Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
iScience ; 27(2): 108802, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38318380

ABSTRACT

Inflammation is consistently linked to dysmetabolism. In transgenic mice (Def+/+) model the neutrophilic peptide, alpha defensin, proved atherogenic. This phenotype occurred despite favorable cholesterol and glucose levels, and lower body weight and blood pressure. In this study, integration of metabolic&behavioral phenotyping system, endocrine, biochemical and mitochondrial assessment, pathological and immunohistochemical tests, and multiple challenge tests was established to explore the metabolic impact of alpha defensin. Compared to the control group, Def+/+ mice exhibited lower total energy expenditure and carbohydrate utilization, and higher fat oxidation. Their ACTH-cortisol and thyroid profiles were intact. Intriguingly, they had low levels of glucagon, with high ammonia, uric acid, triglyceride, and lactate. Mitochondrial evaluations were normal. Overall, defensin-induced hypoglucagonemia is associated with lipolysis, restricted glucose oxidation, and enhanced wasting. Def+/+ mice may be a useful model for studying the category of lean, apparently metabolically healthy, and atherosclerotic phenotype, with insight into a potential inflammatory-metabolic link.

2.
Medicina (Kaunas) ; 59(5)2023 May 19.
Article in English | MEDLINE | ID: mdl-37241215

ABSTRACT

Background and Objectives: The neutrophilic peptide, alpha-defensin, is considered an evolving risk factor intimately linked with lipid mobilization. It was previously linked to augmented liver fibrosis. Here, we assess a potential association between alpha-defensin and fatty liver. Materials and Methods: A cohort of transgenic C57BL/6JDef+/+ male mice that overexpress the human neutrophil-derived alpha-defensin in their polymorphonuclear neutrophils (PMNs) were assessed for liver steatosis and fibrosis development. Wild type (C57BL/6JDef.Wt) and transgenic (C57BL/6JDef+/+) mice were maintained on a standard rodent chow diet for 8.5 months. At the termination of the experiment, systemic metabolic indices and hepatic immunological cell profiling were assessed. Results: The Def+/+ transgenic mice exhibited lower body and liver weights, lower serum fasting glucose and cholesterol, and significantly lower liver fat content. These results were associated with impaired liver lymphocytes count and function (lower CD8, NK cells, and killing marker CD107a). The metabolic cage demonstrated dominant fat utilization with a comparable food intake in the Def+/+ mice. Conclusions: Chronic physiological expression of alpha-defensin induces favorable blood metabolic profile, increased systemic lipolysis, and decreased hepatic fat accumulation. Further studies are needed to characterize the defensin net liver effect.


Subject(s)
Non-alcoholic Fatty Liver Disease , alpha-Defensins , Male , Humans , Mice , Animals , Non-alcoholic Fatty Liver Disease/complications , alpha-Defensins/metabolism , Lipolysis , Diet, High-Fat , Mice, Inbred C57BL , Liver/metabolism
3.
J Int Med Res ; 50(10): 3000605221127099, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36268757

ABSTRACT

OBJECTIVE: Inflammation is associated with atherogenesis. Although a higher neutrophil count is associated with the plaque burden, the role of neutrophil activation is unclear. Human neutrophil peptides 1-3 (HNP1-3) are a risk factor for atherogenesis in bench models and are elevated in human atheromas. This study aimed to examine the association between skin HNP1-3 deposition and the severity of coronary artery disease (CAD), including long-term outcomes. METHODS: HNP1-3 levels were immunohistochemically quantified in skin biopsies, which were prospectively taken from 599 consecutive patients before clinically indicated coronary angiography. Established cardiovascular risk factors and blood markers for atheroinflammation were obtained. CAD severity and the incidence of repeat revascularization and mortality at 48 months of follow-up were assessed in relation to HNP1-3 levels. RESULTS: The risk of CAD was independently associated with age and HNP1-3 in the entire cohort (F = 0.71 and F = 7.4, respectively). Additionally, HNP1-3 levels were significantly associated with myocardial necrosis (R = 0.26). At the follow-up, high HNP1-3 levels negatively affected mortality (19.54%) and recurrent revascularization (8.05%). CONCLUSION: HNP1-3 tissue deposition is positively associated with the severity of CAD, myonecrosis, and long-term sequelae. HNP1-3 levels may be suppressed using colchicine.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Plaque, Atherosclerotic , alpha-Defensins , Humans , Prospective Studies , Longitudinal Studies , Cohort Studies , Risk Factors , Phenotype , Colchicine
4.
Br J Haematol ; 196(4): 923-927, 2022 02.
Article in English | MEDLINE | ID: mdl-34622440

