Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422879

ABSTRACT

Altered expression of corin, a cardiac transmembrane serine protease, has been linked to dilated and ischemic cardiomyopathy. However, the potential role of corin in myocardial infarction (MI) is lacking. This study examined the outcomes of MI in wild-type vs. cardiac-specific overexpressed corin transgenic (Corin-Tg) mice during pre-MI, early phase (3, 24, 72 h), and late phase (1, 4 weeks) post-MI. Corin overexpression significantly reduced cardiac cell apoptosis (p < 0.001), infarct size (p < 0.001), and inhibited cleavage of procaspases 3, 9, and 8 (p < 0.05 to p < 0.01), as well as altered the expression of Bcl2 family proteins, Bcl-xl, Bcl2 and Bak (p < 0.05 to p < 0.001) at 24 h post-MI. Overexpressed cardiac corin also significantly modulated heart function (ejection fraction, p < 0.0001), lung congestion (lung weight to body weight ratio, p < 0.0001), and systemic extracellular water (edema, p < 0.05) during late phase post-MI. Overall, cardiac corin overexpression significantly reduced apoptosis, infarct size, and modulated cardiac expression of key members of the apoptotic pathway in early phase post-MI; and led to significant improvement in heart function and reduced congestion in late phase post-MI. These findings suggest that corin may be a useful target to protect the heart from ischemic injury and subsequent post-infarction remodeling.


Subject(s)
Apoptosis/genetics , Myocardial Infarction/genetics , Myocardium/metabolism , Serine Endopeptidases/genetics , Animals , Cell Death/genetics , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Ventricular Remodeling/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-X Protein/genetics
2.
Blood ; 134(12): 970-978, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31395599

ABSTRACT

Stasis of venous blood triggers deep vein thrombosis by activating coagulation, yet its effects on the fibrinolytic system are not fully understood. We examined the relationship between stasis, fibrinolysis, and the development of experimental venous thrombosis. Effects of stasis-induced deep vein thrombosis and fibrinolysis on thrombosis were examined by inferior vena cava ligation in congenic mice with and without α2-antiplasmin (α2AP), the primary inhibitor of plasmin. Venous thrombus weights were measured and thrombus composition was determined by Martius scarlet blue and immunofluorescence staining. Venous thrombi from α2AP+/+ mice contained plasminogen activators, plasminogen activator inhibitor-1, plasminogen, and α2AP, which changed with thrombus age. Normal, α2AP+/+ mice developed large, occlusive thrombi within 5 hours after ligation; thrombi were even larger in plasminogen-deficient mice (P < .001). No significant thrombus formation was seen in α2AP-/- mice (P < .0001) or in α2AP+/+ mice treated with an α2AP-inactivating antibody (P < .001). Venous stasis activated fibrinolysis, measured by D-dimer levels, in α2AP-/- mice vs α2AP+/+ mice (P < .05). Inhibition of fibrinolysis by the indirect plasmin inhibitor ε-aminocaproic acid or by α2AP restored thrombosis in α2AP-/- mice. In addition to its effects on acute thrombosis, thrombus formation was also markedly suppressed in α2AP-/- mice vs α2AP+/+ mice (P < .0001) 1, 7, and 14 days after ligation. We conclude that experimental venous stasis activates the fibrinolytic system to block the development of venous thrombosis. Suppression of fibrinolysis by α2AP appears essential for stasis-induced thrombus development, which suggests that targeting α2AP may prove useful for preventing venous thrombosis.


Subject(s)
Fibrinolysis/physiology , Postthrombotic Syndrome/complications , Venous Thrombosis/prevention & control , alpha-2-Antiplasmin/physiology , Animals , Disease Models, Animal , Female , Fibrinolysis/genetics , Ligation , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Postthrombotic Syndrome/blood , Postthrombotic Syndrome/genetics , Postthrombotic Syndrome/physiopathology , Vena Cava, Inferior/pathology , Vena Cava, Inferior/surgery , Venous Thrombosis/blood , Venous Thrombosis/genetics , Venous Thrombosis/physiopathology , alpha-2-Antiplasmin/genetics
3.
Int J Mol Sci ; 21(1)2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31892216

