Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(5): e0285499, 2023.
Article in English | MEDLINE | ID: mdl-37235594

ABSTRACT

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


Subject(s)
Atherosclerosis , Brain Injuries, Traumatic , Animals , Mice , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Atherosclerosis/pathology , Blood Pressure , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Disease Models, Animal , Metoprolol/pharmacology , Metoprolol/therapeutic use , Mice, Inbred C57BL
2.
Sci Rep ; 12(1): 16437, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36180774

ABSTRACT

Sickle cell disease (SCD) is associated with altered plasma and erythrocyte lipid profiles. In a previous study, SCD mice with deficiency of proprotein convertase subtilisin/kexin type 9 (PCSK9) were observed to have more severe anemia and increased sickling compared to control SCD mice. Although PCSK9 affects circulating low density lipoprotein (LDL) by regulation of the LDL receptor, the effect of PCSK9 on anemia was independent of LDL receptor expression. In the current study, erythrocyte metabolomics were performed and revealed altered erythrocyte lipid species between SCD mice with and without PCSK9. Of particular interest, the late endosome-specific lipid bis(mono)acylglycerol phosphate (BMP) 44:12 was markedly decreased in erythrocytes from SCD mice deficient in PCSK9 mice relative to control SCD mice. Incubation of sickle erythrocytes with a neutralizing antibody to BMP increased erythrocyte sickling in vitro. In vitro treatment of SCD erythrocytes with amiodarone (1.5 µM) or medroxyprogesterone (6.75 µM), two pharmacologic compounds known to increase BMP, resulted in reduced erythrocyte sickling. Treatment of SCD mice with amiodarone (10 mg/kg) for 2 weeks resulted in increased BMP, improvement in anemia with reduced reticulocytosis, and decreased ex vivo sickling. In conclusion, severity of anemia in SCD is improved with amiodarone treatment, an effect which may be mediated through increased erythrocyte BMP.


Subject(s)
Amiodarone , Anemia, Sickle Cell , Amiodarone/pharmacology , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Animals , Antibodies, Neutralizing/pharmacology , Disease Models, Animal , Erythrocytes/metabolism , Lipoproteins, LDL/metabolism , Medroxyprogesterone/pharmacology , Mice , Monoglycerides/metabolism , Phosphates/metabolism , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism , Subtilisins/metabolism
3.
Br J Haematol ; 196(4): 1040-1051, 2022 02.
Article in English | MEDLINE | ID: mdl-34786709

ABSTRACT

Sickle cell disease (SCD) is associated with haemolytic anaemia and secondary activation of leucocytes and platelets, which in turn may further exacerbate haemolysis. As cytokine signalling pathways may participate in this cycle, the present study investigated whether pharmacological blockade of the interleukin-1 receptor (IL-1R) would mitigate anaemia in a murine model of SCD. Within 2 weeks of treatment, reduced markers of haemolysis were observed in anakinra-treated mice compared to vehicle-treated mice. After 4 weeks of anakinra treatment, mice showed increased numbers of erythrocytes, haemoglobin, and haematocrit, along with reduced reticulocytes. Blood from anakinra-treated mice was less susceptible to ex vivo erythrocyte sickling and was resistant to exogenous IL-1ß-mediated sickling. Supernatant generated from IL-1ß-treated platelets was sufficient to promote erythrocyte sickling, an effect not observed with platelet supernatant generated from IL-1R-/- mice. The sickling effect of IL-1ß-treated platelet supernatant was inhibited by a transforming growth factor-ß (TGF-ß) neutralising antibody, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, and superoxide scavengers, but replicated by recombinant TGF-ß. In conclusion, pharmacological IL-1R antagonism leads to improved anaemia in a murine SCD model. IL-1ß stimulation of platelets promotes erythrocyte sickling. This effect may be mediated by platelet-derived TGF-ß-induced reactive oxygen species generation though erythrocyte NADPH oxidase.


