Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Circulation ; 148(8): 661-678, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37427430

RESUMEN

BACKGROUND: Fewer than 50% of patients who develop aortic valve calcification have concomitant atherosclerosis, implying differential pathogenesis. Although circulating extracellular vesicles (EVs) act as biomarkers of cardiovascular diseases, tissue-entrapped EVs are associated with early mineralization, but their cargoes, functions, and contributions to disease remain unknown. METHODS: Disease stage-specific proteomics was performed on human carotid endarterectomy specimens (n=16) and stenotic aortic valves (n=18). Tissue EVs were isolated from human carotid arteries (normal, n=6; diseased, n=4) and aortic valves (normal, n=6; diseased, n=4) by enzymatic digestion, (ultra)centrifugation, and a 15-fraction density gradient validated by proteomics, CD63-immunogold electron microscopy, and nanoparticle tracking analysis. Vesiculomics, comprising vesicular proteomics and small RNA-sequencing, was conducted on tissue EVs. TargetScan identified microRNA targets. Pathway network analyses prioritized genes for validation in primary human carotid artery smooth muscle cells and aortic valvular interstitial cells. RESULTS: Disease progression drove significant convergence (P<0.0001) of carotid artery plaque and calcified aortic valve proteomes (2318 proteins). Each tissue also retained a unique subset of differentially enriched proteins (381 in plaques; 226 in valves; q<0.05). Vesicular gene ontology terms increased 2.9-fold (P<0.0001) among proteins modulated by disease in both tissues. Proteomics identified 22 EV markers in tissue digest fractions. Networks of proteins and microRNA targets changed by disease progression in both artery and valve EVs revealed shared involvement in intracellular signaling and cell cycle regulation. Vesiculomics identified 773 proteins and 80 microRNAs differentially enriched by disease exclusively in artery or valve EVs (q<0.05); multiomics integration found tissue-specific EV cargoes associated with procalcific Notch and Wnt signaling in carotid arteries and aortic valves, respectively. Knockdown of tissue-specific EV-derived molecules FGFR2, PPP2CA, and ADAM17 in human carotid artery smooth muscle cells and WNT5A, APP, and APC in human aortic valvular interstitial cells significantly modulated calcification. CONCLUSIONS: The first comparative proteomics study of human carotid artery plaques and calcified aortic valves identifies unique drivers of atherosclerosis versus aortic valve stenosis and implicates EVs in advanced cardiovascular calcification. We delineate a vesiculomics strategy to isolate, purify, and study protein and RNA cargoes from EVs entrapped in fibrocalcific tissues. Integration of vesicular proteomics and transcriptomics by network approaches revealed novel roles for tissue EVs in modulating cardiovascular disease.


Asunto(s)
Estenosis de la Válvula Aórtica , Aterosclerosis , Calcinosis , Vesículas Extracelulares , MicroARNs , Humanos , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/patología , Multiómica , Calcinosis/metabolismo , Células Cultivadas , MicroARNs/metabolismo , Aterosclerosis/patología , Vía de Señalización Wnt , Vesículas Extracelulares/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 41(9): 2399-2416, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34289702

RESUMEN

Objective: Vascular smooth muscle cell (VSMC) plasticity plays a critical role in the development of atherosclerosis. Long noncoding RNAs (lncRNAs) are emerging as important regulators in the vessel wall and impact cellular function through diverse interactors. However, the role of lncRNAs in regulating VSMCs plasticity and atherosclerosis remains unclear. Approach and Results: We identified a VSMC-enriched lncRNA cardiac mesoderm enhancer-associated noncoding RNA (CARMN) that is dynamically regulated with progression of atherosclerosis. In both mouse and human atherosclerotic plaques, CARMN colocalized with VSMCs and was expressed in the nucleus. Knockdown of CARMN using antisense oligonucleotides in Ldlr−/− mice significantly reduced atherosclerotic lesion formation by 38% and suppressed VSMCs proliferation by 45% without affecting apoptosis. In vitro CARMN gain- and loss-of-function studies verified effects on VSMC proliferation, migration, and differentiation. TGF-ß1 (transforming growth factor-beta) induced CARMN expression in a Smad2/3-dependent manner. CARMN regulated VSMC plasticity independent of the miR143/145 cluster, which is located in close proximity to the CARMN locus. Mechanistically, lncRNA pulldown in combination with mass spectrometry analysis showed that the nuclear-localized CARMN interacted with SRF (serum response factor) through a specific 600­1197 nucleotide domain. CARMN enhanced SRF occupancy on the promoter regions of its downstream VSMC targets. Finally, knockdown of SRF abolished the regulatory role of CARMN in VSMC plasticity. Conclusions: The lncRNA CARMN is a critical regulator of VSMC plasticity and atherosclerosis. These findings highlight the role of a lncRNA in SRF-dependent signaling and provide implications for a range of chronic vascular occlusive disease states.