ABSTRACT

Humans with dilated cardiomyopathy (DCM) and heart failure (HF) develop low levels of corin, a multi-domain, cardiac-selective serine protease involved in natriuretic peptide cleavage and sodium and water regulation. However, experimental restoration of corin levels markedly attenuates HF progression. To determine whether the beneficial effects of corin in HF require catalytic activity, we engineered cardiac overexpression of an enzymatically inactive corin transgene (corin-Tg(i)). On a wild-type (WT) background, corin-Tg(i) had no evident phenotypic effects. However, in a well-established genetic model of DCM, corin-Tg(i)/DCM mice had increased survival (p < 0.01 to 0.001) vs. littermate corin-WT/DCM controls. Pleural effusion (p < 0.01), lung edema (p < 0.05), systemic extracellular free water (p < 0.01), and heart weight were decreased (p < 0.01) in corin-Tg(i)/DCM vs. corin-WT/DCM mice. Cardiac ejection fraction and fractional shortening improved (p < 0.01), while ventricular dilation decreased (p < 0.0001) in corin-Tg(i)/DCM mice. Plasma atrial natriuretic peptide, cyclic guanosine monophosphate, and neprilysin were significantly decreased. Cardiac phosphorylated glycogen synthase kinase-3ß (pSer9-GSK3ß) levels were increased in corin(i)-Tg/DCM mice (p < 0.01). In summary, catalytically inactive corin-Tg(i) decreased fluid retention, improved contractile function, decreased HF biomarkers, and diminished cardiac GSK3ß activity. Thus, the protective effects of cardiac corin on HF progression and survival in experimental DCM do not require the serine protease activity of the molecule.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Edema/metabolism , Myocardial Contraction/physiology , Serine Endopeptidases/metabolism , Animals , Biomarkers/metabolism , Female , Glycogen Synthase Kinase 3 beta/metabolism , Heart/physiopathology , Heart Failure/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism
4.
Neuroscience ; 376: 40-47, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29294343

ABSTRACT

During acute brain ischemia, α2-antiplasmin markedly enhances brain injury, blood-brain barrier breakdown and matrix metalloproteinase-9 (MMP-9) expression. Although α2-antiplasmin inhibits fibrin thrombus-degradation, and MMP-9 is a collagen-degrading enzyme altering blood-brain barrier, both have similar deleterious effects on the ischemic brain. We examined the hypothesis that MMP-9 is an essential downstream mediator of α2-antiplasmin's deleterious effects during brain ischemia. Middle cerebral artery thromboembolic stroke was induced in a randomized, blinded fashion in mice with increased blood levels of α2-antiplasmin. There was a robust increase in MMP-9 expression (immunofluorescence) in the ischemic vs. the non-ischemic hemisphere of MMP-9+/+ but not MMP-9-/- mice, 24 h after stroke. Brain swelling and hemorrhage were significantly increased in the ischemic vs. the non-ischemic hemisphere of MMP-9+/+ mice. By comparison to MMP-9+/+ mice, the ischemic hemispheres of MMP-9-/- mice showed a ∼6-fold reduction in brain swelling (p < 0.001) and a ∼9-fold reduction in brain hemorrhage. Brain infarction (p < 0.0001) and TUNEL-positive cell death (p < 0.001) were significantly diminished in the ischemic hemisphere of MMP-9-/- mice vs. MMP-9+/+ mice. Ischemic breakdown of the blood-brain barrier and fibrin deposition were also significantly reduced in MMP-9-/- mice vs. MMP-9+/+ mice (p < 0.05), as measured by quantitative immunofluorescence. We conclude that MMP-9 deficiency ablates many of the deleterious effects of high α2-antiplasmin levels, significantly reducing blood-brain barrier breakdown, TUNEL-positive cell death, brain hemorrhage, swelling and infarction. This suggests that the two molecules may be in a shared pathway in which MMP-9 is essential downstream for the deleterious effects of α2-antiplasmin in ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Matrix Metalloproteinase 9/metabolism , Stroke/metabolism , alpha-2-Antiplasmin/metabolism , Animals , Blood-Brain Barrier/pathology , Brain Ischemia/pathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Disease Models, Animal , Female , Fibrinogen/metabolism , Male , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , Mice, Knockout , Random Allocation , Single-Blind Method , Stroke/pathology
5.
Semin Thromb Hemost ; 43(2): 191-199, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27472428

ABSTRACT

Thrombotic vascular occlusion is the leading cause of ischemic stroke. High blood levels of α2-antiplasmin (a2AP), an ultrafast, covalent inhibitor of plasmin, have been linked in humans to increased risk of ischemic stroke and failure of tissue plasminogen activator (tPA) therapy. Consistent with these observations, a2AP neutralizes the therapeutic benefit of tPA therapy in experimental stroke. In addition, a2AP has deleterious, dose-related effects on ischemic brain injury in the absence of therapy. Experimental therapeutic inactivation of a2AP markedly reduces microvascular thrombosis, ischemic brain injury, brain swelling, brain hemorrhage, and death after thromboembolic stroke. These data provide new insights into the critical importance of a2AP in the pathogenesis of ischemic brain injury and suggest that transiently inactivating a2AP may have therapeutic value in ischemic stroke.


Subject(s)
Stroke/drug therapy , alpha-2-Antiplasmin/therapeutic use , Humans , Stroke/mortality , Stroke/pathology , alpha-2-Antiplasmin/administration & dosage
6.
Arterioscler Thromb Vasc Biol ; 34(12): 2586-93, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25256235

ABSTRACT

OBJECTIVE: Ischemic stroke is primarily attributable to thrombotic vascular occlusion. Elevated α2-antiplasmin (a2AP) levels correlate with increased stroke risk, but whether a2AP contributes to the pathogenesis of stroke is unknown. We examined how a2AP affects thrombosis, ischemic brain injury, and survival after experimental cerebral thromboembolism. APPROACH AND RESULTS: We evaluated the effects of a2AP on stroke outcomes in mice with increased, normal, or no circulating a2AP, as well as in mice given an a2AP-inactivating antibody. Higher a2AP levels were correlated with greater ischemic brain injury (rs=0.88, P<0.001), brain swelling (rs=0.82, P<0.001), and reduced middle cerebral artery thrombus dissolution (rs=-0.93, P<0.001). In contrast, a2AP deficiency enhanced thrombus dissolution, increased cerebral blood flow, reduced brain infarction, and decreased brain swelling. By comparison to tissue plasminogen activator (TPA), a2AP inactivation hours after thromboembolism still reduced brain infarction (P<0.001) and hemorrhage (P<0.05). Microvascular thrombosis, a process that enhances brain ischemia, was markedly reduced in a2AP-deficient or a2AP-inactivated mice compared with TPA-treated mice or mice with increased a2AP levels (all P<0.001). Matrix metalloproteinase-9 expression, which contributes to acute brain injury, was profoundly decreased in a2AP-deficient or a2AP-inactivated mice versus TPA-treated mice or mice with increased a2AP levels (all P<0.001). a2AP inactivation markedly reduced stroke mortality versus TPA (P<0.0001). CONCLUSIONS: a2AP has profound, dose-related effects on ischemic brain injury, swelling, hemorrhage, and survival after cerebral thromboembolism. By comparison to TPA, the protective effects of a2AP deficiency or inactivation seem to be mediated through reductions in microvascular thrombosis and matrix metalloproteinase-9 expression.


Subject(s)
Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/pathology , alpha-2-Antiplasmin/metabolism , Animals , Brain/metabolism , Brain/pathology , Cerebrovascular Circulation , Disease Models, Animal , Fibrinolysis , Infarction, Middle Cerebral Artery/enzymology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , alpha-2-Antiplasmin/deficiency , alpha-2-Antiplasmin/genetics
7.
Exp Neurol ; 255: 56-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24556477

ABSTRACT

High blood levels of α2-antiplasmin have been associated with failed tissue plasminogen activator (TPA) therapy for ischemic stroke. Yet, other data suggests that α2-antiplasmin may be protective in stroke, because it defends against bleeding and excitotoxicity. To address this paradox, we examined the effects of high α2-antiplasmin levels and α2-antiplasmin inactivation in mice treated with TPA 0.5-2.5h after middle cerebral artery (MCA) thromboembolism. Brain infarction, swelling, hemorrhage, blood brain barrier breakdown and neuronal apoptosis were measured by a blinded observer. Thrombus dissolution was determined by gamma counting. During TPA treatment, high α2-antiplasmin blood levels increased brain infarction (2.2-fold) and swelling (3.7-fold), but decreased MCA thrombus dissolution. Conversely, α2-antiplasmin inactivation during TPA treatment reduced brain infarction, hemorrhage and swelling, but increased MCA thrombus dissolution. Inactivation of α2-antiplasmin during TPA treatment reduced neuronal apoptosis and blood brain barrier breakdown. Inactivation of α2-antiplasmin also reduced short-term mortality. Taken together these data show that α2-antiplasmin opposes the effects of TPA therapy and contributes to enhanced brain injury after experimental thromboembolic stroke. Conversely, α2-antiplasmin inactivation during TPA treatment improves thrombus dissolution and reduces brain infarction, swelling and hemorrhage. Consistent with clinical observations, these data suggest that α2-antiplasmin exerts deleterious effects that reduce the efficacy and safety of TPA therapy for ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Fibrinolytic Agents/therapeutic use , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , alpha-2-Antiplasmin/metabolism , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Ischemia/blood , Disease Models, Animal , Fibrinolytic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Stroke/blood , Tissue Plasminogen Activator/pharmacology
8.
J Cardiovasc Pharmacol ; 63(6): 520-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24477045

ABSTRACT

P2Y(12) receptor antagonism inhibits platelet aggregation by preventing adenosine diphosphate (ADP)-mediated amplification of activation pathways downstream of primary agonists, such as thrombin and collagen. However, the role of ADP signaling in maintaining aggregate stability and the effects of P2Y(12) antagonists on preestablished aggregates in vitro and arterial thrombus in vivo are not well understood. This study evaluated the impact of P2Y(12) signaling on platelet aggregate stability and early thrombotic occlusion using a reversible P2Y(12) antagonist, ticagrelor. There were 2 study objectives: (1) to determine if there was a time-dependent factor on the capacity of a P2Y(12) antagonist to affect human platelet aggregate stability in vitro using light transmission aggregometry and (2) to evaluate the extent of arterial thrombus reversal in a preclinical model upon administration of ticagrelor in vivo. Platelet aggregates were exposed to ticagrelor after ADP or collagen activation, monitored for stability by aggregometry, and visualized by microscopy. Freshly formed ADP- and collagen-induced platelet aggregates were more rapidly dispersed by a P2Y(12) antagonist than drug carrier control at clinically relevant concentrations (P < 0.05). However, stable aggregates were not noticeably affected. A murine arterial thrombosis model was used to evaluate thrombus stability in an in vivo mouse model. Thrombotic occlusion was induced by FeCl(3), followed by a bolus intravenous administration of ticagrelor or vehicle control. Doppler blood flow was monitored before injury and 30 minutes after bolus administration. Arteries were retrieved for inspection for residual thrombus. Early arterial thrombotic occlusion in vivo was partially reversed by ticagrelor administration. Blood flow through the injured artery increased, and thrombus size within the artery decreased (P < 0.05, n = 3). In conclusion, P2Y(12) antagonism disrupts the stability of newly formed platelet aggregates, promoting disaggregation, and reverses thrombotic vascular occlusion. Thus, in addition to activating platelets, signaling via P2Y(12) seems to be required for stabilizing platelet thrombi.


Subject(s)
Platelet Aggregation/physiology , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/metabolism , Signal Transduction/physiology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/therapeutic use , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/therapeutic use , Signal Transduction/drug effects , Thrombosis/drug therapy , Thrombosis/metabolism , Ticagrelor , Time Factors
9.
Hypertension ; 63(3): 514-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24379183

ABSTRACT

Dilated cardiomyopathy is a frequent cause of heart failure and death. Atrial natriuretic peptide (ANP) is a biomarker of dilated cardiomyopathy, but there is controversy whether ANP modulates the development of heart failure. Therefore, we examined whether ANP affects heart failure, cardiac remodeling, function, and survival in a well-characterized, transgenic model of dilated cardiomyopathy. Mice with dilated cardiomyopathy with normal ANP levels survived longer than mice with partial ANP (P<0.01) or full ANP deficiency (P<0.001). In dilated cardiomyopathy mice, ANP protected against the development of heart failure as indicated by reduced lung water, alveolar congestion, pleural effusions, etc. ANP improved systolic function and reduced cardiomegaly. Pathological cardiac remodeling was diminished in mice with normal ANP as indicated by decreased ventricular interstitial and perivascular fibrosis. Mice with dilated cardiomyopathy and normal ANP levels had better systolic function (P<0.001) than mice with dilated cardiomyopathy and ANP deficiency. Dilated cardiomyopathy was associated with diminished cardiac transcripts for NP receptors A and B in mice with normal ANP and ANP deficiency, but transcripts for NP receptor C and C-type natriuretic peptide were selectively altered in mice with dilated cardiomyopathy and ANP deficiency. Taken together, these data indicate that ANP has potent effects in experimental dilated cardiomyopathy that reduce the development of heart failure, prevent pathological remodeling, preserve systolic function, and reduce mortality. Despite the apparent overlap in physiological function between the NPs, these data suggest that the role of ANP in dilated cardiomyopathy and heart failure is not compensated physiologically by other NPs.


Subject(s)
Atrial Natriuretic Factor/metabolism , Cardiomyopathy, Dilated/metabolism , Heart Failure/metabolism , Heart Ventricles/physiopathology , Ventricular Function, Left/physiology , Ventricular Remodeling , Animals , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Disease Progression , Heart Failure/etiology , Heart Failure/mortality , Mice , Mice, Inbred C57BL , Survival Rate/trends
10.
Hypertension ; 61(2): 327-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23232642

ABSTRACT

Heart failure, caused by dilated cardiomyopathy and other cardiac disorders such as hypertension, is a major public health problem with high morbidity and mortality. Corin, a cardiac enzyme that cleaves natriuretic peptides, is a promising biomarker of cardiomyopathy and heart failure, but its functional role in these processes is not understood. We evaluated the potential effects of corin in mice with a well-characterized model of dilated cardiomyopathy. Mice with dilated cardiomyopathy developed heart failure, reduced contractile function, cardiac fibrosis, and accelerated mortality in the setting of low corin expression. In wild-type mice, transgenic, cardiac-targeted, overexpression of corin enhanced cyclic guanosine monophosphate and blood pressure responses to pro-atrial natriuretic peptide, but did not affect heart size, contractility, body weights, survival, and blood pressure. In mice with dilated cardiomyopathy, corin overexpression significantly reduced the development of myocardial fibrosis (P<0.05). Corin overexpression also enhanced heart contractile function (fractional shortening and ejection fraction; P<0.01) and it significantly reduced heart failure as assessed by lung water (P<0.05) and alveolar congestion (P<0.001). Consistent with these observations, corin overexpression significantly prolonged life in mice with dilated cardiomyopathy (P<0.0001). These results provide the first experimental evidence that corin expression plays a role in cardiomyopathy by modulating myocardial fibrosis, cardiac function, heart failure, and survival.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Heart Failure/metabolism , Heart/physiopathology , Myocardium/metabolism , Serine Endopeptidases/metabolism , Animals , Blood Pressure/physiology , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/physiopathology , Fibrosis , Heart Failure/etiology , Heart Failure/physiopathology , Hypertension/etiology , Hypertension/metabolism , Hypertension/physiopathology , Mice , Mice, Transgenic , Myocardium/pathology , Serine Endopeptidases/genetics
11.
J Biol Chem ; 287(23): 19171-6, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22518846

ABSTRACT

Bacterial plasminogen (Pg) activators generate plasmin to degrade fibrin blood clots and other proteins that modulate the pathogenesis of infection, yet despite strong homology between mammalian Pgs, the activity of bacterial Pg activators is thought to be restricted to the Pg of their host mammalian species. Thus, we found that Streptococcus uberis Pg activator (SUPA), isolated from a Streptococcus species that infects cows but not humans, robustly activated bovine but not human Pg in purified systems and in plasma. Consistent with this, SUPA formed a higher avidity complex (118-fold) with bovine Pg than with human Pg and non-proteolytically activated bovine but not human Pg. Surprisingly, however, the presence of human fibrin overrides the species-restricted action of SUPA. First, human fibrin enhanced the binding avidity of SUPA for human Pg by 4-8-fold in the presence and absence of chloride ion (a negative regulator). Second, although SUPA did not protect plasmin from inactivation by α(2)-antiplasmin, fibrin did protect human plasmin, which formed a 31-fold higher avidity complex with SUPA than Pg. Third, fibrin significantly enhanced Pg activation by reducing the K(m) (4-fold) and improving the catalytic efficiency of the SUPA complex (6-fold). Taken together, these data suggest that indirect molecular interactions may override the species-restricted activity of bacterial Pg activators; this may affect the pathogenesis of infections or may be exploited to facilitate the design of new blood clot-dissolving drugs.


Subject(s)
Bacterial Proteins/chemistry , Enzyme Activators/chemistry , Fibrin/chemistry , Plasminogen Activators/chemistry , Plasminogen/chemistry , Streptococcus/enzymology , Animals , Bacterial Proteins/metabolism , Cattle , Cattle Diseases/enzymology , Enzyme Activation , Enzyme Activators/metabolism , Fibrin/metabolism , Humans , Plasminogen/metabolism , Plasminogen Activators/metabolism , Protein Binding , Species Specificity
12.
Circ Heart Fail ; 4(2): 114-20, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21216831

ABSTRACT

BACKGROUND: By promoting salt and water excretion, the corin and the atrial natriuretic peptide (ANP) system should help to maintain fluid balance in heart failure. Yet, the development of fluid retention despite high levels of ANP-related peptides suggests that this compensatory system is limited. METHODS AND RESULTS: Levels of circulating corin (the pro-ANP-converting enzyme) and pro-ANP were measured in hospitalized patients with heart failure, using novel immunoassays. Patients (n=14) had severe heart failure (New York Heart Association class III-IV) with a median ejection fraction of 18% and median brain natriuretic peptide levels of 1940 pg/mL. In heart failure, median plasma corin levels were 7.6-fold lower than measured in plasma from 16 normal control subjects (180 versus 1368 pg/mL, P<0.01). In contrast, in patients with heart failure, levels of plasma N-terminal ANP peptides (N-ANP and pro-ANP) levels were markedly elevated (42.0 versus 7.5 ng/mL, P<0.01). Levels of uncleaved pro-ANP, measured by novel immunoassays, were significantly higher in patients with heart failure (P<0.01), suggesting that corin cleavage of pro-ANP was impaired. Median plasma levels of cyclic guanosine monophosphate were elevated in patients with heart failure (150.0 versus 7.6 pmol/mL, P<0.01), and plasma cyclic guanosine monophosphate levels positively correlated with the fractional amount of cleaved pro-ANP (r(s)=0.59, P<0.03) but not with levels of uncleaved pro-ANP, implying that the cellular response to ANP remained intact. CONCLUSIONS: Taken together, these data suggest that there may be patients for whom low corin levels and impaired pro-ANP cleavage contribute to acute decompensation.


Subject(s)
Atrial Natriuretic Factor/blood , Heart Failure/blood , Serine Endopeptidases/blood , Adolescent , Adult , Aged , Biomarkers/blood , Case-Control Studies , Cyclic GMP/blood , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Heart Failure/physiopathology , Humans , Male , Middle Aged , Severity of Illness Index , Stroke Volume , Ventricular Function , Young Adult
13.
Circulation ; 120(9): 785-91, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19687360

ABSTRACT

BACKGROUND: Platelet aggregation plays a critical role in myocardial infarction and stroke; however, the role of platelet secretion in atherosclerotic vascular disease is poorly understood. Therefore, we examined the hypothesis that platelet dense-granule secretion modulates thrombosis, inflammation, and atherosclerotic vascular remodeling after injury. METHODS AND RESULTS: Functional deletion of the Hermansky-Pudlak syndrome 3 gene (HPS3(-/-)) markedly reduces platelet dense-granule secretion. HPS3(-/-) mice have normal platelet counts, platelet morphology, and alpha-granule number, as well as maximal secretion of the alpha-granule marker P-selectin; however, their capacity to form platelet-leukocyte aggregates is significantly reduced (P<0.05). To examine the role of platelet dense-granule secretion in these processes, atherosclerosis-prone mice with combined genetic deficiency of apolipoprotein E and HPS3 (ApoE(-/-), HPS3(-/-)) were compared with congenic, atherosclerosis-prone mice with normal platelet secretion (ApoE(-/-), HPS3(+/+)). After 16 to 18 weeks on a high-fat diet, both groups of mice had similar fasting cholesterol levels and body weight. Carotid arteries of ApoE(-/-), HPS3(+/+) mice thrombosed rapidly after FeCl(3) injury, but ApoE(-/-), HPS3(-/-) mice were completely resistant to thrombotic arterial occlusion (P<0.01). Three weeks after injury, neointimal hyperplasia (from alpha-smooth muscle actin-positive cells) was significantly less (P<0.001) in arteries from ApoE(-/-), HPS3(-/-) mice. In ApoE(-/-), HPS3(-/-) mice, there were also pronounced reductions in arterial inflammation, as indicated by a 74% decrease in CD45-positive leukocytes (P<0.01) and a 73% decrease in Mac-3-positive macrophages (P<0.05). CONCLUSIONS: In atherosclerotic mice, reduced platelet dense-granule secretion is associated with marked protection against the development of arterial thrombosis, inflammation, and neointimal hyperplasia after vascular injury.


Subject(s)
Blood Platelets/metabolism , Carotid Artery Diseases/blood , Carotid Artery Diseases/pathology , Platelet Aggregation/physiology , Thrombosis/blood , Adenosine Triphosphate/metabolism , Animals , Apolipoproteins E/genetics , Dietary Fats/pharmacology , Disease Models, Animal , Hyperplasia , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Serotonin/metabolism , Tunica Intima/immunology , Tunica Intima/pathology , Vasculitis/blood , Vasculitis/pathology
14.
Am J Physiol Heart Circ Physiol ; 296(3): H655-61, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19122164

ABSTRACT

Acute coronary artery occlusion triggers the release of atrial natriuretic peptide (ANP) from the heart. ANP affects vasodilation, natriuresis, and inflammation, but the integrated biological effects of ANP on myocardial infarction are unknown. To elucidate these effects, the left anterior coronary artery was ligated in anesthetized, ANP-deficient (ANP(-/-)) and congenic wild-type (ANP(+/+)) mice. The survival of ANP(-/-) mice was markedly better (56%) at 30 days postinfarction than the survival of ANP(+/+) mice (20%, P < 0.01). Surviving mice were comparable initially, but ANP(-/-) mice developed more cardiac hypertrophy (P < 0.001) and had lower contractility indexes 30 days after infarction (P < 0.05). An analysis 24 h after coronary occlusion showed that ANP(-/-) mice had smaller infarcts than ANP(+/+) mice (62.6 +/- 12.1 vs. 100.8 +/- 3.8%, P < 0.001) adjusted for comparable areas at risk for ischemia. The administration of ANP to ANP(-/-) mice via osmotic minipumps significantly enlarged infarct size to levels comparable with those observed in ANP(+/+) mice (P < 0.05). There was no difference in neutrophil migration into the noninfarcted myocardium of ANP(-/-) mice undergoing actual versus sham-operated coronary occlusion. By comparison, after coronary occlusion, the neutrophil infiltration into the myocardium was enhanced in ANP(+/+) (P < 0.0005) and ANP(-/-) mice administered ANP (P < 0.0005). The expression of P-selectin, a molecule that mediates neutrophil adhesion, was significantly greater after coronary occlusion in the vasculature of ANP(+/+) or ANP(-/-) mice treated with ANP than in ANP(-/-) mice (P < 0.002). Taken together, these results indicate that ANP increases P-selectin, neutrophil infiltration, infarct size, and mortality following experimental coronary occlusion.


Subject(s)
Atrial Natriuretic Factor/metabolism , Coronary Occlusion/complications , Inflammation/etiology , Myocardial Infarction/etiology , Myocardium/metabolism , Animals , Atrial Natriuretic Factor/administration & dosage , Atrial Natriuretic Factor/deficiency , Atrial Natriuretic Factor/genetics , Cardiomegaly/etiology , Cardiomegaly/metabolism , Coronary Occlusion/metabolism , Coronary Occlusion/pathology , Coronary Occlusion/physiopathology , Disease Models, Animal , Hemodynamics , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Neutrophil Infiltration , P-Selectin/metabolism , Time Factors
15.
Biochem Biophys Res Commun ; 373(1): 130-5, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18549807

ABSTRACT

N-glycosylation may influence the subcellular localization and biological activity of the pro-ANP convertase, corin. In HEK293-corin cells, the inhibition of N-glycosylation, with tunicamycin, reduced the cell-surface expression of murine corin, but did not alter the total expression. Therefore, tunicamycin treatment likely caused the intracellular accumulation of non-glycosylated corin. Tunicamycin treatment also significantly reduced corin activity (pro-ANP cleavage) in these cells. We developed an assay to measure the effect of N-glycosylation on corin activity, independent of its effect on corin localization. We determined that the reduction in corin activity was due to a direct effect of N-glycosylation, and was not secondary to the effect of N-glycosylation on corin cell-surface expression. Our data provide evidence that N-glycosylation is essential for the cell-surface expression of murine corin and modulates its functional activity. N-Glycosylation represents a possible mechanism for the regulation of native corin on the surface of cardiomyocytes.


Subject(s)
Atrial Natriuretic Factor/metabolism , Cell Membrane/enzymology , Serine Endopeptidases/metabolism , Animals , Catalysis , Cell Line , Glycosylation/drug effects , Humans , Mice , Myocytes, Cardiac/enzymology , Recombinant Proteins/metabolism , Tunicamycin/pharmacology
16.
J Mol Cell Cardiol ; 44(1): 131-42, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17996891

ABSTRACT

The multi-domain transmembrane serine protease corin cleaves pro-atrial natriuretic peptide (pro-ANP) in vitro to generate an active hormone, ANP. Corin may also contribute to the regulation of the natriuretic peptide system in vivo, and might be an attractive target for treatment of cardiovascular diseases. In order for corin to cleave its substrate pro-ANP, it should be catalytically active and located proximally. However, because knowledge of native corin is limited, we examined the expression, cardiac localization and molecular forms of the native corin protein. Immunofluorescence studies using a series of anti-corin antibodies directed against the stem and protease domains reveal that corin is present on the cell-surface of rat neonatal cardiomyocytes and murine HL-1 cardiomyocyte-like cells. Furthermore, we immunolocalized native corin in pro-ANP expressing cardiomyocytes. Immunoprecipitation of the membrane fraction of mouse heart extract showed that native corin had a relative mass of 205-210 kDa. Under reducing conditions native corin migrates as several different molecular weight forms corresponding to zymogen (uncleaved) and active (cleaved) forms. Studies using a FITC-tagged chloromethyl ketone that mimics the corin cleavage sequence in pro-ANP, suggest that an enzymatically active form of corin is localized to the cell surface of myocardial cells in vivo. Additionally, we showed that the 205-210 kDa form of corin is a glycosylated protein. Treatment of HL-1 cells with tunicamycin reduced the relative mass of expressed corin. We conclude that native corin is a glycosylated protease that is localized on the cell surface of pro-ANP-expressing cardiomyocytes in both zymogen and catalytically active forms.


Subject(s)
Atrial Natriuretic Factor/metabolism , Enzyme Precursors/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/enzymology , Protein Precursors/metabolism , Serine Endopeptidases/metabolism , Animals , Antibodies, Monoclonal/biosynthesis , Blotting, Western , Catalysis , Cell Line , Enzyme Activation , Female , Humans , Mice , Mice, Inbred BALB C , Molecular Weight , Myocytes, Cardiac/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley , Serine Endopeptidases/chemistry
17.
Circulation ; 110(2): 170-6, 2004 Jul 13.
Article in English | MEDLINE | ID: mdl-15210587

