Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Transl Med ; 22(1): 412, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693516

ABSTRACT

BACKGROUND: Thromboinflammation involving platelet adhesion to endothelial surface-associated von Willebrand factor (VWF) has been implicated in the accelerated progression of non-culprit plaques after MI. The aim of this study was to use arterial endothelial molecular imaging to mechanistically evaluate endothelial-associated VWF as a therapeutic target for reducing remote plaque activation after myocardial infarction (MI). METHODS: Hyperlipidemic mice deficient for the low-density lipoprotein receptor and Apobec-1 underwent closed-chest MI and were treated chronically with either: (i) recombinant ADAMTS13 which is responsible for proteolytic removal of VWF from the endothelial surface, (ii) N-acetylcysteine (NAC) which removes VWF by disulfide bond reduction, (iii) function-blocking anti-factor XI (FXI) antibody, or (iv) no therapy. Non-ischemic controls were also studied. At day 3 and 21, ultrasound molecular imaging was performed with probes targeted to endothelial-associated VWF A1-domain, platelet GPIbα, P-selectin and vascular cell adhesion molecule-1 (VCAM-1) at lesion-prone sites of the aorta. Histology was performed at day 21. RESULTS: Aortic signal for P-selectin, VCAM-1, VWF, and platelet-GPIbα were all increased several-fold (p < 0.01) in post-MI mice versus sham-treated animals at day 3 and 21. Treatment with NAC and ADAMTS13 significantly attenuated the post-MI increase for all four molecular targets by > 50% (p < 0.05 vs. non-treated at day 3 and 21). On aortic root histology, mice undergoing MI versus controls had 2-4 fold greater plaque size and macrophage content (p < 0.05), approximately 20-fold greater platelet adhesion (p < 0.05), and increased staining for markers of platelet transforming growth factor-ß1 signaling. Accelerated plaque growth and inflammatory activation was almost entirely prevented by ADAMTS13 and NAC. Inhibition of FXI had no significant effect on molecular imaging signal or plaque morphology. CONCLUSIONS: Plaque inflammatory activation in remote arteries after MI is strongly influenced by VWF-mediated platelet adhesion to the endothelium. These findings support investigation into new secondary preventive therapies for reducing non-culprit artery events after MI.


Subject(s)
ADAMTS13 Protein , Myocardial Infarction , von Willebrand Factor , Animals , von Willebrand Factor/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/complications , ADAMTS13 Protein/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Mice , Plaque, Atherosclerotic/pathology , P-Selectin/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Male , Molecular Imaging , Aorta/pathology , Aorta/drug effects , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Mice, Inbred C57BL
2.
J Am Soc Echocardiogr ; 37(1): 100-107, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37678655

ABSTRACT

BACKGROUND: Shear created by inertial cavitation of microbubbles by ultrasound augments limb and myocardial perfusion and can reverse tissue ischemia. Our aim was to determine whether this therapeutic bioeffect is attenuated by atherosclerotic risk factors that are known to impair shear-mediated vasodilation and adversely affect microvascular reactivity. METHODS: In mice, lipid-stabilized decafluorobutane microbubbles (2 × 108) were administered intravenously while exposing a proximal hind limb to ultrasound (1.3 MHz, 1.3 mechanical index, pulsing interval 5 seconds) for 10 minutes. Murine strains included wild-type mice and severely hyperlipidemic mice at 15, 35, or 52 weeks of age as a model of aging and elevated cholesterol, and obese db/db mice (≈15 weeks) with severe insulin resistance. Quantitative contrast-enhanced ultrasound perfusion imaging was performed to assess microvascular perfusion in the control and ultrasound-exposed limb. An in situ electrochemical probe and in vivo biophotonic imaging were used to assess limb nitric oxide (NO) and adenosine triphosphosphate concentrations, respectively. RESULTS: Microvascular perfusion was significantly increased by several fold in the cavitation-exposed limb versus control limb for all murine strains and ages (P < .001). In wild-type and hyperlipidemic mice, hyperemia from cavitation was attenuated in the 2 older age groups (P < .01). In young mice (15 weeks), perfusion in cavitation-exposed muscle was less in both the hyperlipidemic mice and the obese db/db mice compared with corresponding wild-type mice. Using young hyperlipidemic mice as a model for flow impairment, limb NO production after cavitation was reduced but adenosine triphosphosphate production was unaltered when compared with age-matched wild-type mice. CONCLUSIONS: In mice, ultrasound cavitation of microbubbles increases limb perfusion by several fold even in the presence of traditional atherosclerotic risk factors. However, older age, hyperlipidemia, and insulin resistance modestly attenuate the degree of flow augmentation, which could impact the degree of flow response in current clinical trials in patients with critical limb ischemia.


Subject(s)
Insulin Resistance , Ultrasonic Therapy , Humans , Mice , Animals , Aged , Infant , Risk Factors , Adenosine , Obesity , Microbubbles
3.
Arterioscler Thromb Vasc Biol ; 43(6): 1041-1053, 2023 06.
Article in English | MEDLINE | ID: mdl-37128919