Asunto(s)
Aterosclerosis/metabolismo , Plasticidad de la Célula , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , ARN Largo no Codificante/metabolismo , Factor de Respuesta Sérica/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Línea Celular , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fenotipo , Placa Aterosclerótica , ARN Largo no Codificante/genética , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factor de Respuesta Sérica/genética , Transducción de Señal
3.
N Engl J Med ; 377(2): 111-121, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28636844

RESUMEN

BACKGROUND: Clonal hematopoiesis of indeterminate potential (CHIP), which is defined as the presence of an expanded somatic blood-cell clone in persons without other hematologic abnormalities, is common among older persons and is associated with an increased risk of hematologic cancer. We previously found preliminary evidence for an association between CHIP and atherosclerotic cardiovascular disease, but the nature of this association was unclear. METHODS: We used whole-exome sequencing to detect the presence of CHIP in peripheral-blood cells and associated such presence with coronary heart disease using samples from four case-control studies that together enrolled 4726 participants with coronary heart disease and 3529 controls. To assess causality, we perturbed the function of Tet2, the second most commonly mutated gene linked to clonal hematopoiesis, in the hematopoietic cells of atherosclerosis-prone mice. RESULTS: In nested case-control analyses from two prospective cohorts, carriers of CHIP had a risk of coronary heart disease that was 1.9 times as great as in noncarriers (95% confidence interval [CI], 1.4 to 2.7). In two retrospective case-control cohorts for the evaluation of early-onset myocardial infarction, participants with CHIP had a risk of myocardial infarction that was 4.0 times as great as in noncarriers (95% CI, 2.4 to 6.7). Mutations in DNMT3A, TET2, ASXL1, and JAK2 were each individually associated with coronary heart disease. CHIP carriers with these mutations also had increased coronary-artery calcification, a marker of coronary atherosclerosis burden. Hypercholesterolemia-prone mice that were engrafted with bone marrow obtained from homozygous or heterozygous Tet2 knockout mice had larger atherosclerotic lesions in the aortic root and aorta than did mice that had received control bone marrow. Analyses of macrophages from Tet2 knockout mice showed elevated expression of several chemokine and cytokine genes that contribute to atherosclerosis. CONCLUSIONS: The presence of CHIP in peripheral-blood cells was associated with nearly a doubling in the risk of coronary heart disease in humans and with accelerated atherosclerosis in mice. (Funded by the National Institutes of Health and others.).


Asunto(s)
Aterosclerosis/genética , Evolución Clonal , Enfermedad Coronaria/genética , Hematopoyesis/genética , Mutación , Animales , Estudios de Casos y Controles , Exoma , Predisposición Genética a la Enfermedad , Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Noqueados , Riesgo , Análisis de Secuencia de ADN/métodos
4.
Circ Res ; 123(1): 33-42, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29572206