ABSTRACT

Patients who are severely affected by coronavirus disease 2019 (COVID-19) may develop a delayed onset 'cytokine storm', which includes an increase in interleukin-6 (IL-6). This may be followed by a pro-thrombotic state and increased D-dimers. It was anticipated that tocilizumab (TCZ), an anti-IL-6 receptor monoclonal antibody, would mitigate inflammation and coagulation in patients with COVID-19. However, clinical trials with TCZ have recorded an increase in D-dimer levels. In contrast to TCZ, colchicine reduced D-dimer levels in patients with COVID-19. To understand how the two anti-inflammatory agents have diverse effects on D-dimer levels, we present data from two clinical trials that we performed. In the first trial, TCZ was administered (8 mg/kg) to patients who had a positive polymerase chain reaction test for COVID-19. In the second trial, colchicine was given (0·5 mg twice a day). We found that TCZ significantly increased IL-6, α-Defensin (α-Def), a pro-thrombotic peptide, and D-dimers. In contrast, treatment with colchicine reduced α-Def and Di-dimer levels. In vitro studies show that IL-6 stimulated the release of α-Def from human neutrophils but in contrast to colchicine, TCZ did not inhibit the stimulatory effect of IL-6; raising the possibility that the increase in IL-6 in patients with COVID-19 treated with TCZ triggers the release of α-Def, which promotes pro-thrombotic events reflected in an increase in D-dimer levels.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , COVID-19 Drug Treatment , Colchicine/therapeutic use , Fibrin Fibrinogen Degradation Products/analysis , alpha-Defensins/immunology , Aged , Blood Coagulation/drug effects , COVID-19/blood , COVID-19/immunology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Female , Fibrin Fibrinogen Degradation Products/immunology , Humans , Interleukin-6/blood , Interleukin-6/immunology , Male , Middle Aged , Neutrophils/drug effects , Neutrophils/immunology
5.
Br J Haematol ; 194(1): 44-52, 2021 07.
Article in English | MEDLINE | ID: mdl-34053084

ABSTRACT

The inflammatory response to SARS/CoV-2 (COVID-19) infection may contribute to the risk of thromboembolic complications. α-Defensins, antimicrobial peptides released from activated neutrophils, are anti-fibrinolytic and prothrombotic in vitro and in mouse models. In this prospective study of 176 patients with COVID-19 infection, we found that plasma levels of α-defensins were elevated, tracked with disease progression/mortality or resolution and with plasma levels of interleukin-6 (IL-6) and D-dimers. Immunohistochemistry revealed intense deposition of α-defensins in lung vasculature and thrombi. IL-6 stimulated the release of α-defensins from neutrophils, thereby accelerating coagulation and inhibiting fibrinolysis in human blood, imitating the coagulation pattern in COVID-19 patients. The procoagulant effect of IL-6 was inhibited by colchicine, which blocks neutrophil degranulation. These studies describe a link between inflammation and the risk of thromboembolism, and they identify a potential new approach to mitigate this risk in patients with COVID-19 and potentially in other inflammatory prothrombotic conditions.


Subject(s)
COVID-19/metabolism , Inflammation/metabolism , Thromboembolism/prevention & control , alpha-Defensins/blood , Adult , Aged , Animals , Blood Coagulation/drug effects , COVID-19/complications , COVID-19/diagnosis , COVID-19/virology , Case-Control Studies , Colchicine/pharmacology , Female , Fibrin Fibrinogen Degradation Products/analysis , Humans , Inflammation/complications , Interleukin-6/blood , Interleukin-6/pharmacology , Male , Mice , Middle Aged , Models, Animal , Neutrophils/drug effects , Prospective Studies , Risk Factors , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Severity of Illness Index , Thromboembolism/etiology , Thrombosis/etiology , Thrombosis/metabolism , Tubulin Modulators/pharmacology , alpha-Defensins/pharmacology
7.
Front Pharmacol ; 11: 846, 2020.
Article in English | MEDLINE | ID: mdl-32595499