ABSTRACT

BACKGROUND: In reperfused myocardial infarction, VWF (von Willebrand factor)-mediated platelet adhesion contributes to impaired microvascular reflow and possibly also to postmyocardial infarction inflammation. We hypothesized that postischemic thromboinflammatory processes are worsened by elevated LDL (low-density lipoprotein) cholesterol. METHODS: Myocardial ischemia-reperfusion or sham procedure was performed in wild-type mice and hyperlipidemic mice deficient for the LDL receptor and Apobec-1 (apolipoprotein-B mRNA editing enzyme catalytic polypeptide-1; DKO [double knockout]). DKO subgroups were treated with N-acetylcysteine, which inhibits pro-adhesive VWF multimers or with recombinant ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin motifs-13), which enzymatically cleaves endothelial surface-associated VWF. Myocardial contrast echocardiography perfusion imaging and molecular imaging for VWF, platelet glycoprotein Ibα, and leukocyte CD18 (cluster of differentiation) were performed 30 minutes post-reperfusion. Histology, infarct sizing, and echocardiography were performed at 1.5 or 72 hours; late echocardiography was performed at day 21. RESULTS: After ischemia-reperfusion, DKO compared with wild-type mice had ≈2-fold higher (P<0.05) risk area signal for microvascular platelet adhesion, VWF, and CD18; greater impairment in microvascular reflow, and 2-fold larger infarct size. Treatment of DKO mice with N-acetylcysteine and ADAMTS13 reduced molecular imaging signal for microvascular platelet adhesion, VWF, and CD18; improved early microvascular reflow; and reduced eventual infarct size. ADAMTS13 suppressed the postmyocardial infarction neutrophil and monocyte infiltration, enhanced the time-dependent recovery of left ventricular systolic function, and prevented late left ventricular remodeling. CONCLUSIONS: In reperfused myocardial infarction, elevated LDL cholesterol promotes thromboinflammation through excess microvascular endothelial VWF and platelet adhesion, resulting in less microvascular reflow and larger infarct size. In the presence of elevated LDL cholesterol, therapies that suppress endothelial-associated VWF can promote recovery of left ventricular function and protect against remodeling.


Subject(s)
Myocardial Infarction , Thromboinflammation , Animals , Mice , Acetylcysteine , ADAMTS13 Protein/genetics , Cholesterol, LDL , Inflammation , Ischemia , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , von Willebrand Factor/genetics
4.
JACC Basic Transl Sci ; 7(7): 642-655, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35958695

ABSTRACT

We hypothesized that excess endothelial-associated von Willebrand factor (vWF) and secondary platelet adhesion contribute to aortic valve stenosis (AS). We studied hyperlipidemic mice lacking ADAMTS13 (LDLR -/- AD13 -/- ), which cleaves endothelial-associated vWF multimers. On echocardiography and molecular imaging, LDLR -/- AD13 -/- compared with control strains had increased aortic endothelial vWF and platelet adhesion and developed hemodynamically significant AS, arterial stiffening, high valvulo-aortic impedance, and secondary load-dependent reduction in LV systolic function. Histology revealed leaflet thickening and calcification with valve interstitial cell myofibroblastic and osteogenic transformation, and evidence for TGFß1 pathway activation. We conclude that valve leaflet endothelial vWF-platelet interactions promote AS through juxtacrine platelet signaling.

5.
Cell Mol Bioeng ; 15(3): 231-243, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35611166

ABSTRACT

Introduction: Inflammatory activation of the vascular endothelium leads to overexpression of adhesion molecules such as vascular cell adhesion molecule-1 (VCAM-1), contributing to the pro-thrombotic state underpinning atherogenesis. While the role of TEC family kinases (TFKs) in mediating inflammatory cell and platelet activation is well defined, the role of TFKs in vascular endothelial activation remains unclear. We investigated the role of TFKs in endothelial cell activation in vitro and in a nonhuman primate model of diet-induced atherosclerosis in vivo. Methods and Results: In vitro, we found that ibrutinib blocked activation of the TFK member, BMX, by vascular endothelial growth factors (VEGF)-A in human aortic endothelial cells (HAECs). Blockade of BMX activation with ibrutinib or pharmacologically distinct BMX inhibitors eliminated the ability of VEGF-A to stimulate VCAM-1 expression in HAECs. We validated that treatment with ibrutinib inhibited TFK-mediated platelet activation and aggregation in both human and primate samples as measured using flow cytometry and light transmission aggregometry. We utilized contrast-enhanced ultrasound molecular imaging to measure platelet GPIbα and endothelial VCAM-1 expression in atherosclerosis-prone carotid arteries of obese nonhuman primates. We observed that the TFK inhibitor, ibrutinib, inhibited platelet deposition and endothelial cell activation in vivo. Conclusion: Herein we found that VEGF-A signals through BMX to induce VCAM-1 expression in endothelial cells, and that VCAM-1 expression is sensitive to ibrutinib in vitro and in atherosclerosis-prone carotid arteries in vivo. These findings suggest that TFKs may contribute to the pathogenesis of atherosclerosis and could represent a novel therapeutic target.

6.
Ultrasound Med Biol ; 47(8): 2277-2285, 2021 08.
Article in English | MEDLINE | ID: mdl-33934942

ABSTRACT

Ultrasound (US) is known to stimulate endogenous shear-dependent pathways, and can lower microvascular resistance through mediators that are conducted downstream from US exposure. We hypothesized that endovascular US, already in use for thrombolysis in humans, can improve tissue perfusion in the setting of acute limb ischemia through downstream-conducted effects. Models of severe peripheral arterial disease were developed in mice and in rhesus macaques. An endovascular US catheter (2.3 MHz, 0.5-1.1 MPa) was used to expose the limb adductor in mice for 10 min or the femoral artery distal to stenosis in macaques for 15 min. Quantitative contrast-enhanced ultrasound perfusion imaging was performed to assess flow augmentation in the adductor muscle of mice and the calf muscle of macaques. Microvascular blood flow in the ischemic limb relative to the contralateral control limb was reduced to 22 ± 8% in mice and 36 ± 20% in macaques. US produced immediate 2.3- and 3-fold increases (p < 0.05) in the murine and macaque ischemic limbs, respectively. In macaques, perfusion in the ischemic limb was increased to a normal level. We conclude that non-cavitating US produced by endovascular catheters that are used to enhance thrombolysis in humans can reduce vascular resistance and increase limb perfusion in the setting of acute ischemia.