RESUMEN

RATIONALE: Neutrophils likely contribute to the thrombotic complications of human atheromata. In particular, neutrophil extracellular traps (NETs) could exacerbate local inflammation and amplify and propagate arterial intimal injury and thrombosis. PAD4 (peptidyl arginine deiminase 4) participates in NET formation, but an understanding of this enzyme's role in atherothrombosis remains scant. OBJECTIVE: This study tested the hypothesis that PAD4 and NETs influence experimental atherogenesis and in processes implicated in superficial erosion, a form of plaque complication we previously associated with NETs. METHODS AND RESULTS: Bone marrow chimeric Ldlr deficient mice reconstituted with either wild-type or PAD4-deficient cells underwent studies that assessed atheroma formation or procedures designed to probe mechanisms related to superficial erosion. PAD4 deficiency neither retarded fatty streak formation nor reduced plaque size or inflammation in bone marrow chimeric mice that consumed an atherogenic diet. In contrast, either a PAD4 deficiency in bone marrow-derived cells or administration of DNaseI to disrupt NETs decreased the extent of arterial intimal injury in mice with arterial lesions tailored to recapitulate characteristics of human atheroma complicated by erosion. CONCLUSIONS: These results indicate that PAD4 from bone marrow-derived cells and NETs do not influence chronic experimental atherogenesis, but participate causally in acute thrombotic complications of intimal lesions that recapitulate features of superficial erosion.


Asunto(s)
Trampas Extracelulares/fisiología , Hidrolasas/fisiología , Placa Aterosclerótica/etiología , Trombosis/etiología , Animales , Trasplante de Médula Ósea , Enfermedades de las Arterias Carótidas/etiología , Enfermedades de las Arterias Carótidas/patología , Muerte Celular , Desoxirribonucleasa I/farmacología , Trampas Extracelulares/efectos de los fármacos , Humanos , Hidrolasas/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología , Osteomielitis/etiología , Placa Aterosclerótica/patología , Arginina Deiminasa Proteína-Tipo 4 , Trombosis/prevención & control , Túnica Íntima/lesiones
5.
Eur Heart J ; 40(30): 2482-2491, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30698710

RESUMEN

AIMS: Targeting interleukin-1 (IL-1) represents a novel therapeutic approach to atherosclerosis. CANTOS demonstrated the benefits of IL-1ß neutralization in patients post-myocardial infarction with residual inflammatory risk. Yet, some mouse data have shown a prominent role of IL-1α rather than IL-1ß in atherosclerosis, or even a deleterious effect of IL-1 on outward arterial remodelling in atherosclerosis-susceptible mice. To shed light on these disparate results, this study investigated the effect of neutralizing IL-1α or/and IL-1ß isoforms starting either early in atherogenesis or later in ApoE-/- mice with established atheroma. METHODS AND RESULTS: The neutralization of IL-1α or of both IL-1 isoforms impaired outward remodelling during early atherogenesis as assessed by micro-computed tomographic and histologic assessment. In contrast, the neutralization of IL-1ß did not impair outward remodelling either during early atherogenesis or in mice with established lesions. Interleukin-1ß inhibition promoted a slant of blood monocytes towards a less inflammatory state during atherogenesis, reduced the size of established atheromata, and increased plasma levels of IL-10 without limiting outward remodelling of brachiocephalic arteries. CONCLUSION: This study established a pivotal role for IL-1α in the remodelling of arteries during early experimental atherogenesis, whereas IL-1ß drives inflammation during atherogenesis and the evolution of advanced atheroma in mice.


Asunto(s)
Aterosclerosis/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Animales , Arteritis/metabolismo , Modelos Animales de Enfermedad , Interleucina-1alfa/antagonistas & inhibidores , Interleucina-1alfa/sangre , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/sangre , Masculino , Ratones , Ratones Noqueados , Monocitos/metabolismo , Placa Aterosclerótica/metabolismo
6.
Circ Res ; 121(1): 31-42, 2017 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-28428204