Subject(s)
Anemia, Sickle Cell/genetics , Anemia/physiopathology , Blood Platelets/metabolism , Receptors, Interleukin-1/genetics , Animals , Disease Models, Animal , Erythrocytes, Abnormal , Humans , Mice
4.
Haematologica ; 106(9): 2469-2477, 2021 09 01.
Article in English | MEDLINE | ID: mdl-32817286

ABSTRACT

Sickle cell disease (SCD) is associated with chronic hemolytic anemia and a heightened inflammatory state. The causal role of inflammatory pathways in stroke associated with SCD is unclear. Therefore, the hypothesis that deletion of the non-hematopoietic interleukin-1 receptor (IL-1R) pool may be beneficial in SCD was pursued. Since potential deleterious effects of IL-1R signaling in SCD could be mediated via downstream production of interleukin-6 (IL-6), the role of the non-hematopoietic IL-6 pool was also addressed. Bone marrow transplantation (BMT) from SCD to wild-type (WT) recipient mice was used to generate SCD mice (Wt,SCDbmt). To generate mice with non-hematopoietic deficiency of IL-1R or IL-6, SCD marrow was transplanted into IL-1R deficient (IL1R-/-,SCDbmt) or IL-6 deficient recipients (IL6-/-,SCDbmt). Blood counts, reticulocytes, soluble E-selectin (Esel), and IL-6 levels were analyzed 14-15 weeks post-BMT. Ischemic stroke was induced by middle cerebral artery (MCA) photothrombosis at 16 weeks post-BMT. A separate group of Wt,SCDbmt mice was given the IL-1R inhibitor, anakinra, following stroke induction. Seventy-two hours after MCA occlusion, stroke volume was assessed by staining brain sections with 2,3,5-triphenyltetrazolium chloride. Formalin-fixed brain sections were also stained for macrophages with MAC3, for endothelial activation with ICAM-1, and for loss of blood brain barrier (BBB) integrity with fibrin(ogen) staining. All SCD mice generated by BMT were anemic and the severity of anemia was not different between Wt,SCDbmt, IL1R-/-,SCDbmt, and IL6-/-,SCDbmt mice. Three days following MCA occlusion, stroke volume was significantly reduced in IL1R-/-,SCDbmt mice compared to Wt,SCDbmt mice and IL6-/-,SCDbmt mice. Plasma levels of sE-sel were lower in IL1R-/-,SCDbmt compared to Wt,SCDbmt and IL-6-/-,SCDbmt mice. Post-stroke treatment of Wt,SCDbmt mice with anakinra decreased stroke size, leukocyte infiltration, ICAM-1 expression, and fibrin(ogen) accumulation compared to vehicle-treated mice. Deficiency of non-hematopoietic IL-1R or treatment with an IL-1R antagonist is sufficient to confer protection against the increased stroke size associated with SCD. These effects of IL1R deficiency are associated with reduced endothelial activation, leukocyte infiltration, and blood brain barrier disruption, and are independent of non-hematopoietic IL-6 signaling.


Subject(s)
Anemia, Sickle Cell , Receptors, Interleukin-1 , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Disease Models, Animal , Interleukin 1 Receptor Antagonist Protein , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
Arterioscler Thromb Vasc Biol ; 40(6): 1533-1542, 2020 06.
Article in English | MEDLINE | ID: mdl-32268786

ABSTRACT

OBJECTIVE: Clopidogrel is a commonly used P2Y12 inhibitor to treat and prevent arterial thrombotic events. Clopidogrel is a prodrug that requires bioactivation by CYP (cytochrome P450) enzymes to exert antiplatelet activity. Diabetes mellitus is associated with an increased risk of ischemic events, and impaired ability to generate the active metabolite (AM) from clopidogrel. The objective of this study is to identify the mechanism of clopidogrel resistance in a murine model of diet-induced obesity (DIO). Approach and Results: C57BL/6J mice and IL-1R-/- mice were given high-fat diet for 10 weeks to generate a murine model of diet-induced obesity. Platelet aggregation and carotid arterial thrombosis were assessed in response to clopidogrel treatment. Wild-type DIO mice exhibited resistance to antiplatelet and antithrombotic effects of clopidogrel that was associated with reduced hepatic expression of CYP genes and reduced generation of the AM. IL (Interleukin)-1 receptor-deficient DIO (IL1R-/- DIO) mice showed no resistance to clopidogrel. Lack of resistance was accompanied by increased exposure of the clopidogrel AM. This resistance was also absent when wild-type DIO mice were treated with the conjugate of the clopidogrel AM, DT-678. CONCLUSIONS: These findings indicate that antiplatelet effects of clopidogrel may be impaired in the setting of diabetes mellitus due to reduced prodrug bioactivation related to IL-1 receptor signaling. Therapeutic targeting of P2Y12 in patients with diabetes mellitus using the conjugate of clopidogrel AM may lead to improved outcomes.


Subject(s)
Clopidogrel/pharmacokinetics , Clopidogrel/therapeutic use , Drug Resistance , Obesity/complications , Receptors, Interleukin-1/physiology , Animals , Carotid Artery Thrombosis/prevention & control , Clopidogrel/pharmacology , Cytochrome P-450 Enzyme System/genetics , Diabetes Mellitus , Diet, High-Fat , Disease Models, Animal , Fibrinolytic Agents , Gene Expression , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/enzymology , Obesity/etiology , Obesity/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Receptors, Interleukin-1/deficiency
6.
Sci Rep ; 8(1): 5639, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29618740

ABSTRACT

Traumatic brain injury (TBI) has been associated with atherosclerosis and cardiovascular mortality in humans. However the causal relationship between TBI and vascular disease is unclear. This study investigated the direct role of TBI on vascular disease using a murine model of atherosclerosis. Apolipoprotein E deficient mice were placed on a western diet beginning at 10 weeks of age. Induction of TBI or a sham operation was performed at 14 weeks of age and mice were sacrificed 6 weeks later at 20 weeks of age. MRI revealed evidence of uniform brain injury in all mice subjected to TBI. There were no differences in total cholesterol levels or blood pressure between the groups. Complete blood counts and flow cytometry analysis performed on peripheral blood 6 weeks following TBI revealed a higher percentage of Ly6C-high monocytes in mice subjected to TBI compared to sham-treated mice. Mice with TBI also showed elevated levels of plasma soluble E-selectin and bone marrow tyrosine hydroxylase. Analysis of atherosclerosis at the time of sacrifice revealed increased atherosclerosis with increased Ly6C/G immunostaining in TBI mice compared to sham-treated mice. In conclusion, progression of atherosclerosis is accelerated following TBI. Targeting inflammatory pathways in patients with TBI may reduce subsequent vascular complications.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/pathology , Brain Injuries, Traumatic/physiopathology , Animals , Blood Pressure Determination , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Sci Rep ; 8(1): 4881, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29559676

ABSTRACT

Endothelial dysfunction precedes atherosclerosis and may constitute a critical link between obesity-related inflammation and cardiovascular disease. Neutrophil extracellular traps (NETs) have been shown to promote vascular damage in murine models of autoimmune disease and atherosclerosis. The impact of NETs towards endothelial dysfunction associated with obesity is unknown. Using a diet-induced obesity (DIO) mouse model, this study investigated whether the inhibition or degradation of NETs could reduce the endothelial dysfunction observed in DIO mice. Following induction of DIO, there were elevated plasma concentrations of monocyte chemoattractant protein-1 (MCP-1) and impairment of mesenteric arteriolar vasorelaxation in response to acetylcholine as measured by pressure myography. A marker of NET formation, cathelicidin-related antimicrobial peptide (CRAMP), was markedly increased in mesenteric arterial walls of DIO mice compared to mice on standard chow. Prevention of NET formation with Cl-amidine or dissolution of NETs with DNase restored endothelium-dependent vasodilation to the mesenteric arteries of DIO mice. These findings suggest an instrumental role for NETs in obesity-induced endothelial dysfunction.


Subject(s)
Endothelium, Vascular/metabolism , Extracellular Traps/metabolism , Animals , Antimicrobial Cationic Peptides/analysis , Antimicrobial Cationic Peptides/blood , Cardiovascular Diseases/physiopathology , Chemokine CCL2/analysis , Chemokine CCL2/blood , Diet, High-Fat , Disease Models, Animal , Extracellular Traps/physiology , Inflammation/metabolism , Male , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/physiopathology , Vascular Diseases/physiopathology , Vasodilation , Cathelicidins
8.
Sci Rep ; 8(1): 3223, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459637

ABSTRACT

Identification of inflammatory mediators that regulate the vascular response to vasopressor molecules may aid in the development of novel therapeutic agents to treat or prevent hypertensive vascular diseases. Leukocytes have recently been shown to be capable of modifying blood pressure responses to vasopressor molecules. The purpose of this study was to test the hypothesis that deficiency of the leukocyte ligand, Psgl-1, would reduce the pressor response to angiotensin II (Ang II). Mice deficient in Psgl-1 (Psgl-1-/-) along with wild-type (WT) controls were treated for 2 weeks with a continuous infusion of Ang II. No differences in blood pressure between the groups were noted at baseline, however after 5 days of Ang II infusion, systolic blood pressures were higher in WT compared to Psgl-1-/- mice. The pressor response to acute administration of high dose Ang II was also attenuated in Psgl-1-/- compared to WT mice. Chimeric mice with hematopoietic deficiency of Psgl-1 similarly showed a reduced pressor response to Ang II. This effect was associated with reduced plasma interleukin-17 (IL-17) levels in Psgl-1-/- mice and the reduced pressor response was restored by administration of recombinant IL-17. In conclusion, hematopoietic deficiency of Psgl-1 attenuates Ang II-induced hypertension, an effect that may be mediated by reduced IL-17.


Subject(s)
Angiotensin II/metabolism , Hypertension/physiopathology , Membrane Glycoproteins/metabolism , Vasoconstrictor Agents/metabolism , Angiotensin II/administration & dosage , Animals , Blood Pressure/drug effects , Disease Models, Animal , Interleukin-17/metabolism , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout
9.
Sci Rep ; 7(1): 14360, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29084995

ABSTRACT

The effect of lipid lowering on the incidence of deep venous thrombosis (DVT) is controversial. The purpose of this study was to determine the effect of proprotein convertase subtilisin/kexin type 9 (PCSK9) deficiency on development of DVT in mice. Pcsk9 deficient (pcsk9 -/-) and wild-type (WT) littermates underwent partial inferior vena cava (IVC) ligation to induce venous thrombosis. 48 hours following IVC ligation, IVC thrombosis was evident in 60% of WT mice and 25% of pcsk9 -/- mice (p < 0.05). Analysis of IVC thrombosis revealed greater thrombus weight, length, myeloid cell recruitment, and more neutrophil extracellular trap formation (NETs) in WT compared to pcsk9 -/- mice. Intravital microscopy performed two hours following partial IVC ligation revealed that leukocyte firm attachment was increased in WT mice compared to mice undergoing a sham operation, however leukocyte attachment was reduced in pcsk9 -/- mice compared to WT mice. In conclusion, deficiency of PCSK9 is associated with protection from venous thrombosis. This protection is associated with reduced leukocyte recruitment and NET formation at the site of thrombosis.


Subject(s)
Proprotein Convertase 9/physiology , Venous Thrombosis/prevention & control , Animals , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Lipid Metabolism/physiology , Lipids/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proprotein Convertase 9/deficiency , Proprotein Convertase 9/metabolism , Proprotein Convertases , Serine Endopeptidases , Subtilisins , Venous Thrombosis/metabolism
10.
Sci Rep ; 7(1): 1606, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28487522

ABSTRACT

Some studies have shown that levels of MicroRNA (miR)-223 derived from platelets in the plasma are reduced following inhibition of platelet function, while others have shown a correlation between low plasma miR-223 and high on-treatment platelet reactivity. The present study seeks to investigate the role of miR-223 in arterial thrombosis. A model of photochemical-induced carotid thrombosis was applied to miR-223 deficient mice and littermate (WT) controls. Mice deficient in miR-223 exhibited significantly prolonged times to occlusive thrombosis compared to WT mice indicating a protective effect of miR-223 deficiency. Bone marrow transplantation experiments confirmed that the hematopoietic pool of miR-223 was responsible for differences in thrombosis times. Transfusion of either WT platelets or extracellular vesicles derived from WT platelets were both sufficient to shorten thrombosis times in miR-223 deficient recipients. The effect of platelet transfusions on IGF-1R was explored. These experiments revealed that vascular IGF-1R was down-regulated by platelet miR-223. Furthermore, inhibition of IGF-1R abolished the protection conferred by miR-223 deficiency on thrombosis. In conclusion, platelet miR-223 is a regulator of arterial thrombosis following endothelial injury through effects on vascular wall IGF-1R. This study indicates that platelet miR-223 is a potential therapeutic target for prevention of arterial thrombosis.


Subject(s)
Hematopoiesis , MicroRNAs/metabolism , Photochemical Processes , Thrombosis/genetics , Animals , Carotid Arteries/pathology , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Hematopoiesis/genetics , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Receptor, IGF Type 1/metabolism , Time Factors
11.
Sci Rep ; 6: 28997, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27357136

ABSTRACT

Systemic lupus erythematosus (SLE) is associated with an elevated risk of vascular complications, including premature stroke. Therapies targeting leukocyte recruitment may be beneficial in reducing vascular complications associated with SLE. Lupus was induced in female wild-type (WT) and P-selectin glycoprotein ligand-1 deficient (Psgl-1(-/-)) mice with pristane. Stroke was induced following photochemical injury to the middle cerebral artery (MCA). Stroke size was increased in pristane-treated WT mice compared to non-pristane-treated WT controls. However, stroke size was not increased in pristane-treated Psgl-1(-/-) mice compared to controls, despite evidence of increased nephritis in Psgl-1(-/-) mice. Pristane-treated WT mice showed elevated anti-dsDNA, anti-snRNP, CXCL1, and MCP-1 levels compared to untreated mice; however levels of anti-snRNP, MCP-1, and CXCL1 were reduced in pristane-treated Psgl-1(-/-) mice compared to pristane-treated WT mice. Infiltration of neutrophils and macrophages at the cerebral infarction site were reduced in pristane-treated Psgl-1(-/-) mice compared to pristane-treated WT mice. In conclusion, the increase in stroke size associated with lupus is prevented by Psgl-1 deficiency while nephritis is exacerbated. Therapies targeting Psgl-1 may be useful in the management of SLE patients at high risk of acute vascular complications although elucidation of downstream pathways will be necessary to identify targets that do not promote nephritis.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Membrane Glycoproteins/deficiency , Stroke/prevention & control , Animals , Antibodies, Antinuclear/blood , Brain/pathology , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery/etiology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Inflammation Mediators/blood , Kidney/pathology , Lupus Erythematosus, Systemic/chemically induced , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/chemically induced , Lupus Nephritis/genetics , Lupus Nephritis/pathology , Macrophages/physiology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Specific Pathogen-Free Organisms , Stroke/etiology , Terpenes/toxicity
12.
Br J Haematol ; 174(3): 461-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27030515

ABSTRACT

Mechanistic target of rapamycin (mTOR) has been shown to play an important role in red blood cell physiology, with inhibition of mTOR signalling leading to alterations in erythropoiesis. To determine if mTOR inhibition would improve anaemia in sickle cell disease (SCD), mice with SCD were treated with the dual mTORC1/2 inhibitor, INK128. One week after daily oral drug treatment, erythrocyte count, haemoglobin, and haematocrit were all significantly increased while reticulocyte counts were reduced. These parameters remained stable during 3 weeks of treatment. Similar effects were observed following oral treatment with the mTORC1 inhibitor, sirolimus. Sirolimus treatment prolonged the lifespan of sickle cell erythrocytes in circulation, reduced spleen size, and reduced renal and hepatic iron accumulation in SCD mice. Following middle cerebral artery occlusion, stroke size was reduced in SCD mice treated with sirolimus. In conclusion, mTOR inhibition is protective against anaemia and organ damage in a murine model of SCD.


Subject(s)
Anemia, Sickle Cell/drug therapy , Benzoxazoles/pharmacology , Pyrimidines/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Anemia/prevention & control , Anemia, Sickle Cell/complications , Animals , Benzoxazoles/administration & dosage , Disease Models, Animal , Erythrocytes/drug effects , Hematocrit , Hemoglobins , Mice , Pyrimidines/administration & dosage , Splenomegaly/drug therapy , Splenomegaly/prevention & control
13.
Exp Hematol Oncol ; 4: 19, 2015.
Article in English | MEDLINE | ID: mdl-26161296

ABSTRACT

BACKGROUND: The effect of sickle cell disease (SCD) on tumor growth is unknown. Sickled red blood cells may form aggregates within the microvasculature of hypoxic tumors and reduce blood flow leading to impairment of tumor growth. However, there is a paucity of data related to tumor growth in SCD. METHODS: To investigate the effect of SCD on tumor growth in a melanoma model, we generated SCD and control mice using bone marrow transplantation and inoculated the chest wall with B16-F10 melanoma cells. Tumor growth was monitored and angiogenesis was studied in vivo and in vitro. RESULTS: From day 1 to 21, tumor growth rate was nearly identical between SCD and WT mice, however from day 22 to day 29 tumor growth was accelerated in SCD mice compared to WT mice. Disparity in tumor size was confirmed at autopsy with an approximate 2-fold increase in tumor weights from SCD mice. Tumors from SCD mice showed increased vascularity and elevated levels of heme oxygenase-1 (HO-1). HO-1 inhibition with zinc protoporphyrin (ZnPP) blocked the angiogenic and tumor growth response to SCD in vivo and the response to hemin in vitro. CONCLUSIONS: Growth of melanoma tumors is potentiated in a mouse model of SCD. Therapies targeting angiogenesis or HO-1 may be useful in SCD patients with malignant tumors.

14.
Hypertension ; 65(4): 758-65, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646301

ABSTRACT

The renal autonomic nervous system may contribute to hypertension and vascular disease. Although the effects of renal artery denervation on blood pressure lowering are controversial, there may be other beneficial vascular effects independent of blood pressure lowering. Bilateral renal denervation (RDN) or sham operation (SO) was performed in 14-week-old male apolipoprotein E-deficient mice on a Western diet starting at 10 weeks of age. Efficacy of RDN was confirmed by reduction of renal norepinephrine levels (SO: 3.8±0.1 versus RDN: 1.7±0.3 ng/mL; P<0.01) at 6 weeks after procedure. Compared with SO, RDN had no effect on blood pressure (SO: 101.0±2.4 versus RDN: 97.5±1.6 mm Hg; P=0.25), total cholesterol (SO: 536.7±28.5 versus RDN: 535.7±62.9 mg/dL; P=0.99), or triglycerides (SO: 83.7±3.5 versus RDN: 86.9±10.2 mg/dL; P=0.78). Quantification of atherosclerosis at 20 weeks of age demonstrated reduced atherosclerosis in mice receiving RDN compared with SO (arterial tree oil-red-O surface staining RDN: 4.2±0.5% versus SO: 6.3±0.7%; P<0.05). Reduced atherosclerosis was associated with increased smooth muscle cell content in atherosclerotic plaques (RDN: 13.3±2.1 versus SO: 8.1±0.6%; P<0.05). Serum levels of aldosterone, monocyte chemoattractant protein-1, and 8-isoprostane were lower in mice that received RDN compared with sham-operated mice (aldosterone; RDN: 206.8±33.2 versus SO: 405.5±59.4 pg/mL, P<0.05; monocyte chemoattractant protein-1; RDN: 51.7±7.9 versus SO: 91.71±4.6 pg/mL, P<0.05; 8-isoprostane; RDN: 331.9±38.2 versus SO: 468.5±42.0 pg/mL, P<0.05). RDN reduces progression of atherosclerosis in apolipoprotein E-deficient mice. These changes are associated with reduced aldosterone levels, monocyte chemoattractant protein-1, and markers of oxidative stress.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/surgery , Hypertension/surgery , Sympathectomy/methods , Animals , Apolipoproteins E/blood , Atherosclerosis/blood , Atherosclerosis/physiopathology , Blood Pressure Determination , Chemokine CCL2/blood , Dinoprost/analogs & derivatives , Dinoprost/blood , Disease Models, Animal , Disease Progression , Hypertension/blood , Hypertension/physiopathology , Kidney , Male , Mice , Oxidative Stress
15.
PLoS One ; 9(2): e90146, 2014.
Article in English | MEDLINE | ID: mdl-24587248

ABSTRACT

Mebendazole is an antihelminthic drug that exerts its effects via interference with microtubule function in parasites. To determine the utility of mebendazole as a potential treatment for vascular diseases involving proliferation of vascular smooth muscle cells, the effects of mebendazole on vascular smooth muscle cell proliferation were tested in vitro and in a mouse model of arterial injury. In vitro, mebendazole inhibited proliferation and migration of murine vascular smooth muscle cells and this was associated with altered intracellular microtubule organization. To determine in vivo effects of mebendazole following vascular injury, femoral arterial wire injury was induced in wild-type mice treated with either mebendazole or placebo control. Compared with placebo-treated mice, mebendazole-treated mice formed less neointima at the site of injury. Mebendazole is effective at inhibiting vascular smooth muscle cell proliferation and migration, and neointimal formation following arterial injury in mice.


Subject(s)
Mebendazole/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Neointima/drug therapy , Vascular System Injuries/drug therapy , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Hyperplasia , Male , Mebendazole/administration & dosage , Mice , Neointima/pathology , Vascular System Injuries/pathology
17.
Circ Res ; 114(6): 947-56, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24425713

ABSTRACT

RATIONALE: Neutrophil extracellular trap (NET) formation promotes vascular damage, thrombosis, and activation of interferon-α-producing plasmacytoid dendritic cells in diseased arteries. Peptidylarginine deiminase inhibition is a strategy that can decrease in vivo NET formation. OBJECTIVE: To test whether peptidylarginine deiminase inhibition, a novel approach to targeting arterial disease, can reduce vascular damage and inhibit innate immune responses in murine models of atherosclerosis. METHODS AND RESULTS: Apolipoprotein-E (Apoe)(-/-) mice demonstrated enhanced NET formation, developed autoantibodies to NETs, and expressed high levels of interferon-α in diseased arteries. Apoe(-/-) mice were treated for 11 weeks with daily injections of Cl-amidine, a peptidylarginine deiminase inhibitor. Peptidylarginine deiminase inhibition blocked NET formation, reduced atherosclerotic lesion area, and delayed time to carotid artery thrombosis in a photochemical injury model. Decreases in atherosclerosis burden were accompanied by reduced recruitment of netting neutrophils and macrophages to arteries, as well as by reduced arterial interferon-α expression. CONCLUSIONS: Pharmacological interventions that block NET formation can reduce atherosclerosis burden and arterial thrombosis in murine systems. These results support a role for aberrant NET formation in the pathogenesis of atherosclerosis through modulation of innate immune responses.


Subject(s)
Atherosclerosis/prevention & control , Enzyme Inhibitors/therapeutic use , Hydrolases/antagonists & inhibitors , Immunity, Innate/drug effects , Ornithine/analogs & derivatives , Animals , Aortic Diseases/drug therapy , Aortic Diseases/etiology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Atherosclerosis/enzymology , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/pathology , Autoantibodies/biosynthesis , Autoantibodies/immunology , Citrulline/analysis , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Extracellular Space , Histones/metabolism , Hydrolases/physiology , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , L-Selectin/analysis , Lipids/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutropenia/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/ultrastructure , Ornithine/pharmacology , Ornithine/therapeutic use , Photochemical Processes , Protein Processing, Post-Translational/drug effects , Protein-Arginine Deiminase Type 4 , Receptor, Interferon alpha-beta/deficiency , Sinus of Valsalva/pathology , Tunica Intima/pathology
18.
J Am Heart Assoc ; 2(5): e000299, 2013 Sep 16.
Article in English | MEDLINE | ID: mdl-24042086

ABSTRACT

BACKGROUND: The adipocyte-derived hormone leptin is elevated in obesity and may contribute to vascular risk associated with obesity. The mechanism(s) by which leptin affects vascular disease is unclear, although leptin has been shown to increase sympathetic activity. The aim of this study was to investigate the effect of leptin treatment on endothelial function and the role of the local sympathetic nervous system in mediating these effects. METHODS AND RESULTS: Recombinant leptin was administered to C57BL6/J mice every other day for 1 week. Mesenteric arteriole myography revealed that leptin treatment caused significant impairment of endothelium-dependent vasorelaxation. Although leptin alone did not raise aortic blood pressure, leptin treatment augmented the blood pressure response to angiotensin II. The effects of leptin on mesenteric arteriolar function and aortic blood pressure response to angiotensin II were neutralized following sympathetic denervation to the mesenteric vasculature. The superoxide scavenger TEMPOL was also effective in preventing the effects of leptin on endothelial dysfunction. CONCLUSIONS: Leptin causes endothelial dysfunction and enhances the effects of angiotensin II on blood pressure. These effects of leptin are mediated by sympathetic nervous system activation and superoxide and may contribute to vascular stiffness and hypertension in obesity.


Subject(s)
Endothelium, Vascular/physiopathology , Leptin/physiology , Sympathetic Nervous System/physiology , Animals , Blood Pressure , Endothelium, Vascular/drug effects , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Obesity
19.
Br J Haematol ; 162(1): 120-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23590132

ABSTRACT

Sickle cell disease (SCD) is associated with vascular complications including premature stroke. The role of atherothrombosis in these vascular complications is unclear. To determine the effect of SCD on atherosclerosis and thrombosis, mice with SCD along with controls were generated by transplantation of bone marrow from mice carrying the homozygous sickle cell mutation (Hbb(hßs/hßs) ) or wild-type mice (Hbb(+/+) ) into C57BL6/J or apolipoprotein E deficient (Apoe(-/-) ) recipient mice. At the time of sacrifice, 23-28 weeks following bone marrow transplantation, anaemia, reticulocytosis, and splenomegaly were present in mice receiving Hbb(hßs/hßs) bone marrow compared with control mice. Analysis of atherosclerosis involving the aortic root revealed reduced atherosclerotic lesion area with reduced macrophage content and increased collagen content in Apoe(-/-) , Hbb(hßs/hßs) mice compared to Apoe(-/-) , Hbb(+/+) mice. In a carotid thrombosis model, the time to thrombosis was prolonged in Hbb(hßs/hßs) mice compared to Hbb(+/+) mice. This apparent protective effect of SCD on atherosclerosis and thrombosis was diminished by inhibition of heme oxygenase-1 (HMOX1) using zinc protoporphyrin IX. We conclude that SCD in mice is paradoxically protective against atherosclerosis and thrombosis, highlighting the complexity of vascular events in SCD. This protective effect is at least partially mediated by induction of HMOX1.


Subject(s)
Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Atherosclerosis/prevention & control , Thrombosis/prevention & control , Anemia, Sickle Cell/complications , Animals , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/etiology , Carotid Arteries/pathology , Disease Models, Animal , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Lipids/blood , Male , Mice , Mice, Knockout , Protoporphyrins/pharmacology , Thrombosis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...