Subject(s)
Endosonography/methods , Extremities/blood supply , Hindlimb/blood supply , Ischemia/therapy , Peripheral Arterial Disease/therapy , Ultrasonography, Interventional/methods , Animals , Catheters , Endosonography/instrumentation , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Ultrasonography, Interventional/instrumentation
7.
J Am Soc Echocardiogr ; 34(4): 433-442.e3, 2021 04.
Article in English | MEDLINE | ID: mdl-33253812

ABSTRACT

BACKGROUND: Echocardiographic molecular imaging techniques are beginning to be applied to evaluate preclinical efficacy of new drugs. In a large clinical trial, anti-interleukin-1ß (IL-1ß) immunotherapy reduced atherosclerotic events, yet treatment effects were modest, and the mechanisms of action were not fully elucidated. We tested the hypothesis that echocardiographic molecular imaging can assess changes in vascular thromboinflammatory status in response to anti-IL-1ß therapy. METHODS: In wild-type and atherosclerotic mice deficient for the low-density lipoprotein-receptor and Apobec-1, closed-chest myocardial infarction (MI) was performed to mimic high-risk clinical cohorts. Control animals had sham surgery. Post-MI animals were randomized to either no therapy or anti-IL-1ß immunotherapy, which was continued weekly. At post-MI day 3 or 21, in vivo ultrasound molecular imaging of aortic VCAM-1, P-selectin, von Willebrand factor A1-domain, and platelet GPIbα in the thoracic aorta was performed. Aortic histology and NF-κB activity were assessed in atherosclerotic mice. RESULTS: In both atherosclerotic and wild-type mice, MI produced a several-fold increase (P < .05) in aortic molecular signals for P-selectin, VCAM-1, von Willebrand factor, and GPIbα. In atherosclerotic mice, signal remained elevated at day 21. Anti-IL-1ß therapy completely abolished the post-MI increase in signal for all endothelial targets (P < .05 vs nontreated) at day 3 and 21. In atherosclerotic mice, MI triggered an increase in aortic plaque growth and macrophage content, a decrease in plaque collagen, and elevated aortic NF-κB (P < .05 for all changes). All of these remote plaque adverse changes were inhibited by anti-IL-1ß therapy. CONCLUSIONS: Echocardiographic molecular imaging of the vascular endothelium can quantify the beneficial effects of therapies designed to suppress the proatherosclerotic arterial thromboinflammatory effects of alarmins such as IL-1ß. This approach could potentially be used to evaluate the biologic variables that influence response in preclinical studies, and possibly to select patients most likely to benefit from therapy.


Subject(s)
Atherosclerosis , Animals , Disease Models, Animal , Echocardiography , Humans , Immunotherapy , Mice , Molecular Imaging
8.
JACC Basic Transl Sci ; 5(10): 1017-1028, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33145464

ABSTRACT

This study used in vivo molecular imaging to characterize endotheliall activation attributable to von Willebrand factor (vWF)-mediated platelet adhesion in atherosclerosis. In atherosclerotic mice lacking the low-density lipoprotein receptor on Western diet, the additional genetic deletion of the ADAMTS13, which cleaves endothelial-associated vWF, produced greater aortic molecular imaging signal for not only vWF and platelets, but also for endothelial adhesion molecules VCAM1 and P-selectin, larger plaque size, and lower aortic distensibility. Sustained ADAMTS13 therapy reduced signal for all 4 molecular targets and plaque size. We conclude that excess endothelial-associated vWF contributes to not only platelet adhesion, but also to up-regulation of endothelial cell adhesion molecules.

9.
Ultrasound Med Biol ; 46(9): 2361-2369, 2020 09.
Article in English | MEDLINE | ID: mdl-32522456

ABSTRACT

Intra-vascular ultrasound catheters are used clinically to facilitate clot lysis. We hypothesized that these devices could also directly lower microvascular resistance and increase tissue perfusion through established shear-dependent pathways. In mice, either the proximal hind-limb muscles or the upstream femoral artery alone was exposed to an endovascular ultrasound catheter (2.3 MHz, 0.5-1.1 MPa) for 10 min. Quantitative microvascular perfusion imaging in the hind limbs exposed to the endovascular ultrasound system exhibited a more-than-twofold increase in flow (p < 0.01) compared with the contralateral control limb after exposure of either the muscle or the femoral artery alone. Using an in vivo optical imaging reporting system, an eight- to ninefold increase in tissue adenosine triphosphate (ATP) was detected in the region of insonification (p = 0.006). Ultrasound was found to produce an immediate release of ATP from ex vivo erythrocytes (p = 0.03). In situ electrochemical sensing revealed an immediate increase in nitric oxide with initiation of ultrasound which returned to baseline within 5 min of termination, as well as ultrasound-triggered nitric oxide (NO) release from erythrocytes. These data indicate that non-cavitating ultrasound produced by endovascular catheters can reduce vascular resistance and increase flow through recognized shear-dependent vasodilator pathways involving purinergic signaling and NO.