RESUMEN

RATIONALE: Superficial erosion currently causes up to a third of acute coronary syndromes; yet, we lack understanding of its mechanisms. Thrombi because of superficial intimal erosion characteristically complicate matrix-rich atheromata in regions of flow perturbation. OBJECTIVE: This study tested in vivo the involvement of disturbed flow and of neutrophils, hyaluronan, and Toll-like receptor 2 ligation in superficial intimal injury, a process implicated in superficial erosion. METHODS AND RESULTS: In mouse carotid arteries with established intimal lesions tailored to resemble the substrate of human eroded plaques, acute flow perturbation promoted downstream endothelial cell activation, neutrophil accumulation, endothelial cell death and desquamation, and mural thrombosis. Neutrophil loss-of-function limited these findings. Toll-like receptor 2 agonism activated luminal endothelial cells, and deficiency of this innate immune receptor decreased intimal neutrophil adherence in regions of local flow disturbance, reducing endothelial cell injury and local thrombosis (P<0.05). CONCLUSIONS: These results implicate flow disturbance, neutrophils, and Toll-like receptor 2 signaling as mechanisms that contribute to superficial erosion, a cause of acute coronary syndrome of likely growing importance in the statin era.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Endotelio Vascular/metabolismo , Infiltración Neutrófila/fisiología , Receptor Toll-Like 2/deficiencia , Animales , Trasplante de Médula Ósea/métodos , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Células Cultivadas , Endotelio Vascular/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Circ Res ; 114(1): 32-40, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24084690

RESUMEN

RATIONALE: Activated nuclear factor (NF)-κB signaling in the vascular endothelium promotes the initiation and progression of atherosclerosis. Targeting endothelial NF-κB may provide a novel strategy to limit chronic inflammation. OBJECTIVE: To examine the role of microRNA-181b (miR-181b) in endothelial NF-κB signaling and effects on atherosclerosis. METHODS AND RESULTS: MiR-181b expression was reduced in the aortic intima and plasma in apolipoprotein E-deficient mice fed a high-fat diet. Correspondingly, circulating miR-181b in the plasma was markedly reduced in human subjects with coronary artery disease. Systemic delivery of miR-181b resulted in a 2.3-fold overexpression of miR-181b in the aortic intima of apolipoprotein E-deficient mice and suppressed NF-κB signaling revealed by bioluminescence imaging and reduced target gene expression in the aortic arch in apolipoprotein E-deficient/NF-κB-luciferase transgenic mice. MiR-181b significantly inhibited atherosclerotic lesion formation, proinflammatory gene expression and the influx of lesional macrophages and CD4+ T cells in the vessel wall. Mechanistically, miR-181b inhibited the expression of the target gene importin-α3, an effect that reduced NF-κB nuclear translocation specifically in the vascular endothelium of lesions, whereas surprisingly leukocyte NF-κB signaling was unaffected despite a 7-fold overexpression of miR-181b. Our findings uncover that NF-κB nuclear translocation in leukocytes does not involve importin-α3, but rather importin-α5, which miR-181b does not target, highlighting that inhibition of NF-κB signaling in the endothelium is sufficient to mediate miR-181b's protective effects. CONCLUSIONS: Systemic delivery of miR-181b inhibits the activation of NF-κB and atherosclerosis through cell-specific mechanisms in the vascular endothelium. These findings support the rationale that delivery of miR-181b may provide a novel therapeutic approach to treat chronic inflammatory diseases such as atherosclerosis.


Asunto(s)
Aorta/metabolismo , Aterosclerosis/terapia , MicroARNs/uso terapéutico , FN-kappa B/metabolismo , Túnica Íntima/metabolismo , Animales , Aorta/patología , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Linfocitos T CD4-Positivos/metabolismo , Dieta Alta en Grasa/efectos adversos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/administración & dosificación , MicroARNs/sangre , MicroARNs/metabolismo , FN-kappa B/antagonistas & inhibidores , Túnica Íntima/patología
8.
Eur Heart J ; 36(22): 1394-404, 2015 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-25755115