ABSTRACT

Malaria caused by Plasmodium falciparum causes numerous cases of morbidity with about 400,000 deaths yearly owing, mainly, to inflammation leading to cerebral malaria (CM). CM conventionally is treated by repetitive administration of anti-plasmodial drugs and supportive non-specific drugs, for about a week. A mouse model of CM caused by Plasmodium berghei ANKA, in which brain and systemic clinical pathologies occur followed by sudden death within about a week, was used to study the effect of artemisone, a relatively new artemisinin, within an injectable pasty polymer formulated for its controlled release. The parasites were exposed to the drug over several days at a non-toxic concentrations for the mice but high enough to affect the parasites. Artemisone was also tested in cultures of bacteria, cancer cells and P. falciparum to evaluate the specificity and suitability of these cells for examining the release of artemisone from its carrier. Cultures of P. falciparum were the most suitable. Artemisone released from subcutaneous injected poly(sebacic acid-ricinoleic acid) (PSARA) pasty polymer, reduced parasitemias in infected mice, prolonged survival and prevented death in most of the infected mice. Successful prophylactic treatment before infection proved that there was a slow release of the drug for about a week, which contrasts with the three hour half-life that occurs after injection of just the drug. Treatment with artemisone within the polymer, even at a late stage of the disease, helped to prevent or, at least, delay accompanying severe symptoms. In some cases, treatment prevented death of CM and the mice died later of anemia. Postponing the severe clinical symptoms is also beneficial in cases of human malaria, giving more time for an appropriate diagnosis and treatment before severe symptoms appear. The method presented here may also be useful for combination therapy of anti-plasmodial and immunomodulatory drugs.

8.
PLoS One ; 15(4): e0231582, 2020.
Article in English | MEDLINE | ID: mdl-32302327

ABSTRACT

Atherosclerosis, the predominant cause of death in well-resourced countries, may develop in the presence of plasma lipid levels within the normal range. Inflammation may contribute to lesion development in these individuals, but the underlying mechanisms are not well understood. Transgenic mice expressing α-def-1 released from activated neutrophils develop larger lipid and macrophage-rich lesions in the proximal aortae notwithstanding hypocholesterolemia caused by accelerated clearance of α-def-1/low-density lipoprotein (LDL) complexes from the plasma. The phenotype does not develop when the release of α-def-1 is prevented with colchicine. However, ApoE-/- mice crossed with α-def-1 mice or given exogenous α-def-1 develop smaller aortic lesions associated with reduced plasma cholesterol, suggesting a protective effect of accelerated LDL clearance. Experiments were performed to address this seeming paradox and to determine if α-def-1 might provide a means to lower cholesterol and thereby attenuate atherogenesis. We confirmed that exposing ApoE-/- mice to α-def-1 lowers total plasma cholesterol and decreases lesion size. However, lesion size was larger than in mice with total plasma cholesterol lowered to the same extent by inhibiting its adsorption or by ingesting a low-fat diet. Furthermore, α-def-1 levels correlated independently with lesion size in ApoE-/- mice. These studies show that α-def-1 has competing effects on atherogenesis. Although α-def-1 accelerates LDL clearance from plasma, it also stimulates deposition and retention of LDL in the vasculature, which may contribute to development of atherosclerosis in individuals with normal or even low plasma levels of cholesterol. Inhibiting α-def-1 may attenuate the impact of chronic inflammation on atherosclerotic vascular disease.


Subject(s)
Aorta/pathology , Atherosclerosis/pathology , Cholesterol/blood , alpha-Defensins/metabolism , Animals , Anticholesteremic Agents/administration & dosage , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cholesterol/metabolism , Cholestyramine Resin/administration & dosage , Colchicine/administration & dosage , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Humans , Lipid Metabolism/drug effects , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Mice , Mice, Knockout, ApoE , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/metabolism , alpha-Defensins/genetics
9.
Transl Stroke Res ; 10(1): 104-111, 2019 02.
Article in English | MEDLINE | ID: mdl-29476447

ABSTRACT

The sole FDA-approved drug treatment for ischemic stroke is tissue-type plasminogen activator (tPA). However, upregulation of JNK mitogen-activated protein kinase (MAPK) and endothelin 1 (ET-1) by tPA after stroke contributes to impaired cerebrovascular autoregulation. Wild-type (wt) tPA can bind to the lipoprotein-related receptor (LRP), which mediates vasodilation, or NMDA receptors (NMDA-Rs), exacerbating vasoconstriction. Elevations in IL-6, a marker of inflammation that accompanies stroke, are reported to be an adverse prognostic factor. We hypothesized that IL-6 released into CSF after stroke by wt-tPA through activation of NMDA-Rs and upregulation of ET-1 and JNK contribute to impairment of cerebrovascular autoregulation and increased histopathology. Results show that IL-6 was increased post stroke in pigs, which was increased further by wt-tPA. Co-administration of the IL-6 antagonist LMT-28 with wt-tPA prevented impairment of cerebrovascular autoregulation and necrosis of hippocampal cells. wt-tPA co-administered with the JNK inhibitor SP 600125 and the ET-1 antagonist BQ 123 blocked stroke-induced elevation of IL-6. Co-administration of LMT-28 with wt-tPA blocked the augmentation of JNK and ET-1 post stroke. In conclusion, IL-6 released after stroke, which is enhanced by wt-tPA through activation of NMDA-Rs and upregulation of ET-1 and JNK, impairs cerebrovascular autoregulation and increases histopathology. Strategies that promote fibrinolysis while limiting activation of NMDA-Rs and upregulation of IL-6 may improve the benefit/risk ratio compared to wt-tPA in treatment of stroke.