Subject(s)
Catheters , Endosonography/instrumentation , Femoral Artery/physiology , Femoral Artery/radiation effects , Hindlimb/blood supply , Hindlimb/radiation effects , Muscle, Skeletal/blood supply , Muscle, Skeletal/radiation effects , Regional Blood Flow , Ultrasonography, Interventional/instrumentation , Animals , Mice , Mice, Inbred C57BL , Vascular Resistance/radiation effects
10.
J Am Soc Echocardiogr ; 33(8): 1023-1031.e2, 2020 08.
Article in English | MEDLINE | ID: mdl-32532642

ABSTRACT

BACKGROUND: Ultrasound-mediated cavitation of microbubble contrast agents produces high intravascular shear. We hypothesized that microbubble cavitation increases myocardial microvascular perfusion through shear-dependent purinergic pathways downstream from ATP release that is immediate and sustained through cellular ATP channels such as Pannexin-1. METHODS: Quantitative myocardial contrast echocardiography perfusion imaging and in vivo optical imaging of ATP was performed in wild-type and Pannexin-1-deficient (Panx1-/-) mice before and 5 and 30 minutes after 10 minutes of ultrasound-mediated (1.3 MHz, mechanical index 1.3) myocardial microbubble cavitation. Flow augmentation in a preclinical model closer to humans was evaluated in rhesus macaques undergoing myocardial contrast echocardiography perfusion imaging after high-power cavitation in the apical four-chamber plane for 10 minutes. RESULTS: Microbubble cavitation in wild-type mice (n = 7) increased myocardial perfusion by 64% ± 25% at 5 minutes and 95% ± 55% at 30 minutes compared with baseline (P < .05). In Panx1-/- mice (n = 5), perfusion increased by 28% ± 26% at 5 minutes (P = .04) but returned to baseline at 30 minutes. Myocardial ATP signal in wild-type (n = 7) mice undergoing cavitation compared with sham-treated controls (n = 3) was 450-fold higher at 5 minutes and 90-fold higher at 30 minutes after cavitation (P < .001). The ATP signal in Panx1-/- mice (n = 4) was consistently 10-fold lower than that in wild-type mice and was similar to sham controls at 30 minutes. In macaques (n = 8), myocardial perfusion increased twofold in the cavitation-exposed four-chamber plane, similar in degree to that produced by adenosine, but did not increase in the control two-chamber plane. CONCLUSIONS: Cavitation of microbubbles in the myocardial microcirculation produces an immediate release of ATP, likely from cell microporation, as well as sustained release, which is channel dependent and responsible for persistent flow augmentation. These findings provide mechanistic insight by which cavitation improves perfusion and reduces infarct size in patients with myocardial infarction.


Subject(s)
Contrast Media , Microbubbles , Animals , Connexins , Macaca mulatta , Mice , Mice, Inbred C57BL , Myocardium , Nerve Tissue Proteins , Ultrasonography
11.
Blood ; 133(14): 1597-1606, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30692122

ABSTRACT

The third-generation tyrosine kinase inhibitor (TKI) ponatinib has been associated with high rates of acute ischemic events. The pathophysiology responsible for these events is unknown. We hypothesized that ponatinib produces an endothelial angiopathy involving excessive endothelial-associated von Willebrand factor (VWF) and secondary platelet adhesion. In wild-type mice and ApoE-/- mice on a Western diet, ultrasound molecular imaging of the thoracic aorta for VWF A1-domain and glycoprotein-Ibα was performed to quantify endothelial-associated VWF and platelet adhesion. After treatment of wild-type mice for 7 days, aortic molecular signal for endothelial-associated VWF and platelet adhesion were five- to sixfold higher in ponatinib vs sham therapy (P < .001), whereas dasatinib had no effect. In ApoE-/- mice, aortic VWF and platelet signals were two- to fourfold higher for ponatinib-treated compared with sham-treated mice (P < .05) and were significantly higher than in treated wild-type mice (P < .05). Platelet and VWF signals in ponatinib-treated mice were significantly reduced by N-acetylcysteine and completely eliminated by recombinant ADAMTS13. Ponatinib produced segmental left ventricular wall motion abnormalities in 33% of wild-type and 45% of ApoE-/- mice and corresponding patchy perfusion defects, yet coronary arteries were normal on angiography. Instead, a global microvascular angiopathy was detected by immunohistochemistry and by intravital microscopy observation of platelet aggregates and nets associated with endothelial cells and leukocytes. Our findings reveal a new form of vascular toxicity for the TKI ponatinib that involves VWF-mediated platelet adhesion and a secondary microvascular angiopathy that produces ischemic wall motion abnormalities. These processes can be mitigated by interventions known to reduce VWF multimer size.


Subject(s)
Cardiovascular Diseases/chemically induced , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/toxicity , Pyridazines/toxicity , Thrombotic Microangiopathies/complications , Animals , Aorta/metabolism , Endothelium/metabolism , Humans , Ischemia/chemically induced , Mice , Mice, Knockout , Platelet Adhesiveness/drug effects , Protein Kinase Inhibitors/toxicity , Ventricular Dysfunction/chemically induced , von Willebrand Factor/drug effects , von Willebrand Factor/metabolism
12.
Circ Cardiovasc Imaging ; 11(11): e007913, 2018 11.
Article in English | MEDLINE | ID: mdl-30571316

ABSTRACT

BACKGROUND: Complete mechanistic understanding of impaired microvascular reflow after myocardial infarction will likely lead to new therapies for reducing infarct size. Myocardial contrast echocardiography perfusion imaging and molecular imaging were used to evaluate the contribution of microvascular endothelial-associated VWF (von Willebrand factor) and platelet adhesion to microvascular no-reflow. METHODS AND RESULTS: Myocardial infarction was produced by transient LAD ligation in WT (wild type) mice, WT mice treated with the VWF proteolytic enzyme ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13), and ADAMTS13-deficient (ADAMTS13-/-) mice. Myocardial contrast echocardiography perfusion imaging and molecular imaging of VWF and platelet GP (glycoprotein) Ibα were performed 30 minutes after ischemia-reperfusion. Infarct size was measured at 3 days. Mortality during ischemia-reperfusion incrementally increased in WT+ADAMTS13, WT, and ADAMTS13-/- mice (14%, 43%, and 63%, respectively; P<0.05). For WT mice, molecular imaging signal for platelets and VWF in the postischemic risk area was 4- to 5-fold higher ( P<0.05) compared with both the remote nonischemic regions or to sham-treated mice. Signal enhancement in the risk area was completely abolished by ADAMTS13 treatment for both platelets (12.8±3.3 versus -1.0±4.4 IU; P<0.05) and VWF (13.9±4.0 versus -1.0±3.0 IU; P<0.05). ADAMTS13-/- compared with WT mice had 2- to 3-fold higher risk area signal for platelets (33.1±8.5 IU) and VWF (30.9±1.9 IU). Microvascular reflow in the risk area incrementally decreased for WT+ADAMTS13, WT, and ADAMTS13-/- mice ( P<0.05), whereas infarct size incrementally increased ( P<0.05). CONCLUSIONS: Mechanistic information on microvascular no-reflow is possible by combining perfusion and molecular imaging. In reperfused myocardial infarction, excess endothelial-associated VWF and secondary platelet adhesion in the risk area microcirculation contribute to impaired reflow and are modifiable.


Subject(s)
Blood Platelets/physiology , Molecular Imaging/methods , No-Reflow Phenomenon/diagnosis , Platelet Adhesiveness/physiology , von Willebrand Factor/metabolism , Animals , Disease Models, Animal , Echocardiography , Immunohistochemistry , Mice , Mice, Inbred C57BL , No-Reflow Phenomenon/blood , No-Reflow Phenomenon/physiopathology
13.
J Am Soc Echocardiogr ; 31(11): 1252-1259.e1, 2018 11.
Article in English | MEDLINE | ID: mdl-30213420

ABSTRACT

BACKGROUND: Ultrasound molecular imaging was used to evaluate the therapeutic effects of antioxidant therapy with EUK-207, which has superoxide dismutase and catalase activities, on suppressing high-risk atherosclerotic features. METHODS: Mice with age-dependent atherosclerosis produced by deletion of the low-density lipoprotein receptor and Apobec-1 were studied at 20 and 40 weeks of age. EUK-207 or vehicle was administered for the preceding 8 weeks. Therapy for 28 weeks was also studied for 40-week-old mice. Ultrasound molecular imaging of the thoracic aorta was performed with contrast agents targeted to endothelial P-selectin, von Willebrand factor A1-domain, and platelet glycoprotein Ibα or control agent. Aortic plaque area and macrophage content were assessed by histology. RESULTS: In 20-week-old double-knockout mice, EUK-207 compared with sham therapy produced only nonsignificant trends for reduction in molecular imaging signal for endothelial P-selectin, von Willebrand factor A1-domain, and platelet adhesion. At 40 weeks, EUK-207 given for 8 or 28 weeks significantly (P < .05) reduced signal for all three endothelial-associated events essentially to background levels, with the exception of glycoprotein Ibα signal after 8 weeks (P = .06). On aortic histology, EUK-207 therapy for 8 weeks did not affect plaque area or macrophage content at either age. However, EUK-207 for 28 weeks almost completely suppressed plaque development (350 ± 258 vs 4 ± 6 × 103 µm2, P = .014) and macrophage content (136 ± 103 vs 3 ± 2 × 103 µm2, P = .002) compared with control mice at 40 weeks. CONCLUSIONS: Molecular imaging can be used to assess vascular responses to antioxidants and has demonstrated that certain antioxidants reduce vascular endothelial activation and platelet adhesion, but reductions in plaque size and macrophage content occurs only with long-duration therapy that is started early.


Subject(s)
Antioxidants/therapeutic use , Aorta, Thoracic/diagnostic imaging , Aortic Diseases/drug therapy , Atherosclerosis/drug therapy , Contrast Media/pharmacology , Molecular Imaging/methods , Ultrasonography/methods , Animals , Aortic Diseases/diagnosis , Atherosclerosis/diagnosis , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Treatment Outcome
14.
J Am Coll Cardiol ; 72(9): 1015-1026, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30139430

ABSTRACT

BACKGROUND: In the months after acute myocardial infarction (MI), risk for acute atherothrombotic events in nonculprit arteries increases several fold. OBJECTIVES: This study investigated whether sustained proinflammatory and prothrombotic endothelial alterations occur in remote vessels after MI. METHODS: Wild-type mice, atherosclerotic mice with double knockout (DKO) of the low-density lipoprotein receptor and Apobec-1, and DKO mice treated with the Nox-inhibitor apocynin were studied at baseline and at 3 and 21 days after closed-chest MI. Ultrasound molecular imaging of P-selectin, vascular cell adhesion molecule (VCAM)-1, von Willebrand factor (VWF) A1-domain, and platelet GPIbα was performed. Intravital microscopy was used to characterize post-MI leukocyte and platelet recruitment in the remote microcirculation after MI. RESULTS: Aortic molecular imaging for P-selectin, VCAM-1, VWF-A1, and platelets was increased several-fold (p < 0.01) 3 days post-MI for both wild-type and DKO mice. At 21 days, these changes resolved in wild-type mice but persisted in DKO mice. Signal for platelet adhesion was abolished 1 h after administration of ADAMTS13, which regulates VWF multimerization. In DKO and wild-type mice, apocynin significantly attenuated the post-MI increase for molecular targets, and platelet depletion significantly reduced P-selectin and VCAM-1 signal. On intravital microscopy, MI resulted in remote vessel leukocyte adhesion and platelet string or net complexes. On histology, high-risk inflammatory features in aortic plaque increased in DKO mice 21 days post-MI, which were completely prevented by apocynin. CONCLUSIONS: Acute MI stimulates a spectrum of changes in remote vessels, including up-regulation of endothelial inflammatory adhesion molecules and platelet-endothelial adhesion from endothelial-associated VWF multimers. These remote arterial alterations persist longer in the presence of hyperlipidemia, are associated with accelerated plaque growth and inflammation, and are attenuated by Nox inhibition.