RESUMEN

AIMS: Superficial erosion of atheromata causes many acute coronary syndromes, but arises from unknown mechanisms. This study tested the hypothesis that Toll-like receptor-2 (TLR2) activation contributes to endothelial apoptosis and denudation and thus contributes to the pathogenesis of superficial erosion. METHODS AND RESULTS: Toll-like receptor-2 and neutrophils localized at sites of superficially eroded human plaques. In vitro, TLR2 ligands (including hyaluronan, a matrix macromolecule abundant in eroded lesions) induced endothelial stress, characterized by reactive oxygen species production, endoplasmic reticulum (ER) stress, and apoptosis. Co-incubation of neutrophils with endothelial cells (ECs) potentiated these effects and induced EC apoptosis and detachment. We then categorized human atherosclerotic plaques (n = 56) based on morphologic features associated with superficial erosion, 'stable' fibrotic, or 'vulnerable' lesions. Morphometric analyses of the human atheromata localized neutrophils and neutrophil extracellular traps (NETs) near clusters of apoptotic ECs in smooth muscle cell (SMC)-rich plaques. The number of luminal apoptotic ECs correlated with neutrophil accumulation, amount of NETs, and TLR2 staining in SMC-rich plaques, but not in 'vulnerable' atheromata. CONCLUSION: These in vitro observations and analyses of human plaques indicate that TLR2 stimulation followed by neutrophil participation may render smooth muscle cell-rich plaques susceptible to superficial erosion and thrombotic complications by inducing ER stress, apoptosis, and favouring detachment of EC.


Asunto(s)
Apoptosis/fisiología , Neutrófilos/fisiología , Placa Aterosclerótica/fisiopatología , Receptor Toll-Like 2/fisiología , Síndrome Coronario Agudo/etiología , Síndrome Coronario Agudo/fisiopatología , Biomarcadores/metabolismo , Caspasas/metabolismo , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/metabolismo , Citocinas/metabolismo , Fluoresceínas/farmacología , Colorantes Fluorescentes/farmacología , Humanos , Ácido Hialurónico/farmacología , Elastasa de Leucocito/metabolismo , Leucocitos Mononucleares/metabolismo , Músculo Liso Vascular/metabolismo , Proteína Fosfatasa 1/metabolismo , ARN Interferente Pequeño/fisiología , Especies Reactivas de Oxígeno/metabolismo , Rotura Espontánea/fisiopatología , Estrés Fisiológico/fisiología , Receptor Toll-Like 2/agonistas , Factor de Transcripción CHOP/metabolismo
9.
Nat Cardiovasc Res ; 2(9): 805-818, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39196062

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) is defined by the presence of a cancer-associated somatic mutation in white blood cells in the absence of overt hematological malignancy. It arises most commonly from loss-of-function mutations in the epigenetic regulators DNMT3A and TET2. CHIP predisposes to both hematological malignancies and atherosclerotic cardiovascular disease in humans. Here we demonstrate that loss of Dnmt3a in myeloid cells increased murine atherosclerosis to a similar degree as previously seen with loss of Tet2. Loss of Dnmt3a enhanced inflammation in macrophages in vitro and generated a distinct adventitial macrophage population in vivo which merges a resident macrophage profile with an inflammatory cytokine signature. These changes surprisingly phenocopy the effect of loss of Tet2. Our results identify a common pathway promoting heightened innate immune cell activation with loss of either gene, providing a biological basis for the excess atherosclerotic disease burden in carriers of these two most prevalent CHIP mutations.


Asunto(s)
Aterosclerosis , ADN (Citosina-5-)-Metiltransferasas , ADN Metiltransferasa 3A , Proteínas de Unión al ADN , Dioxigenasas , Modelos Animales de Enfermedad , Mutación con Pérdida de Función , Macrófagos , Fenotipo , Proteínas Proto-Oncogénicas , ADN Metiltransferasa 3A/genética , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/inmunología , Macrófagos/metabolismo , Macrófagos/inmunología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Masculino , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Ratones , Citocinas/metabolismo , Citocinas/genética , Inmunidad Innata/genética , Mediadores de Inflamación/metabolismo , Hematopoyesis Clonal/genética , Femenino
10.
Arterioscler Thromb Vasc Biol ; 31(11): 2464-72, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21903941

RESUMEN

OBJECTIVE: Evidence has linked collagen loss with the onset of acute coronary events. This study tested the hypothesis that selective matrix metalloproteinase-13 (MMP-13) collagenase inhibition increases collagen content in already established and nascent mouse atheromas. METHODS AND RESULTS: In vitro and in situ experiments documented the selectivity and efficacy of an orally available MMP-13 inhibitor (MMP13i-A). In vivo observations monitored macrophage accumulation and MMP-13 activity using molecular imaging. After 10 weeks of MMP13i-A treatment, apolipoprotein E-deficient mice with evolving or established lesions exhibited reduced MMP-13 activity without affecting macrophage content, measured either by intravital microscopy or fluorescence reflectance imaging. Histological analysis indicated that MMP13-iA did not affect plaque size or macrophage or smooth muscle cell accumulation. Administration of MMP13i-A to mice with evolving or established atheromas substantially increased plaque interstitial collagen content in the intima and locally in the fibrous cap, compared with vehicle-treated controls. Analysis of collagen revealed thicker collagen fibers within the plaques of treated groups. CONCLUSION: Pharmacological MMP-13 inhibition yields collagen accumulation in plaques (a feature associated in humans with resistance to rupture), even in established plaques. This study, of considerable clinical relevance, furnishes new mechanistic insight into regulation of the plaque's extracellular matrix and validates molecular imaging for studying plaque biology.


Asunto(s)
Aterosclerosis/metabolismo , Colágeno/metabolismo , Inhibidores Enzimáticos/farmacología , Metaloproteinasa 13 de la Matriz/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/patología , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Modelos Animales de Enfermedad , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología
11.
Arterioscler Thromb Vasc Biol ; 31(2): 261-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21088251

RESUMEN

OBJECTIVE: To examine whether a lack of prostaglandin E receptor 4 (EP4) on bone marrow-derived cells would increase local inflammation and enhance the formation of abdominal aortic aneurysm (AAA) in vivo. METHODS AND RESULTS: Prostaglandin E(2) (PGE(2)) through activation of EP4, can mute inflammation. Hypercholesterolemic low-density lipoprotein receptor knockout (LDLR(-/-)) mice transplanted with either EP4(+/+) (EP4(+/+)/LDLR(-/-)) or EP4(-/-) (EP4(-/-)/LDLR(-/-)) bone marrow received infusions of angiotensin II to induce AAA. Deficiency of EP4 on bone marrow-derived cells increased the incidence (50% of male EP4(+/+)/LDLR(-/-) mice versus 88.9% of male EP4(-/-)/LDLR(-/-) mice developed AAA; and 22% of female EP4(+/+)/LDLR(-/-) mice versus 83.3% of female EP4(-/-)/LDLR(-/-) mice developed AAA) and severity of AAA, increased monocyte chemoattractant protein-1 (2.72-fold in males and 1.64-fold in females), and enhanced infiltration of macrophages (3.8-fold in males and 2.44-fold in females) and T cells (1.88-fold in males and 1.66-fold in females) into AAA lesions. Lack of EP4 on bone marrow-derived cells augmented elastin fragmentation, increased apoptotic markers, and decreased smooth muscle cell accumulation within AAA lesions. CONCLUSIONS: Deficiency of EP4 on bone marrow-derived cells boosted inflammation and AAA formation induced by angiotensin II in hyperlipidemic mice. This study affirms the pathophysiologic importance of PGE(2) signaling through EP4 as an endogenous anti-inflammatory pathway involved in experimental aneurysm formation.


Asunto(s)
Angiotensina II/efectos adversos , Aneurisma de la Aorta Abdominal/inducido químicamente , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Eliminación de Gen , Inflamación/inducido químicamente , Subtipo EP4 de Receptores de Prostaglandina E/genética , Animales , Aneurisma de la Aorta Abdominal/epidemiología , Aneurisma de la Aorta Abdominal/metabolismo , Células de la Médula Ósea/citología , Quimiocina CCL2/metabolismo , Elastina/metabolismo , Femenino , Hiperlipidemias/complicaciones , Hiperlipidemias/metabolismo , Inflamación/epidemiología , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Receptores de LDL/genética , Receptores de LDL/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Factores de Riesgo
12.
Cardiovasc Res ; 117(13): 2652-2663, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-33751034