Subject(s)
Cerebral Cortex/physiopathology , Hippocampus/pathology , Homeostasis/physiology , Interleukin-6/metabolism , Stroke , Animals , Anthracenes/therapeutic use , Disease Models, Animal , Endothelin-1 , Necrosis/etiology , Oxazolidinones/therapeutic use , Random Allocation , Receptors, N-Methyl-D-Aspartate , Signal Transduction , Stroke/complications , Stroke/drug therapy , Stroke/metabolism , Stroke/pathology , Swine , Tissue Plasminogen Activator/therapeutic use , Up-Regulation
10.
Blood ; 133(5): 481-493, 2019 01 31.
Article in English | MEDLINE | ID: mdl-30442678

ABSTRACT

Inflammation and thrombosis are integrated, mutually reinforcing processes, but the interregulatory mechanisms are incompletely defined. Here, we examined the contribution of α-defensins (α-defs), antimicrobial proteins released from activated human neutrophils, on clot formation in vitro and in vivo. Activation of the intrinsic pathway of coagulation stimulates release of α-defs from neutrophils. α-Defs accelerate fibrin polymerization, increase fiber density and branching, incorporate into nascent fibrin clots, and impede fibrinolysis in vitro. Transgenic mice (Def++) expressing human α-Def-1 developed larger, occlusive, neutrophil-rich clots after partial inferior vena cava (IVC) ligation than those that formed in wild-type (WT) mice. IVC thrombi extracted from Def++ mice were composed of a fibrin meshwork that was denser and contained a higher proportion of tightly packed compressed polyhedral erythrocytes than those that developed in WT mice. Def++ mice were resistant to thromboprophylaxis with heparin. Inhibiting activation of the intrinsic pathway of coagulation, bone marrow transplantation from WT mice or provision of colchicine to Def++ mice to inhibit neutrophil degranulation decreased plasma levels of α-defs, caused a phenotypic reversion characterized by smaller thrombi comparable to those formed in WT mice, and restored responsiveness to heparin. These data identify α-defs as a potentially important and tractable link between innate immunity and thrombosis.


Subject(s)
Fibrin/immunology , Neutrophil Activation , Thrombosis/immunology , alpha-Defensins/immunology , Animals , Blood Coagulation , Fibrin/analysis , Fibrinolysis , Humans , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Kallikreins/blood , Kallikreins/immunology , Male , Mice , Protein Conformation , Protein Stability , Thrombosis/blood , Thrombosis/pathology , alpha-Defensins/blood
11.
J Neurosci Res ; 96(1): 128-137, 2018 01.
Article in English | MEDLINE | ID: mdl-28703856

ABSTRACT

Tissue-type plasminogen activator (tPA) is neurotoxic and exacerbates uncoupling of cerebral blood flow (CBF) and metabolism after stroke, yet it remains the sole FDA-approved drug for treatment of ischemic stroke. Upregulation of c-Jun-terminal kinase (JNK) after stroke contributes to tPA-mediated impairment of autoregulation, but the role of endothelin-1 (ET-1) is unknown. Based on the Glasgow Coma Scale, impaired autoregulation is linked to adverse outcomes after TBI, but correlation with hippocampal histopathology after stroke has not been established. We propose that given after stroke, tPA activates N-Methyl-D-Aspartate receptors (NMDA-Rs) and upregulates ET-1 in a JNK dependent manner, imparing autoregulation and leading to histopathology. After stroke, CBF was reduced in the hippocampus and reduced further during hypotension, which did not occur in hypotensive sham pigs, indicating impairment of autoregulation. Autoregulation and necrosis of hippocampal CA1 and CA3 neurons were further impaired by tPA, but were preserved by the ET-1 antagonist BQ 123 and tPA-A,296-299 a variant that is fibrinolytic but does not bind to NMDA-Rs. Expression of ET-1 was increased by stroke and potentiated by tPA but returned to sham levels by tPA-A296-299 and the JNK antagonist SP600125. Results show that JNK releases ET-1 after stroke. Tissue-type plasminogen activator -A296-299 prevents impairment of cerebral autoregulation and histopathology after stroke by inhibiting upregulation of ET-1.


Subject(s)
Cerebrovascular Circulation/physiology , Endothelin-1/antagonists & inhibitors , Hippocampus/metabolism , Homeostasis/physiology , Stroke/metabolism , Tissue Plasminogen Activator/therapeutic use , Animals , Cerebrovascular Circulation/drug effects , Endothelin-1/biosynthesis , Female , Hippocampus/drug effects , Hippocampus/pathology , Homeostasis/drug effects , Male , Necrosis , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Stroke/drug therapy , Stroke/pathology , Swine , Tissue Plasminogen Activator/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
12.
Stroke ; 47(8): 2096-102, 2016 08.
Article in English | MEDLINE | ID: mdl-27354223

ABSTRACT

BACKGROUND AND PURPOSE: The sole Food and Drug Administration-approved treatment for stroke is tissue-type plasminogen activator (tPA), but its brief therapeutic window and complications of treatment constrain its use. One limitation may be its potential to exacerbate impairment of cerebral autoregulation after stroke. Vasodilation is maintained by elevations in cAMP. However, cAMP levels fall after stroke because of overactivation of N-methyl-d-aspartate receptors by toxic levels of glutamate, an effect that is exacerbated by tPA. Binding of wild-type (wt) tPA to the low-density lipoprotein-related receptor (LRP) mediates dilation. We propose that binding of wt-tPA to N-methyl-d-aspartate receptor reduces cAMP and impairs vasodilation. We hypothesize that tPA-A(296-299), a variant that is fibrinolytic but cannot bind to N-methyl-d-aspartate receptor, preferentially binds to LRP and increases cAMP and p38, limiting autoregulation impairment after stroke. METHODS: Stroke was induced by photothrombosis in pigs equipped with a closed cranial window, cerebral blood flow determined by microspheres, and cerebrospinal fluid cAMP and p38 determined by ELISA. RESULTS: Stroke decreased cerebral blood flow. Cerebral blood flow was reduced further during hypotension, indicating impairment of autoregulation. Autoregulation was further impaired by wt-tPA, which was prevented by MK801 and tPA-A(296-299). Protection by tPA-A(296-299) was blocked by anti-LRP Ab, the LRP antagonist receptor-associated protein, and the p38 inhibitor SB 203580, but not by control IgG. Stroke reduced cerebrospinal fluid cAMP, which was reduced further by wt-tPA, but augmented by tPA-A(296-299). Cerebrospinal fluid p38 was unchanged by wt-tPA, increased by tPA-A(296-299), and decreased by anti-LRP Ab and receptor-associated protein. CONCLUSIONS: tPA-A(296-299) prevents impairment of cerebral autoregulation after stroke through an LRP-dependent increase in cAMP and p38.


Subject(s)
Cerebral Cortex/drug effects , Cyclic AMP/metabolism , Homeostasis/drug effects , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cerebral Cortex/metabolism , Cerebrovascular Circulation/drug effects , Female , Male , Receptors, N-Methyl-D-Aspartate/metabolism , Stroke/metabolism , Swine , Thrombolytic Therapy , Tissue Plasminogen Activator/pharmacology
13.
J Biol Chem ; 291(6): 2777-86, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26518877

ABSTRACT

Approximately one-half of the patients who develop clinical atherosclerosis have normal or only modest elevations in plasma lipids, indicating that additional mechanisms contribute to pathogenesis. In view of increasing evidence that inflammation contributes to atherogenesis, we studied the effect of human neutrophil α-defensins on low density lipoprotein (LDL) trafficking, metabolism, vascular deposition, and atherogenesis using transgenic mice expressing human α-defensins in their polymorphonuclear leukocytes (Def(+/+)). Accelerated Def(+/+) mice developed α-defensin·LDL complexes that accelerate the clearance of LDL from the circulation accompanied by enhanced vascular deposition and retention of LDL, induction of endothelial cathepsins, increased endothelial permeability to LDL, and the development of lipid streaks in the aortic roots when fed a regular diet and at normal plasma levels of LDL. Transplantation of bone marrow from Def(+/+) to WT mice increased LDL clearance, increased vascular permeability, and increased vascular deposition of LDL, whereas transplantation of WT bone marrow to Def(+/+) mice prevented these outcomes. The same outcome was obtained by treating Def(+/+) mice with colchicine to inhibit the release of α-defensins. These studies identify a potential new link between inflammation and the development of atherosclerosis.


Subject(s)
Atherosclerosis/blood , Cholesterol/blood , Endothelial Cells/metabolism , Lipoproteins, LDL/blood , Protein Processing, Post-Translational , alpha-Defensins/blood , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Cathepsins/blood , Cathepsins/genetics , Cholesterol/genetics , Colchicine/pharmacology , Endothelial Cells/pathology , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/pathology , Lipoproteins, LDL/genetics , Male , Mice , Mice, Transgenic , Multiprotein Complexes/blood , Multiprotein Complexes/genetics , alpha-Defensins/genetics
14.
Blood ; 125(16): 2558-67, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25673638

ABSTRACT

Persistent intracerebral hemorrhage (ICH) is a major cause of death and disability after traumatic brain injury (TBI) for which no medical treatment is available. Delayed bleeding is often ascribed to consumptive coagulopathy initiated by exposed brain tissue factor. We examined an alternative hypothesis, namely, that marked release of tissue-type plasminogen activator (tPA) followed by delayed synthesis and release of urokinase plasminogen activator (uPA) from injured brain leads to posttraumatic bleeding by causing premature clot lysis. Using a murine model of severe TBI, we found that ICH is reduced in tPA(-/-) and uPA(-/-) mice but increased in PAI-1(-/-) mice compared with wild-type (WT) mice. tPA(-/-), but not uPA(-/-), mice developed a systemic coagulopathy post-TBI. Tranexamic acid inhibited ICH expansion in uPA(-/-)mice but not in tPA(-/-) mice. Catalytically inactive tPA-S(481)A inhibited plasminogen activation by tPA and uPA, attenuated ICH, lowered plasma d-dimers, lessened thrombocytopenia, and improved neurologic outcome in WT, tPA(-/-), and uPA(-/-) mice. ICH expansion was also inhibited by tPA-S(481)A in WT mice anticoagulated with warfarin. These data demonstrate that protracted endogenous fibrinolysis induced by TBI is primarily responsible for persistent ICH and post-TBI coagulopathy in this model and offer a novel approach to interrupt bleeding.


Subject(s)
Brain Injuries/complications , Cerebral Hemorrhage/metabolism , Tissue Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Antifibrinolytic Agents/pharmacology , Brain/blood supply , Brain/metabolism , Brain Injuries/blood , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/genetics , Fibrin/metabolism , Fibrinolysis/drug effects , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Plasminogen/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Time Factors , Tissue Plasminogen Activator/genetics , Tranexamic Acid/pharmacology , Urokinase-Type Plasminogen Activator/genetics
15.
Neurocrit Care ; 20(1): 111-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24248736

ABSTRACT

BACKGROUND: Babies are frequently exposed to cerebral hypoxia and ischemia (H/I) during the perinatal period as a result of stroke, problems with delivery or post delivery respiratory management. The sole FDA approved treatment for acute stroke is tissue-type plasminogen activator (tPA). Endogenous tPA is upregulated and potentiates impairment of pial artery dilation in response to hypotension after H/I in pigs. Mitogen-activated protein kinase (MAPK), a family of at least 3 kinases, ERK, p38 and JNK, is also upregulated after H/I, with ERK contributing to impaired vasodilation. This study examined the hypothesis that H/I aggravates the vascular response to two important procontractile mediators released during CNS ischemia, endothelin-1 (ET-1) and thromboxane, which is further enhanced by tPA and ERK MAPK. METHODS: Cerebral hypoxia (pO(2) 35 mmHg for 10 min via inhalation of N(2)) followed immediately by ischemia (global intracranial pressure elevation for 20 min) was produced in chloralose anesthetized piglets equipped with a closed cranial window. RESULTS: H/I aggravated pial artery vasconstriction induced by ET-1 and the thromboxane mimetic U 46619. Potentiated vasoconstrictor responses were blocked by EEIIMD, an inhibitor of tPA's signaling and vascular activities, but unchanged by its inactive analogue EEIIMR. The cerebrospinal fluid concentration of ERK MAPK determined by ELISA was increased by H/I, potentiated by tPA, but blocked by EEIIMD. The ERK MAPK antagonist U 0126 blocked H/I augmented enhancement of ET-1 and U 46619 vasoconstriction. CONCLUSIONS: These data indicate that H/I aggravates ET-1 and thromboxane mediated cerebral vasoconstriction by upregulating endogenous tPA and ERK MAPK.


Subject(s)
Brain/blood supply , Cerebrovascular Circulation/drug effects , Endothelin-1/pharmacology , Hypoxia-Ischemia, Brain/drug therapy , Oligopeptides/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Thromboxanes/pharmacology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Animals , Animals, Newborn , Brain/drug effects , Craniotomy/methods , Disease Models, Animal , Female , Male , Pia Mater/blood supply , Swine , Translational Research, Biomedical
16.
J Neurotrauma ; 30(22): 1898-907, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23731391

ABSTRACT

Traumatic brain injury (TBI) is associated with loss of cerebrovascular autoregulation, which leads to cerebral hypoperfusion. Mitogen activated protein kinase (MAPK) isoforms ERK, p38, and JNK and endothelin-1 (ET-1) are mediators of impaired cerebral hemodynamics after TBI. Excessive tissue plasminogen activator (tPA) released after TBI may cause loss of cerebrovascular autoregulation either by over-activating N-methyl-D-aspartate receptors (NMDA-Rs) or by predisposing to intracranial hemorrhage. Our recent work shows that a catalytically inactive tPA variant (tPA-S(481)A) that competes with endogenous wild type (wt) tPA for binding to NMDA-R through its receptor docking site but that cannot activate it, prevents activation of ERK by wt tPA and impairment of autoregulation when administered 30 min after fluid percussion injury (FPI). We investigated the ability of variants that lack proteolytic activity but bind/block activation of NMDA-Rs by wt tPA (tPA-S(481)A), do not bind/block activation of NMDA-Rs but are proteolytic (tPA-A(296-299)), or neither bind/block NMDA-Rs nor are proteolytic (tPA-A(296-299)S(481)A) to prevent impairment of autoregulation after TBI and the role of MAPK and ET-1 in such effects. Results show that tPA-S(481)A given 3 h post-TBI, but not tPA-A(296-299) or tPA-A(296-299)S(481)A prevents impaired autoregulation by upregulating p38 and inhibiting ET-1, suggesting that tPA-S(481)A has a realistic therapeutic window and focuses intervention on NMDA-Rs to improve outcome.


Subject(s)
Brain Injuries/drug therapy , Endothelin-1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Tissue Plasminogen Activator/pharmacology , Up-Regulation , p38 Mitogen-Activated Protein Kinases/biosynthesis , Animals , Animals, Newborn , Brain Injuries/etiology , Brain Injuries/metabolism , Disease Models, Animal , Endothelin-1/metabolism , Female , Male , Swine , Time Factors , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/metabolism , Up-Regulation/drug effects
18.
Neurol Res ; 34(6): 530-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22642975

ABSTRACT

OBJECTIVE: Outcome of traumatic brain injury (TBI) is impaired by hypotension and glutamate, and TBI-associated release of endogenous tissue plasminogen activator (tPA) impairs cerebral autoregulation. Glucagon decreases central nervous system glutamate, lessens neuronal cell injury, and improves neurological score in mice after TBI. Glucagon partially protects against impaired cerebrovasodilation during hypotension after TBI in piglets by upregulating cAMP which decreases release of tPA. Pial artery dilation during hypotension is due to release of cAMP-dependent dilator prostaglandins (PG), such as PGE2 and PGI2. TBI impairs PGE2 and PGI2-mediated pial artery dilation, which contributes to disturbed cerebral autoregulation post-insult, by upregulating mitogen-activated protein kinase (MAPK). This study was designed to investigate relationships between tPA, prostaglandins, and MAPK as a mechanism to improve the efficacy of glucagon-mediated preservation of cerebrovasodilation during hypotension after TBI. METHODS: Lateral fluid percussion brain injury (FPI) was induced in piglets equipped with a closed cranial window. ERK and JNK MAPK concentrations in cerebrospinal fluid were quantified by enzyme-linked immunosorbent assay. RESULTS: Cerebrospinal fluid JNK MAPK was increased by FPI, but blunted by glucagon and the novel plasminogen activator inhibitor-1-derived peptide (PAI-1DP), Ac-RMAPEEIIMDRPFLYVVR-amide. FPI modestly increased, while glucagon and PAI-1DP decreased ERK MAPK. PGE2, PGI2, N-methyl-D-aspartate, and hypotension-induced pial artery dilation was blunted after FPI, partially protected by glucagon, and fully protected by glucagon+PAI-1DP, glucagon+JNK antagonist SP600125 or glucagon+ERK inhibitor U 0126. DISCUSSION: Glucagon+PAI-1DP act in concert to protect against impairment of cerebrovasodilation during hypotension after TBI via inhibition of ERK and JNK MAPK.


Subject(s)
Brain Injuries/drug therapy , Cerebrovascular Circulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucagon/pharmacology , MAP Kinase Kinase 4/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Animals , Brain Injuries/complications , Brain Injuries/metabolism , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Hypotension/etiology , Hypotension/prevention & control , Peptides , Swine , Vasodilation/drug effects
19.
J Neurotrauma ; 29(9): 1794-802, 2012 Jun 10.
Article in English | MEDLINE | ID: mdl-22435890

ABSTRACT

Traumatic brain injury (TBI) is associated with loss of autoregulation due to impaired responsiveness to cerebrovascular dilator stimuli, which leads to cerebral hypoperfusion and neuronal impairment or death. Upregulation of tissue plasminogen activator (tPA) post-TBI exacerbates loss of cerebral autoregulation and NMDA-receptor-mediated impairment of cerebral hemodynamics, and enhances excitotoxic neuronal death. However, the relationship between NMDA-receptor activation, loss of autoregulation, and neurological dysfunction is unclear. Here, we evaluated the potential therapeutic efficacy of a catalytically inactive tPA variant, tPA S481A, that acts by competing with wild-type tPA for binding, cleavage, and activation of NMDA receptors. Lateral fluid percussion brain injury was produced in anesthetized piglets. Pial artery reactivity was measured via a closed cranial window, and cerebrospinal fluid (CSF) extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) was quantified by enzyme-linked immunosorbent assay (ELISA). tPA-S481A prevented impairment of cerebral autoregulation and reduced histopathologic changes after TBI by inhibiting upregulation of the ERK isoform of MAPK. Treatment with this tPA variant provides a novel approach for limiting neuronal toxicity caused by untoward NMDA-receptor activation mediated by increased tPA and glutamate following TBI.


Subject(s)
Brain Injuries/drug therapy , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/toxicity , Tissue Plasminogen Activator/therapeutic use , Animals , Animals, Newborn , Apoptosis/drug effects , Brain Injuries/cerebrospinal fluid , Brain Injuries/pathology , CA1 Region, Hippocampal/pathology , CA3 Region, Hippocampal/pathology , Cerebral Arteries/pathology , Cerebrovascular Circulation/physiology , Enzyme-Linked Immunosorbent Assay , Female , Glutamic Acid/metabolism , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Necrosis/pathology , Polymerase Chain Reaction , Prostaglandins/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Swine , Tissue Plasminogen Activator/cerebrospinal fluid
20.
Transl Stroke Res ; 3(1): 114-21, 2012 Mar.
Article in English | MEDLINE | ID: mdl-23577046

ABSTRACT

The sole Food and Drug Administration-approved treatment for acute stroke is tissue-type plasminogen activator (tPA), but tPA aggravates impairment of cerebrovasodilation during hypotension in a newborn pig photothrombotic model of stroke. Coupling to carrier red blood cells (RBC) enhances thrombolytic effects of tPA, while reducing its side effects. ATP- and Ca-sensitive K channels (Katp and Kca) are important regulators of cerebrovascular tone and mediate cerebrovasodilation during hypotension. Mitogen-activated protein kinase, a family of at least three kinases, ERK, p38, and c-Jun-N-terminal kinase (JNK), is upregulated after photothrombosis. This study examined the effect of photothrombosis on Katp- and Kca-induced cerebrovasodilation and the roles of tPA and JNK during/after injury. Photothrombosis blunted vasodilation induced by the Katp agonists cromakalim, calcitonin gene-related peptide, and the Kca agonist NS 1619, which was aggravated by injection of tPA. In contrast, both pre- or post-injury thrombosis injection of RBC-tPA and JNK antagonist SP 600125 prevented impairment of Katp- and Kca-induced vasodilation. Therefore, JNK activation in thrombosis impairs K channel-mediated cerebrovasodilation. Standard thrombolytic therapy of central nervous system ischemic disorders using free tPA poses the danger of further dysregulation of cerebrohemodynamics by impairing cation-mediated control of cerebrovascular tone, whereas RBC-coupled tPA both restores reperfusion and normalizes cerebral hemodynamics.

SELECTION OF CITATIONS
SEARCH DETAIL
...