Subject(s)
Myocardial Infarction/blood , Animals , Blood Cell Count , Disease Models, Animal , Mice , P-Selectin/blood , Platelet Activation , Vascular Cell Adhesion Molecule-1/blood , von Willebrand Factor/metabolism
15.
J Am Soc Echocardiogr ; 30(5): 503-510.e1, 2017 May.
Article in English | MEDLINE | ID: mdl-28238588

ABSTRACT

BACKGROUND: Contrast-enhanced ultrasound (CEU) limb perfusion imaging is a promising approach for evaluating peripheral artery disease (PAD). However, low signal enhancement in skeletal muscle has necessitated high-power intermittent imaging algorithms, which are not clinically feasible. We hypothesized that CEU using a combination of intermediate power and a contrast agent resistant to inertial cavitation would allow real-time limb stress perfusion imaging. METHODS: In normal volunteers, CEU of the calf skeletal muscle was performed on separate days with Sonazoid, Optison, or Definity. Progressive reduction in the ultrasound pulsing interval was used to assess the balance between signal enhancement and agent destruction at escalating mechanical indices (MI, 0.1-0.4). Real-time perfusion imaging at MI 0.1-0.4 using postdestructive replenishment kinetics was performed at rest and during 25 W plantar flexion contractile exercise. RESULTS: For Optison, limb perfusion imaging was unreliable at rest due to very low signal enhancement generated at all MIs and was possible during exercise-induced hyperemia only at MI 0.1 due to agent destruction at higher MIs. For Definity, signal intensity progressively increased with MI but was offset by microbubble destruction, which resulted in modest signal enhancement during CEU perfusion imaging and distortion of replenishment curves at MI ≥ 0.2. For Sonazoid, there strong signal enhancement at MI ≥ 0.2, with little destruction detected only at MI 0.4. Accordingly, high signal intensity and nondistorted perfusion imaging was possible at MI 0.2-0.3 and detected an 8.0- ± 5.7-fold flow reserve. CONCLUSIONS: Rest-stress limb perfusion imaging in humans with real-time CEU, which requires only seconds to perform, is possible using microbubbles with viscoelastic properties that produce strong nonlinear signal generation without destruction at intermediate acoustic pressures.


Subject(s)
Blood Flow Velocity/physiology , Exercise Test/methods , Fluorocarbons , Leg/physiology , Muscle, Skeletal/physiology , Perfusion Imaging/methods , Ultrasonography/methods , Adult , Contrast Media , Female , Humans , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Leg/blood supply , Male , Microbubbles , Muscle, Skeletal/blood supply , Reproducibility of Results , Rest , Sensitivity and Specificity
16.
Circulation ; 135(13): 1240-1252, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28174191

ABSTRACT

BACKGROUND: Augmentation of tissue blood flow by therapeutic ultrasound is thought to rely on convective shear. Microbubble contrast agents that undergo ultrasound-mediated cavitation markedly amplify these effects. We hypothesized that purinergic signaling is responsible for shear-dependent increases in muscle perfusion during therapeutic cavitation. METHODS: Unilateral exposure of the proximal hindlimb of mice (with or without ischemia produced by iliac ligation) to therapeutic ultrasound (1.3 MHz, mechanical index 1.3) was performed for 10 minutes after intravenous injection of 2×108 lipid microbubbles. Microvascular perfusion was evaluated by low-power contrast ultrasound perfusion imaging. In vivo muscle ATP release and in vitro ATP release from endothelial cells or erythrocytes were assessed by a luciferin-luciferase assay. Purinergic signaling pathways were assessed by studying interventions that (1) accelerated ATP degradation; (2) inhibited P2Y receptors, adenosine receptors, or KATP channels; or (3) inhibited downstream signaling pathways involving endothelial nitric oxide synthase or prostanoid production (indomethacin). Augmentation in muscle perfusion by ultrasound cavitation was assessed in a proof-of-concept clinical trial in 12 subjects with stable sickle cell disease. RESULTS: Therapeutic ultrasound cavitation increased muscle perfusion by 7-fold in normal mice, reversed tissue ischemia for up to 24 hours in the murine model of peripheral artery disease, and doubled muscle perfusion in patients with sickle cell disease. Augmentation in flow extended well beyond the region of ultrasound exposure. Ultrasound cavitation produced an ≈40-fold focal and sustained increase in ATP, the source of which included both endothelial cells and erythrocytes. Inhibitory studies indicated that ATP was a critical mediator of flow augmentation that acts primarily through either P2Y receptors or adenosine produced by ectonucleotidase activity. Combined indomethacin and inhibition of endothelial nitric oxide synthase abolished the effects of therapeutic ultrasound, indicating downstream signaling through both nitric oxide and prostaglandins. CONCLUSIONS: Therapeutic ultrasound using microbubble cavitation to increase muscle perfusion relies on shear-dependent increases in ATP, which can act through a diverse portfolio of purinergic signaling pathways. These events can reverse hindlimb ischemia in mice for >24 hours and increase muscle blood flow in patients with sickle cell disease. CLINICAL TRIAL REGISTRATION: URL: http://clinicaltrials.gov. Unique identifier: NCT01566890.


Subject(s)
Adenosine Triphosphate/metabolism , Muscle, Skeletal/blood supply , Purinergic Agents/metabolism , Ultrasonography/methods , Animals , Hemodynamics , Humans , Male , Mice , Mice, Inbred C57BL , Microbubbles , Signal Transduction
17.
J Am Soc Echocardiogr ; 29(8): 812-818, 2016 08.
Article in English | MEDLINE | ID: mdl-27267307

ABSTRACT

BACKGROUND: Contrast ultrasound-mediated gene delivery (CUMGD) is a promising approach for enhancing gene therapy that relies on microbubble (MB) cavitation to augment complementary deoxyribonucleic acid (cDNA) transfection. The aims of this study were to determine optimal conditions for charge-coupling cDNA to MBs and to evaluate the advantages of surface loading for gene transfection in muscle and liver. METHODS: Charge coupling of fluorescently labeled cDNA to either neutral MBs (MBN) or cationic MBs (MB+) in low- to high-ionic conditions (0.3%-1.8% NaCl) was assessed by flow cytometry. MB aggregation from cDNA coupling was determined by electrozone sensing. Tissue transfection of luciferase in murine hindlimb skeletal muscle and liver was made by CUMGD with MBN or MB+ combined with subsaturated, saturated, or supersaturated cDNA concentrations (2.5, 50, and 200 µg/10(8) MBs). RESULTS: Charge-coupling of cDNA was detected for MB+ but not MBN. Coupling occurred over almost the entire range of ionic conditions, with a peak at 1.2% NaCl, although electrostatic interference occurred at >1.5% NaCl. DNA-mediated aggregation of MB+ was observed at ≤0.6% NaCl but did not reduce the ability to produce inertial cavitation. Transfection with CUMGD in muscle and liver was low for both MBs at subsaturation concentrations. In muscle, higher cDNA concentrations produced a 10-fold higher degree of transfection with MB+, which was approximately fivefold higher (P < .05) than that for MBN. There was no effect of DNA supersaturation. The same pattern was seen for liver except that supersaturation further increased transfection with MBN equal to that of MB+. CONCLUSIONS: Efficient charge-coupling of cDNA to MB+ but not MBN occurs over a relatively wide range of ionic conditions without aggregation. Transfection with CUMGD is much more efficient with charge-coupling of cDNA to MBs and is not affected by supersaturation except in the liver, which is specialized for macromolecular and cDNA uptake.


Subject(s)
DNA/pharmacokinetics , Delayed-Action Preparations/radiation effects , High-Energy Shock Waves , Liver/metabolism , Muscle, Skeletal/metabolism , Sonication/methods , Transfection/methods , Animals , Contrast Media/chemistry , Contrast Media/radiation effects , DNA/chemistry , Delayed-Action Preparations/chemistry , Genetic Therapy/methods , Mice , Mice, Inbred C57BL , Microbubbles
18.
JACC Cardiovasc Imaging ; 9(8): 937-46, 2016 08.
Article in English | MEDLINE | ID: mdl-27318722

ABSTRACT

OBJECTIVES: This study hypothesized that microvascular retention of phosphatidylserine-containing microbubbles (MB-PS) would allow detection of recent but resolved myocardial ischemia with myocardial contrast echocardiographic (MCE) molecular imaging. BACKGROUND: Techniques for ischemic memory imaging which can detect and spatially assess resolved myocardial ischemia are being developed for rapid evaluation of patients with chest pain. METHODS: MCE molecular imaging with MB-PS was performed 1.5 h, 3.0 h, and 6.0 h after brief (10 min) myocardial ischemia in mice; data were compared to selectin-targeted microbubbles. MCE molecular imaging with Sonazoid (GE Healthcare, Amersham, United Kingdom), a commercially produced phosphatidylserine (PS) - containing agent, was performed in separate mice at 1.5 h and 3.0 h after ischemia-reperfusion; and in dogs undergoing 135 min of ischemia and 60 min of reflow as well as in closed-chest nonischemic control dogs. The mechanism for MB-PS attachment was assessed by intravital microscopy of post-ischemic muscle and by flow cytometry analysis of cell-MB interactions. RESULTS: In mice undergoing ischemia-reperfusion without infarction, signal enhancement in the risk area for MB-PS and p-selectin glycoprotein ligand-1-targeted microbubbles was similar at reflow times of 1.5 h (23.3 ± 7.3 IU vs. 30.7 ± 4.1 IU), 3.0 h (42.2 ± 6.2 IU vs. 33.9 ± 7.4 IU), and 6.0 h (24.1 ± 4.3 IU vs. 25.5 ± 4.7 IU). For both agents, signal in the risk area was significantly (p < 0.05) higher than remote region at all reflow times. Sonazoid also produced strong risk area enhancement at 1.5 h (34.7 ± 5.0 IU) and 3.0 h (52.5 ± 4.5 IU) which was approximately 3-fold greater than in the control region, and which correlated spatially with the microsphere-derived risk area. In dogs, Sonazoid signal in the risk area was >5-fold higher than in closed-chest control myocardium (42.2 ± 8.1 IU vs. 7.9 ± 3.3 IU; p < 0.001). Mechanistic studies indicated that MB-PS attached directly to venular endothelium and adherent leukocytes which was dependent on serum complement components C1q and C3. CONCLUSIONS: Ischemic memory imaging with MCE is possible using MB-PS which may obviate the need for ligand-directed targeting.


Subject(s)
Complement System Proteins/metabolism , Contrast Media/administration & dosage , Coronary Vessels/metabolism , Echocardiography/methods , Ferric Compounds/administration & dosage , Iron/administration & dosage , Microbubbles , Molecular Imaging/methods , Myocardial Infarction/diagnostic imaging , Myocardial Reperfusion Injury/diagnostic imaging , Oxides/administration & dosage , Phosphatidylserines/administration & dosage , Animals , Complement C1q/metabolism , Complement C3/metabolism , Contrast Media/metabolism , Coronary Vessels/pathology , Disease Models, Animal , Dogs , Ferric Compounds/metabolism , Flow Cytometry , Intravital Microscopy , Iron/metabolism , Male , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Oxides/metabolism , Phosphatidylserines/metabolism , Predictive Value of Tests , Time Factors
19.
Circ Cardiovasc Imaging ; 8(7): e002765, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26156014

ABSTRACT

BACKGROUND: Nonthrombotic platelet-endothelial interactions may contribute to atherosclerotic plaque development, although in vivo studies examining mechanism without platelet preactivation are lacking. Using in vivo molecular imaging at various stages of atherosclerosis, we quantified platelet-endothelial interactions and evaluated the contribution of major adhesion pathways. METHODS AND RESULTS: Mice deficient for the low-density lipoprotein receptor and Apobec-1 were studied as an age-dependent model of atherosclerosis at 10, 20, 30, and 40 weeks of age, which provided progressive increase in stage from early fatty streak (10 weeks) to large complex plaques without rupture (40 weeks). Platelet-targeted contrast ultrasound molecular imaging of the thoracic aorta performed with microbubbles targeted to GPIbα demonstrated selective signal enhancement as early as 10 weeks of age. This signal increased progressively with age (almost 8-fold increase from 10 to 40 weeks, analysis of variance P<0.001). Specificity for platelet targeting was confirmed by the reduction in platelet-targeted signal commensurate with the decrease in platelet count after immunodepletion with anti-GPIb or anti-CD41 antibody. Inhibition of P-selectin in 20 and 40 weeks atherosclerotic mice resulted in a small (15% to 30%) reduction in platelet signal. Molecular imaging with microbubbles targeted to the A1 domain of von Willebrand factor demonstrated selective signal enhancement at all time points, which did not significantly increase with age. Treatment of 20 and 40 week mice with recombinant ADAMTS13 eliminated platelet and von Willebrand factor molecular imaging signal. CONCLUSIONS: Platelet-endothelial interactions occur in early atherosclerosis. These interactions are in part caused by endothelial von Willebrand factor large multimers, which can be reversed with exogenous ADAMTS13.


Subject(s)
Aorta, Thoracic/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Blood Platelets/metabolism , Endothelial Cells/metabolism , Molecular Imaging/methods , Platelet Adhesiveness , von Willebrand Factor/metabolism , ADAM Proteins/metabolism , ADAMTS13 Protein , APOBEC-1 Deaminase , Animals , Aorta, Thoracic/diagnostic imaging , Aortic Diseases/diagnostic imaging , Aortic Diseases/genetics , Atherosclerosis/diagnostic imaging , Atherosclerosis/genetics , Blood Platelets/diagnostic imaging , Contrast Media , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Disease Models, Animal , Endothelial Cells/diagnostic imaging , Mice, Inbred C57BL , Mice, Knockout , Microbubbles , Microscopy, Fluorescence , P-Selectin/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Multimerization , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Ultrasonography
20.
Article in English | MEDLINE | ID: mdl-25834183

ABSTRACT

BACKGROUND: Ultrasound can increase tissue blood flow, in part, through the intravascular shear produced by oscillatory pressure fluctuations. We hypothesized that ultrasound-mediated increases in perfusion can be augmented by microbubble contrast agents that undergo ultrasound-mediated cavitation and sought to characterize the biological mediators. METHODS AND RESULTS: Contrast ultrasound perfusion imaging of hindlimb skeletal muscle and femoral artery diameter measurement were performed in nonischemic mice after unilateral 10-minute exposure to intermittent ultrasound alone (mechanical index, 0.6 or 1.3) or ultrasound with lipid microbubbles (2×10(8) IV). Studies were also performed after inhibiting shear- or pressure-dependent vasodilator pathways, and in mice with hindlimb ischemia. Ultrasound alone produced a 2-fold increase (P<0.05) in muscle perfusion regardless of ultrasound power. Ultrasound-mediated augmentation in flow was greater with microbubbles (3- and 10-fold higher than control for mechanical index 0.6 and 1.3, respectively; P<0.05), as was femoral artery dilation. Inhibition of endothelial nitric oxide synthase attenuated flow augmentation produced by ultrasound and microbubbles by 70% (P<0.01), whereas inhibition of adenosine-A2a receptors and epoxyeicosatrienoic acids had minimal effect. Limb nitric oxide production and muscle phospho-endothelial nitric oxide synthase increased in a stepwise fashion by ultrasound and ultrasound with microbubbles. In mice with unilateral hindlimb ischemia (40%-50% reduction in flow), ultrasound (mechanical index, 1.3) with microbubbles increased perfusion by 2-fold to a degree that was greater than the control nonischemic limb. CONCLUSIONS: Increases in muscle blood flow during high-power ultrasound are markedly amplified by the intravascular presence of microbubbles and can reverse tissue ischemia. These effects are most likely mediated by cavitation-related increases in shear and activation of endothelial nitric oxide synthase.


Subject(s)
Femoral Artery/diagnostic imaging , Hindlimb/blood supply , Hindlimb/diagnostic imaging , Ischemia/diagnostic imaging , Ischemia/physiopathology , Microbubbles , Muscle, Skeletal/diagnostic imaging , Animals , Dilatation, Pathologic , Endothelium, Vascular/physiopathology , Femoral Artery/pathology , Male , Mice, Inbred C57BL , Myocardial Perfusion Imaging , Regional Blood Flow , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...