RESUMEN

AIMS: Recent evidence suggests that 'vulnerable plaques', which have received intense attention as underlying mechanism of acute coronary syndromes over the decades, actually rarely rupture and cause clinical events. Superficial plaque erosion has emerged as a growing cause of residual thrombotic complications of atherosclerosis in an era of increased preventive measures including lipid lowering, antihypertensive therapy, and smoking cessation. The mechanisms of plaque erosion remain poorly understood, and we currently lack validated effective diagnostics or therapeutics for superficial erosion. Eroded plaques have a rich extracellular matrix, an intact fibrous cap, sparse lipid, and few mononuclear cells, but do harbour neutrophil extracellular traps (NETs). We recently reported that NETs amplify and propagate the endothelial damage at the site of arterial lesions that recapitulate superficial erosion in mice. We showed that genetic loss of protein arginine deiminase (PAD)-4 function inhibited NETosis and preserved endothelial integrity. The current study used systemic administration of targeted nanoparticles to deliver an agent that limits NETs formation to probe mechanisms of and demonstrate a novel therapeutic approach to plaque erosion that limits endothelial damage. METHODS AND RESULTS: We developed Collagen IV-targeted nanoparticles (Col IV NP) to deliver PAD4 inhibitors selectively to regions of endothelial cell sloughing and collagen IV-rich basement membrane exposure. We assessed the binding capability of the targeting ligand in vitro and evaluated Col IV NP targeting to areas of denuded endothelium in vivo in a mouse preparation that recapitulates features of superficial erosion. Delivery of the PAD4 inhibitor GSK484 reduced NET accumulation at sites of intimal injury and preserved endothelial continuity. CONCLUSIONS: NPs directed to Col IV show selective uptake and delivery of their payload to experimentally eroded regions, illustrating their translational potential. Our results further support the role of PAD4 and NETs in superficial erosion.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Colágeno Tipo IV/metabolismo , Portadores de Fármacos , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Trampas Extracelulares/metabolismo , Nanopartículas , Arginina Deiminasa Proteína-Tipo 4/antagonistas & inhibidores , Animales , Aterosclerosis/enzimología , Aterosclerosis/patología , Membrana Basal/metabolismo , Técnicas de Cultivo Tridimensional de Células , Células Cultivadas , Colágeno Tipo IV/química , Modelos Animales de Enfermedad , Composición de Medicamentos , Liberación de Fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Ratones Noqueados para ApoE , Nanotecnología , Placa Aterosclerótica , Unión Proteica , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Propiedades de Superficie , Distribución Tisular
13.
Sci Transl Med ; 12(531)2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075942

RESUMEN

Long noncoding RNAs (lncRNAs) are emerging regulators of biological processes in the vessel wall; however, their role in atherosclerosis remains poorly defined. We used RNA sequencing to profile lncRNAs derived specifically from the aortic intima of Ldlr -/- mice on a high-cholesterol diet during lesion progression and regression phases. We found that the evolutionarily conserved lncRNA small nucleolar host gene-12 (SNHG12) is highly expressed in the vascular endothelium and decreases during lesion progression. SNHG12 knockdown accelerated atherosclerotic lesion formation by 2.4-fold in Ldlr -/- mice by increased DNA damage and senescence in the vascular endothelium, independent of effects on lipid profile or vessel wall inflammation. Conversely, intravenous delivery of SNHG12 protected the tunica intima from DNA damage and atherosclerosis. LncRNA pulldown in combination with liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis showed that SNHG12 interacted with DNA-dependent protein kinase (DNA-PK), an important regulator of the DNA damage response. The absence of SNHG12 reduced the DNA-PK interaction with its binding partners Ku70 and Ku80, abrogating DNA damage repair. Moreover, the anti-DNA damage agent nicotinamide riboside (NR), a clinical-grade small-molecule activator of NAD+, fully rescued the increases in lesional DNA damage, senescence, and atherosclerosis mediated by SNHG12 knockdown. SNHG12 expression was also reduced in pig and human atherosclerotic specimens and correlated inversely with DNA damage and senescent markers. These findings reveal a role for this lncRNA in regulating DNA damage repair in the vessel wall and may have implications for chronic vascular disease states and aging.