ABSTRACT

BACKGROUND: Activated factor XIII (FXIIIa) mediates fibrinolytic resistance and is a hallmark of newly formed thrombi. In vivo imaging of FXIIIa activity could further elucidate the role of this molecule in thrombosis and other biological processes and aid in the clinical detection of acute thrombi. METHODS AND RESULTS: An FXIIIa-sensitive near-infrared fluorescence imaging agent (A15) was engineered by conjugating a near-infrared fluorochrome to a peptide ligand derived from the amino terminus of alpha2-antiplasmin. To evaluate the molecular specificity of A15 for FXIIIa, a control agent (C15) was also synthesized by modifying a single key glutamine residue in A15. Fluorescence imaging experiments with A15 demonstrated stronger thrombosis enhancement in human plasma clots in vitro (P<0.001 versus C15 clots and other controls). A15 was found to be highly specific for the active site of FXIIIa and was covalently bound to fibrin. In vivo murine experiments with A15 demonstrated significant signal enhancement in acute intravascular thrombi (P<0.05 versus C15 group). Minimal A15 enhancement was seen in older aged thrombi (>24 hours), consistent with an expected decline of FXIIIa activity over time. Imaging results were confirmed on correlative histopathology and fluorescence microscopy. CONCLUSIONS: A15 is a novel optical imaging agent that is specifically crosslinked to fibrin by FXIIIa, permitting detection of FXIIIa activity in experimental thrombi in vivo. This agent should permit assessment of FXIIIa activity in a broad range of biological processes and could aid in the clinical diagnosis of acute thrombi.


Subject(s)
Contrast Media/pharmacokinetics , Factor XIIIa/analysis , Fibrin/drug effects , Fluorescent Dyes/pharmacokinetics , Peptide Fragments/pharmacokinetics , Spectroscopy, Near-Infrared , Thrombosis/metabolism , alpha-2-Antiplasmin/pharmacokinetics , Animals , Chlorides , Cross-Linking Reagents/pharmacology , Feasibility Studies , Ferric Compounds/toxicity , Fibrin/analysis , Humans , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Protein Binding , Thrombosis/chemically induced , Time Factors
18.
J Biol Chem ; 278(45): 44369-76, 2003 Nov 07.
Article in English | MEDLINE | ID: mdl-12930825

ABSTRACT

Phosphorylation of SNARE proteins may provide a critical link between cell activation and secretory processes. Platelets contain all three members of the SNAP-23/25/29 gene family, but by comparison to brain tissue, SNAP-23 is the most highly enriched of these proteins in platelets. SNAP-23 function is required for exocytosis from platelet alpha, dense, and lysosomal granules. SNAP-23 was phosphorylated largely on serine residues in platelets activated with thrombin. Phosphorylation kinetics paralleled or preceded granule secretion. Inhibition studies suggested that SNAP-23 phosphorylation proceeds largely through a protein kinase C (PKC) mechanism and purified PKC directly phosphorylated recombinant (r-) SNAP-23 (up to 0.3 mol of phosphate/mol of protein). Five major tryptic phosphopeptides were identified in cellular SNAP-23 isolated from activated platelets; three phosphopeptides co-migrated with those identified in PKC-phosphorylated r-SNAP-23. In contrast, only one major phosphopeptide was identified when SNAP-23, engaged in a ternary SNARE complex, was phosphorylated by PKC. Ion trap mass spectrometry revealed that platelet SNAP-23 was phosphorylated at Ser23/Thr24 and Ser161, after cell activation by thrombin; these sites were also identified in PKC-phosphorylated r-SNAP-23. SNAP-23 mutants that mimic phosphorylation at Ser23/Thr24 inhibited syntaxin 4 interactions, whereas a phosphorylation mutant of Ser161 had only minor effects. Taken together these studies show that SNAP-23 is phosphorylated in platelets during cell activation through a PKC-related mechanism at two or more sites with kinetics that parallel or precede granule secretion. Because mutants that mimic SNAP-23 phosphorylation affect syntaxin 4 interactions, we hypothesize that SNAP-23 phosphorylation may be important for modulating SNARE-complex interactions during membrane trafficking and fusion.


Subject(s)
Blood Platelets/metabolism , Carrier Proteins/blood , Platelet Activation , Vesicular Transport Proteins , Amino Acid Sequence , Binding Sites , Blood Platelets/ultrastructure , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cytoplasmic Granules/metabolism , Enzyme Inhibitors/pharmacology , Epoprostenol/pharmacology , Humans , Immunoblotting , Kinetics , Membrane Proteins/blood , Molecular Sequence Data , Mutagenesis , Nerve Tissue Proteins/blood , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Platelet Activation/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/blood , Qa-SNARE Proteins , Qb-SNARE Proteins , Qc-SNARE Proteins , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , SNARE Proteins , Serine/blood , Synaptosomal-Associated Protein 25 , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL