of the central nervous system and the first immune cells to become activated in stroke. Neuroserpin, an endogenous inhibitor of tPA, is up-regulated following cerebral ischemia. To examine neuroserpin-dependent mechanisms of neuroprotection in stroke, we studied neuroserpin deficient (Ns(-/-))mice in an animal model of temporal focal ischemic stroke. Infarct size and neurological outcome were worse in neuroserpin deficient mice even though the fibrinolytic activity in the ischemic brain was increased. The increased infarct size was paralleled by a selective increase in proinflammatory microglia activation in Ns(-/-) mice. Our results show excessive microglial activation in Ns(-/-) mice mediated by an increased activity of tPA. This activation results in a worse outcome further underscoring the potential detrimental proinflammatory effects of tPA.


Subject(s)
Brain Ischemia/pathology , Microglia/pathology , Neuropeptides/genetics , Serpins/genetics , Stroke/pathology , Tissue Plasminogen Activator/genetics , Animals , Brain Ischemia/genetics , Brain Ischemia/immunology , Disease Models, Animal , Gene Expression Regulation , Inflammation , Mice , Mice, Knockout , Microglia/immunology , Neuropeptides/deficiency , Neutrophil Infiltration , Serpins/deficiency , Stroke/genetics , Stroke/immunology , Tissue Plasminogen Activator/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Neuroserpin
17.
Stroke ; 42(8): 2315-22, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680906

ABSTRACT

BACKGROUND AND PURPOSE: Tissue-type plasminogen activator (tPA) is the only drug approved for the acute treatment of ischemic stroke but with two faces in the disease: beneficial fibrinolysis in the vasculature and damaging effects on the neurovascular unit and brain parenchyma. To improve this profile, we developed a novel strategy, relying on antibodies targeting the proneurotoxic effects of tPA. METHODS: After production and characterization of antibodies (αATD-NR1) that specifically prevent the interaction of tPA with the ATD-NR1 of N-methyl-d-aspartate receptors, we have evaluated their efficacy in a model of murine thromboembolic stroke with or without recombinant tPA-induced reperfusion, coupled to MRI, near-infrared fluorescence imaging, and behavior assessments. RESULTS: In vitro, αATD-NR1 prevented the proexcitotoxic effect of tPA without altering N-methyl-d-aspartate-induced neurotransmission. In vivo, after a single administration alone or with late recombinant tPA-induced thrombolysis, antibodies dramatically reduced brain injuries and blood-brain barrier leakage, thus improving long-term neurological outcome. CONCLUSIONS: Our strategy limits ischemic damages and extends the therapeutic window of tPA-driven thrombolysis. Thus, the prospect of this immunotherapy is an extension of the range of treatable patients.


Subject(s)
Antibodies/therapeutic use , Brain Ischemia/drug therapy , Fibrinolytic Agents/therapeutic use , Receptors, N-Methyl-D-Aspartate/immunology , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Animals , Antibodies/immunology , Brain/drug effects , Brain/immunology , Brain Ischemia/immunology , Fibrinolytic Agents/immunology , Mice , Stroke/immunology , Tissue Plasminogen Activator/immunology
18.
Appl Microbiol Biotechnol ; 91(3): 731-40, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21556920

ABSTRACT

Ideal immunogenicity in antigens is a prerequisite to eliciting a sufficiently strong immune and memory response via either DNA or protein vaccines. To improve immunogenicity, efforts have focused on high-level expression of target proteins and on maintaining their natural conformations. In the present work, two trimer motifs (MTQ and MTI) were designed and introduced into a plasmid vector with the tissue plasminogen activator signal peptide (tPA-SP). Next, we examined the efficacy and the efficiency of the two motifs as well as the introduction of tPA-SP and its mutant forms, 22P/A and 22P/G, in facilitating the secretory expression of trimeric proteins in mammalian cells. We found that both trimer motifs could produce the target protein in a trimeric form at a high level. Introduction of tPA-SP 22P/A markedly increased the secretory expression level. The combination of the trimer motif, MTQ, and the signal peptide, 22P/A, may serve as a universal mammalian vector for producing trimeric proteins in vaccine development.


Subject(s)
Amino Acid Motifs/immunology , Multiprotein Complexes , Protein Sorting Signals/genetics , Tissue Plasminogen Activator/genetics , Amino Acid Sequence , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , HIV/genetics , HIV/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/metabolism , Mutation , Protein Structure, Secondary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tissue Plasminogen Activator/immunology , Vaccines, DNA/immunology , Vaccines, Subunit/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/metabolism
19.
Blood Press ; 20(4): 218-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21388252

ABSTRACT

OBJECTIVE. Recent evidence demonstrates that masked hypertension (MH) is a significant predictor of cardiovascular disease. The aim of our study was to examine the impact of MH on haemostasis parameters and to compare the findings to those of healthy normotensives matched for age, sex, body mass index and the rest of risk factors. DESIGN AND METHOD. 130 (60 male, 70 female) healthy subjects mean age 45 ± 12 years who had clinic blood pressure < 140/90 mmHg were studied. The whole study population underwent 24-h ambulatory blood pressure monitoring (ABPM). According to the ABPM recordings, 24 individuals (eight males, 16 females) had MH (daytime systolic blood pressure ≥ 135 mmHg or daytime diastolic blood pressure ≥ 85 mmHg - group A) and the remaining 106 subjects (52 males, 54 females) had normal ABPM recordings - group B. Fibrinogen, thrombomodulin ™, the antigens of plasminogen activator inhibitor 1 (PAI-1Ag) and tissue plasminogen activator (tPA-Ag) were determined in the two groups. Results. The PAI-1 Ag, tPA-Ag, fibrinogen and TM levels were significantly higher in the masked hypertensive group than to normotensive control group. CONCLUSIONS. Our findings suggest that subjects with MH have significantly higher fibrinogen, TM, PAI-1Ag and tPA-Ag plasma levels compared with normotensives. This observation may have prognostic significance for future cardiovascular events in subjects with MH and needs further investigation.


Subject(s)
Hypertension/physiopathology , Antigens/blood , Blood Pressure/physiology , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Cohort Studies , Female , Fibrinogen/metabolism , Hemostasis , Humans , Hypertension/blood , Hypertension/complications , Male , Middle Aged , Plasminogen Activator Inhibitor 1/immunology , Thrombomodulin/metabolism , Tissue Plasminogen Activator/immunology
20.
Stroke ; 41(12): 2950-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20966414

ABSTRACT

BACKGROUND AND PURPOSE: Despite its fibrinolytic effect, tissue-type plasminogen activator (tPA) displays deleterious effects in the brain, including proexcitotoxicity, that can reduce the overall benefit from thrombolysis during stroke. We have proposed that tPA potentiates excitotoxicity by interacting with and cleaving the aminoterminal end of the NR1 subunit of N-methyl-d-aspartate receptors, leading to an increased calcium influx, Erk1/2 activation, and neurotoxicity. Because this mechanism is debated, our aim was to demonstrate its in vivo occurrence and relevance. Because tPA is released under ischemic conditions, we hypothesized that if it indeed processes NR1, then the released fragment should reactivate the immune system in animals that had been immunized long before with recombinant aminoterminal end of the NR1. This effect should be exacerbated in ischemic animals thrombolysed with recombinant tPA. METHODS: At a time when specific antibodies could not be detected any longer, mice previously vaccinated with recombinant aminoterminal end of the NR1 were subjected to thromboembolic stroke induced by injecting thrombin in the middle cerebral artery alone or with intravenous thrombolysis. RESULTS: Stroke performed 1 year after active immunization induced the reappearance of antibodies against the aminoterminal end of the NR1 in the plasma, an effect significantly increased when ischemia was followed by recombinant tPA-induced reperfusion. Moreover, immunization preventing the interaction of tPA with aminoterminal end of the NR1 reduced ischemic brain damages and extended the therapeutic window of tPA-induced thrombolysis. CONCLUSIONS: We demonstrate that the tPA-dependent interaction and cleavage of the NR1 subunit of N-methyl-d-aspartate receptors occurs in vivo after stroke and that this interaction is a relevant therapeutic target for stroke treatment.


Subject(s)
Receptors, N-Methyl-D-Aspartate/physiology , Stroke/physiopathology , Tissue Plasminogen Activator/physiology , Animals , Autoantibodies/biosynthesis , Immunization , Immunohistochemistry , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Stroke/drug therapy , Stroke/immunology , Thrombin/pharmacology , Thrombolytic Therapy , Tissue Plasminogen Activator/immunology , Tissue Plasminogen Activator/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...