Asunto(s)
Daño del ADN , Proteína Quinasa Activada por ADN , Endotelio Vascular/patología , ARN Largo no Codificante , Animales , Movimiento Celular , Proliferación Celular , Cromatografía Liquida , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Proteínas Quinasas , ARN Largo no Codificante/genética , Porcinos , Espectrometría de Masas en Tándem
14.
J Am Coll Cardiol ; 58(6): 603-14, 2011 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-21798423

RESUMEN

OBJECTIVES: This study investigated the regulation of glucose uptake in cells that participate in atherogenesis by stimuli relevant to this process, to gain mechanistic insight into the origin of the (18)fluorine-labeled 2-deoxy-D-glucose (FdG) uptake signals observed clinically. BACKGROUND: Patient studies suggest that positron emission tomography (PET) using FdG can detect "active" atherosclerotic plaques, yet the mechanism giving rise to FdG signals remains unknown. METHODS: We exposed cells to conditions thought to operate in atheroma and determined rates of glucose uptake. RESULTS: Hypoxia, but not pro-inflammatory cytokines, potently stimulated glucose uptake in human macrophages and foam cells. Statins attenuated this process in vitro, suggesting that these agents have a direct effect on human macrophages. Immunohistochemical study of human plaques revealed abundant expression of proteins regulating glucose utilization, predominantly in macrophage-rich regions of the plaques-regions previously proved hypoxic. Smooth-muscle cells and endothelial cells markedly increased rates of glucose uptake when exposed to pro-inflammatory cytokines. CONCLUSIONS: Glucose uptake and, probably, FdG uptake signals in atheroma may reflect hypoxia-stimulated macrophages rather than mere inflammatory burden. Cytokine-activated smooth-muscle cells also may contribute to the FdG signal.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Aterosclerosis/terapia , Glucosa/farmacocinética , Hipoxia , Inflamación , Macrófagos/efectos de los fármacos , Aorta/patología , Aterosclerosis/patología , Citocinas/metabolismo , Células Endoteliales/citología , Fluorodesoxiglucosa F18/farmacología , Células Espumosas/metabolismo , Glucosa/metabolismo , Humanos , Hidrólisis , Macrófagos/metabolismo , Monocitos/citología , Miocitos del Músculo Liso/metabolismo , Tomografía de Emisión de Positrones/métodos
15.
Cardiovasc Res ; 89(1): 234-43, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20736236

RESUMEN

AIM: prostaglandin E(2), by ligation of its receptor EP4, suppresses the production of inflammatory cytokines and chemokines in macrophages in vitro. Thus, activation of EP4 may constitute an endogenous anti-inflammatory pathway. This study investigated the role of EP4 in atherosclerosis in vivo, and particularly its impact on inflammation. METHODS AND RESULTS: Ldlr(-/-) mice transplanted with EP4(+/+) or EP4(-/-) bone marrow consumed a high-fat diet for 5 or 10 weeks. Allogenic bone marrow transplantation promoted exacerbation of atherosclerosis irrespective of EP4 genotype, compatible with prior observations of exacerbated atherogenesis by allogenicity. EP4 deficiency had little effect on plaque size or morphology in early atherosclerosis, but at the later time point, mice deficient in EP4 displayed enhanced inflammation in their atherosclerotic plaques. Expression of monocyte chemoattractant protein-1 and interferon-γ inducible protein 10 increased, and there was a corresponding increase in macrophage and T-cell infiltration. These plaques also contained fewer smooth muscle cells. Despite these changes, mice deficient in EP4 in bone marrow-derived cells at an advanced stage had similar lesion size (in both aorta and aortic root) as mice with EP4. CONCLUSION: this study shows that in advanced atherosclerosis, EP4 deficiency did not alter atherosclerotic lesion size, but yielded plaques with exacerbated inflammation and altered lesion composition.


Asunto(s)
Células de la Médula Ósea/metabolismo , Placa Aterosclerótica/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/deficiencia , Animales , Apoptosis , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Quimiocina CCL2/genética , Quimiocina CXCL10/genética , Cartilla de ADN/genética , Femenino , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Modelos Cardiovasculares , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Receptores de LDL/deficiencia , Receptores de LDL/genética , Subtipo EP4 de Receptores de Prostaglandina E/genética , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA