Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Circ Res ; 127(11): 1422-1436, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32951519

ABSTRACT

RATIONALE: Decades of research have examined immune-modulatory strategies to protect the heart after an acute myocardial infarction and prevent progression to heart failure but have failed to translate to clinical benefit. OBJECTIVE: To determine anti-inflammatory actions of n-apo AI (Apo AI nanoparticles) that contribute to cardiac tissue recovery after myocardial infarction. METHODS AND RESULTS: Using a preclinical mouse model of myocardial infarction, we demonstrate that a single intravenous bolus of n-apo AI (CSL111, 80 mg/kg) delivered immediately after reperfusion reduced the systemic and cardiac inflammatory response. N-apo AI treatment lowered the number of circulating leukocytes by 30±7% and their recruitment into the ischemic heart by 25±10% (all P<5.0×10-2). This was associated with a reduction in plasma levels of the clinical biomarker of cardiac injury, cardiac troponin-I, by 52±17% (P=1.01×10-2). N-apo AI reduced the cardiac expression of chemokines that attract neutrophils and monocytes by 60% to 80% and lowered surface expression of integrin CD11b on monocytes by 20±5% (all P<5.0×10-2). Fluorescently labeled n-apo AI entered the infarct and peri-infarct regions and colocalized with cardiomyocytes undergoing apoptosis and with leukocytes. We further demonstrate that n-apo AI binds to neutrophils and monocytes, with preferential binding to the proinflammatory monocyte subtype and partially via SR-BI (scavenger receptor BI). In patients with type 2 diabetes, we also observed that intravenous infusion of the same n-apo AI (CSL111, 80 mg/kg) similarly reduced the level of circulating leukocytes by 12±5% (all P<5.0×10-2). CONCLUSIONS: A single intravenous bolus of n-apo AI delivered immediately post-myocardial infarction reduced the systemic and cardiac inflammatory response through direct actions on both the ischemic myocardium and leukocytes. These data highlight the anti-inflammatory effects of n-apo AI and provide preclinical support for investigation of its use for management of acute coronary syndromes in the setting of primary percutaneous coronary interventions.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Apolipoprotein A-I/administration & dosage , Inflammation/prevention & control , Leukocytes/drug effects , Myocardial Infarction/drug therapy , Nanoparticles , Administration, Intravenous , Adult , Animals , CD11b Antigen/metabolism , Cells, Cultured , Chemokines/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , Disease Models, Animal , Drug Administration Schedule , Humans , Inflammation/immunology , Inflammation/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Randomized Controlled Trials as Topic , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Troponin I/blood
2.
Sci Transl Med ; 9(411)2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29021167

ABSTRACT

Protecting the heart after an acute coronary syndrome is a key therapeutic goal to support cardiac recovery and prevent progression to heart failure. A potential strategy is to target cardiac glucose metabolism at the early stages after ischemia when glycolysis is critical for myocyte survival. Building on our discovery that high-density lipoprotein (HDL) modulates skeletal muscle glucose metabolism, we now demonstrate that a single dose of reconstituted HDL (rHDL) delivered after myocardial ischemia increases cardiac glucose uptake, reduces infarct size, and improves cardiac remodeling in association with enhanced functional recovery in mice. These findings applied equally to metabolically normal and insulin-resistant mice. We further establish direct effects of HDL on cardiomyocyte glucose uptake, glycolysis, and glucose oxidation via the Akt signaling pathway within 15 min of reperfusion. These data support the use of infusible HDL preparations for management of acute coronary syndromes in the setting of primary percutaneous interventions.


Subject(s)
Lipoproteins, HDL/therapeutic use , Myocardial Infarction/drug therapy , Animals , Glucose/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/drug therapy , Myocardium/metabolism , Myocardium/pathology , Signal Transduction/drug effects
5.
Cardiovasc Res ; 88(1): 186-95, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20501509

ABSTRACT

AIMS: Monocyte systemic levels are known to be a major determinant of ischaemic tissue revascularization, but the mechanisms mediating mobilization of different monocyte subsets-Ly6C(hi) and Ly6C(lo)-to the blood and their respective role in post-ischaemic neovascularization are not clearly understood. Here, we hypothesized that distinct chemokine/chemokine receptor pathways, namely CCL2/CCR2, CX3CL1/CX3CR1, and CCL5/CCR5, differentially control monocyte subset systemic levels, and might thus impact post-ischaemic vessel growth. METHODS AND RESULTS: In a model of murine hindlimb ischaemia, both Ly6C(hi) and Ly6C(lo) monocyte circulating levels were increased after femoral artery ligation. CCL2/CCR2 activation enhanced blood Ly6C(hi) and Ly6C(lo) monocyte counts, although the opposite effect was seen in mice with CCL2 or CCR2 deficiency. CX3CL1/CX3CR1 strongly impacted Ly6C(lo) monocyte levels, whereas CCL5/CCR5 had no role. Only CCL2/CCR2 signalling influenced neovascularization, which was increased in mice overexpressing CCL2, whereas it markedly decreased in CCL2-/- mice. Moreover, adoptive transfer of Ly6C(hi)-but not Ly6C(lo)-monocytes enhanced vessel growth and blood flow recovery. CONCLUSION: Altogether, our data demonstrate that regulation of proangiogenic Ly6C(hi) monocytes systemic levels by CCL2/CCR2 controls post-ischaemic vessel growth, whereas Ly6C(lo) monocytes have no major role in this setting.


Subject(s)
Ischemia/immunology , Monocytes/immunology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Receptors, Chemokine/metabolism , Animals , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1 , Chemokine CCL2/blood , Chemokine CCL5/blood , Chemokine CX3CL1/blood , Chemotaxis, Leukocyte , Disease Models, Animal , Hindlimb , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, CCR2/metabolism , Receptors, CCR5/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL