Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 605(7908): 152-159, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35477759

RESUMEN

Atherosclerotic plaques develop in the inner intimal layer of arteries and can cause heart attacks and strokes1. As plaques lack innervation, the effects of neuronal control on atherosclerosis remain unclear. However, the immune system responds to plaques by forming leukocyte infiltrates in the outer connective tissue coat of arteries (the adventitia)2-6. Here, because the peripheral nervous system uses the adventitia as its principal conduit to reach distant targets7-9, we postulated that the peripheral nervous system may directly interact with diseased arteries. Unexpectedly, widespread neuroimmune cardiovascular interfaces (NICIs) arose in mouse and human atherosclerosis-diseased adventitia segments showed expanded axon networks, including growth cones at axon endings near immune cells and media smooth muscle cells. Mouse NICIs established a structural artery-brain circuit (ABC): abdominal adventitia nociceptive afferents10-14 entered the central nervous system through spinal cord T6-T13 dorsal root ganglia and were traced to higher brain regions, including the parabrachial and central amygdala neurons; and sympathetic efferent neurons projected from medullary and hypothalamic neurons to the adventitia through spinal intermediolateral neurons and both coeliac and sympathetic chain ganglia. Moreover, ABC peripheral nervous system components were activated: splenic sympathetic and coeliac vagus nerve activities increased in parallel to disease progression, whereas coeliac ganglionectomy led to the disintegration of adventitial NICIs, reduced disease progression and enhanced plaque stability. Thus, the peripheral nervous system uses NICIs to assemble a structural ABC, and therapeutic intervention in the ABC attenuates atherosclerosis.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Aterosclerosis/prevención & control , Progresión de la Enfermedad , Ganglios Espinales , Ganglios Simpáticos , Ratones , Neuronas/fisiología , Placa Aterosclerótica/prevención & control
2.
Eur Heart J ; 43(16): 1533-1541, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-34897403

RESUMEN

The increasing use of single-cell immune profiling and advanced microscopic imaging technologies has deepened our understanding of the cardiac immune system, confirming that the heart contains a broad repertoire of innate and adaptive immune cells. Leucocytes found in the healthy heart participate in essential functions to preserve cardiac homeostasis, not only by defending against pathogens but also by maintaining normal organ function. In pathophysiological conditions, cardiac inflammation is implicated in healing responses after ischaemic or non-ischaemic cardiac injury. The aim of this review is to provide a concise overview of novel methodological advancements to the non-expert readership and summarize novel findings on immune cell heterogeneity and functions in cardiac disease with a focus on myocardial infarction as a prototypic example. In addition, we will briefly discuss how biological sex modulate the cardiac immune response. Finally, we will highlight emerging concepts for novel therapeutic applications, such as targeting immunometabolism and nanomedicine.


Asunto(s)
Cardiopatías , Infarto del Miocardio , Corazón , Homeostasis , Humanos , Cicatrización de Heridas
3.
Am J Physiol Cell Physiol ; 320(5): C761-C770, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33596150

RESUMEN

Monocytes are a heterogeneous cell population of innate immune cells with distinct cell surface markers that help them in carrying out different functions. In humans, there are three well-characterized subsets, namely, classical (CD14++CD16-), intermediate (CD14++CD16+), and nonclassical (CD14+ CD16++) monocytes. There is an emerging focus on the not yet well explored nonclassical monocytes that maintain vascular integrity by slowly patrolling on the endothelium, reacting to inflammatory signals, and clearing cell debris. In this manner, they are not only crucial for vascular homeostasis but also play a vital role in wound healing and resolution of inflammation by linking innate to adaptive immune response. Although they have been shown to be protective, yet they are also associated with inflammatory disease progression. This short review will give an insight about the emerging role of nonclassical monocytes in vascular homeostasis, inflammation, and protection in the context of cardiovascular disease.


Asunto(s)
Inmunidad Adaptativa , Enfermedades Cardiovasculares/inmunología , Células Endoteliales/inmunología , Endotelio Vascular/inmunología , Inmunidad Innata , Inflamación/inmunología , Monocitos/inmunología , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Monocitos/metabolismo , Monocitos/patología , Fenotipo , Transducción de Señal
4.
Eur Heart J ; 40(48): 3937-3946, 2019 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-31121017

RESUMEN

The outcomes of the Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) trial have unequivocally proven that inflammation is a key driver of atherosclerosis and that targeting inflammation, in this case by using an anti-interleukin-1ß antibody, improves cardiovascular disease (CVD) outcomes. This is especially true for CVD patients with a pro-inflammatory constitution. Although CANTOS has epitomized the importance of targeting inflammation in atherosclerosis, treatment with canakinumab did not improve CVD mortality, and caused an increase in infections. Therefore, the identification of novel drug targets and development of novel therapeutics that block atherosclerosis-specific inflammatory pathways and exhibit limited immune-suppressive side effects, as pursued in our collaborative research centre, are required to optimize immunotherapy for CVD. In this review, we will highlight the potential of novel immunotherapeutic targets that are currently considered to become a future treatment for CVD.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Citocinas/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Interleucina-1beta/antagonistas & inhibidores , Antiinflamatorios/uso terapéutico , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/uso terapéutico , Aterosclerosis/complicaciones , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Enfermedades Cardiovasculares/mortalidad , Enfermedad de la Arteria Coronaria/prevención & control , Citocinas/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Países Bajos/epidemiología , Ensayos Clínicos Controlados Aleatorios como Asunto
5.
Circulation ; 137(9): 948-960, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29167227

RESUMEN

BACKGROUND: The pericardial adipose tissue (AT) contains a high density of lymphoid clusters. It is unknown whether these clusters play a role in post-myocardial infarction (MI) inflammatory responses and cardiac outcome. METHODS: Lymphoid clusters were examined in epicardial AT of humans with or without coronary artery disease. Murine pericardial lymphoid clusters were visualized in mice subjected to coronary artery ligation. To study the relevance of pericardial clusters during inflammatory responses after MI, we surgically removed the pericardial AT and performed B-cell depletion and granulocyte-macrophage colony-stimulating factor blockade. Leukocytes in murine hearts, pericardial AT, spleen, mediastinal lymph nodes, and bone marrow were quantified by flow cytometry. Cannabinoid receptor CB2 (CB2-/-) mice were used as a model for enhanced B-cell responses. The effect of impaired dendritic cell (DC) trafficking on pericardial AT inflammatory responses was tested in CCR7-/- mice subjected to MI. Cardiac fibrosis and ventricular function were assessed by histology and echocardiography. RESULTS: We identified larger B-cell clusters in epicardial AT of human patients with coronary artery disease in comparison with controls without coronary artery disease. Infarcted mice also had larger pericardial clusters and 3-fold upregulated numbers of granulocyte-macrophage colony-stimulating factor-producing B cells within pericardial AT, but not spleen or lymph nodes. This was associated with higher DC and T-cell counts in pericardial AT, which outnumbered DCs and T cells in lymph nodes. Analysis of DC maturation markers, tracking experiments with fluorescently labeled cells, and use of CCR7-deficient mice suggested that activated DCs migrate from infarcts into pericardial AT via CCR7. B-cell depletion or granulocyte-macrophage colony-stimulating factor neutralization inhibited DC and T-cell expansion within pericardial AT, and translated into reduced bone marrow granulopoiesis and cardiac neutrophil infiltration 3 days after MI. The relevance of the pericardial AT in mediating all these effects was confirmed by removal of pericardial AT and ex vivo coculture with pericardial AT and granulocyte progenitors. Finally, enhanced fibrosis and worsened ejection fraction in CB2-/- mice were limited by pericardial AT removal. CONCLUSIONS: Our findings unveil a new mechanism by which the pericardial AT coordinates immune cell activation, granulopoiesis, and outcome after MI.


Asunto(s)
Tejido Adiposo/fisiología , Granulocitos/inmunología , Infarto del Miocardio/patología , Miocardio/patología , Pericardio/patología , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/metabolismo , Receptor Cannabinoide CB2/genética , Receptores CCR7/genética , Cicatrización de Heridas
6.
J Transl Med ; 17(1): 180, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142323

RESUMEN

BACKGROUND: ST-segment elevation myocardial infarction (STEMI) displays circadian variability with the highest incidence in the morning hours. Data on whether the time-of-day at symptom onset affects infarct size or patients' long-term prognosis are conflicting. We sought to investigate the association of time-of-day at symptom onset with infarct size or long-term mortality in patients with STEMI undergoing primary percutaneous coronary intervention (PPCI). METHODS: This study included 1206 STEMI patients undergoing PPCI. All patients underwent single photon emission computed tomography (SPECT) imaging with 99mTc-sestamibi before and 7-14 days after PPCI. The co-primary endpoints were final infarct size on day 10 after STEMI and all-cause mortality at 5-year follow-up. Time-of-day at symptom onset of STEMI was categorized in 6-h intervals. RESULTS: In patients presenting from 0 to 6 h, 6 to 12 h, 12 to 18 h, and 18 to 24 h, the infarct sizes (median [25th-75th percentiles]) were 10.0 [3.0-24.7], 10.0 [3.0-24.0], 10.0 [3.0-22.0], and 9.0 [3.0-21.0] of the left ventricle, respectively (p = 0.87); the Kaplan-Meier estimates of 5-year all-cause mortality were 13.6%, 8.7%, 13.7% and 9.3%, respectively (log-rank test p = 0.30). After adjustment, time-of-day was not associated with infarct size (p ≥ 0.76 for comparisons with infarct size from reference [6-12 h] time interval) or 5-year all-cause mortality (p ≥ 0.25 for comparisons with mortality from reference [6-12 h] time interval). Time-of-day at symptom onset of STEMI was not associated with differences in the recovery of left ventricular ejection fraction 6 months after STEMI. CONCLUSIONS: In patients with STEMI undergoing PPCI, time-of-day at symptom onset was neither associated with scintigraphic infarct size, left ventricular ejection fraction recovery at 6 months nor with 5-year mortality.


Asunto(s)
Ritmo Circadiano/fisiología , Miocardio/patología , Intervención Coronaria Percutánea , Infarto del Miocardio con Elevación del ST/diagnóstico , Infarto del Miocardio con Elevación del ST/cirugía , Tiempo de Tratamiento , Adulto , Relojes Biológicos/fisiología , Femenino , Estudios de Seguimiento , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Tamaño de los Órganos/fisiología , Pronóstico , Estudios Retrospectivos , Infarto del Miocardio con Elevación del ST/mortalidad , Infarto del Miocardio con Elevación del ST/fisiopatología , Volumen Sistólico/fisiología , Análisis de Supervivencia , Factores de Tiempo , Tomografía Computarizada de Emisión de Fotón Único , Resultado del Tratamiento , Función Ventricular Izquierda
7.
Arterioscler Thromb Vasc Biol ; 38(2): 313-323, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29284608

RESUMEN

OBJECTIVE: The MC1-R (melanocortin 1 receptor) is expressed by monocytes and macrophages where it mediates anti-inflammatory actions. MC1-R also protects against macrophage foam cell formation primarily by promoting cholesterol efflux through the ABCA1 (ATP-binding cassette transporter subfamily A member 1) and ABCG1 (ATP-binding cassette transporter subfamily G member 1). In this study, we aimed to investigate whether global deficiency in MC1-R signaling affects the development of atherosclerosis. APPROACH AND RESULTS: Apoe-/- (apolipoprotein E deficient) mice were crossed with recessive yellow (Mc1re/e) mice carrying dysfunctional MC1-R and fed a high-fat diet to induce atherosclerosis. Apoe-/- Mc1re/e mice developed significantly larger atherosclerotic lesions in the aortic sinus and in the whole aorta compared with Apoe-/- controls. In terms of plaque composition, MC1-R deficiency was associated with less collagen and smooth muscle cells and increased necrotic core, indicative of more vulnerable lesions. These changes were accompanied by reduced Abca1 and Abcg1 expression in the aorta. Furthermore, Apoe-/- Mc1re/e mice showed a defect in bile acid metabolism that aggravated high-fat diet-induced hypercholesterolemia and hepatic lipid accumulation. Flow cytometric analysis of leukocyte profile revealed that dysfunctional MC1-R enhanced arterial accumulation of classical Ly6Chigh monocytes and macrophages, effects that were evident in mice fed a normal chow diet but not under high-fat diet conditions. In support of enhanced arterial recruitment of Ly6Chigh monocytes, these cells had increased expression of L-selectin and P-selectin glycoprotein ligand 1. CONCLUSIONS: The present study highlights the importance of MC1-R in the development of atherosclerosis. Deficiency in MC1-R signaling exacerbates atherosclerosis by disturbing cholesterol handling and by increasing arterial monocyte accumulation.


Asunto(s)
Aorta/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Ratones Noqueados para ApoE , Monocitos/metabolismo , Placa Aterosclerótica , Receptor de Melanocortina Tipo 1/deficiencia , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Colesterol/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Monocitos/patología , Receptor de Melanocortina Tipo 1/genética
8.
Arterioscler Thromb Vasc Biol ; 38(11): 2562-2575, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30354245

RESUMEN

Objective- Palmitoylethanolamide is an endogenous fatty acid mediator that is synthetized from membrane phospholipids by N-acyl phosphatidylethanolamine phospholipase D. Its biological actions are primarily mediated by PPAR-α (peroxisome proliferator-activated receptors α) and the orphan receptor GPR55. Palmitoylethanolamide exerts potent anti-inflammatory actions but its physiological role and promise as a therapeutic agent in chronic arterial inflammation, such as atherosclerosis remain unexplored. Approach and Results- First, the polarization of mouse primary macrophages towards a proinflammatory phenotype was found to reduce N-acyl phosphatidylethanolamine phospholipase D expression and palmitoylethanolamide bioavailability. N-acyl phosphatidylethanolamine phospholipase D expression was progressively downregulated in the aorta of apolipoprotein E deficient (ApoE-/-) mice during atherogenesis. N-acyl phosphatidylethanolamine phospholipase D mRNA levels were also downregulated in unstable human plaques and they positively associated with smooth muscle cell markers and negatively with macrophage markers. Second, ApoE-/- mice were fed a high-fat diet for 4 or 16 weeks and treated with either vehicle or palmitoylethanolamide (3 mg/kg per day, 4 weeks) to study the effects of palmitoylethanolamide on early established and pre-established atherosclerosis. Palmitoylethanolamide treatment reduced plaque size in early atherosclerosis, whereas in pre-established atherosclerosis, palmitoylethanolamide promoted signs of plaque stability as evidenced by reduced macrophage accumulation and necrotic core size, increased collagen deposition and downregulation of M1-type macrophage markers. Mechanistically, we found that palmitoylethanolamide, by activating GPR55, increases the expression of the phagocytosis receptor MerTK (proto-oncogene tyrosine-protein kinase MER) and enhances macrophage efferocytosis, indicative of proresolving properties. Conclusions- The present study demonstrates that palmitoylethanolamide protects against atherosclerosis by promoting an anti-inflammatory and proresolving phenotype of lesional macrophages, representing a new therapeutic approach to resolve arterial inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Etanolaminas/farmacología , Macrófagos/efectos de los fármacos , Ácidos Palmíticos/farmacología , Fagocitosis/efectos de los fármacos , Placa Aterosclerótica , Amidas , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Fenotipo , Fosfolipasa D/metabolismo , Proto-Oncogenes Mas , Ratas , Receptores de Cannabinoides/genética , Receptores de Cannabinoides/metabolismo , Factores de Tiempo , Tirosina Quinasa c-Mer/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 38(5): 1007-1019, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29567680

RESUMEN

OBJECTIVE: Cardiovascular diseases and depression are the leading causes of disability in Western countries. Clinical data on potential cardiovascular effects of serotonin reuptake inhibitors (SSRIs), the most commonly used antidepressant drugs, are controversial. In addition to blocking serotonin reuptake transporter in the brain, SSRIs deplete the major peripheral serotonin (5-hydroxytryptamine [5-HT]) storage by inhibiting serotonin reuptake transporter-mediated uptake in platelets. In this study, we aimed to investigate the effect of chronic SSRI intake on the development of atherosclerosis. APPROACH AND RESULTS: Treatment of apolipoprotein E-deficient mice with the SSRI fluoxetine for 2, 4, or 16 weeks increased atherosclerotic lesion formation, with most pronounced effect during early plaque development. Intravital microscopy of carotid arteries revealed enhanced myeloid cell adhesion on fluoxetine treatment. Mechanistically, we found that fluoxetine augmented vascular permeability and increased chemokine-induced integrin-binding activity of circulating leukocytes. In vitro stimulation of murine blood demonstrated that fluoxetine, but not 5-HT, could directly promote ß1 and ß2 integrin activation provided C-C motif chemokine ligand 5 was also present. Similar effects were observed with the SSRI escitalopram. Enhanced C-C motif chemokine ligand 5-induced integrin activation by fluoxetine was also confirmed in a human neutrophil-like cell line. In contrast to the proatherogenic properties of fluoxetine, pharmacological inhibition of the peripheral 5-HT synthesizing enzyme tryptophan hydroxylase 1 did not promote atherosclerosis, suggesting that the proatherogenic effect of fluoxetine occurs independent of peripheral 5-HT depletion. CONCLUSIONS: SSRI intake may promote atherosclerosis and therefore potentially increase the risk for acute cardiovascular events by a mechanism that is independent of 5-HT depletion.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/inducido químicamente , Aterosclerosis/inducido químicamente , Arterias Carótidas/efectos de los fármacos , Enfermedades de las Arterias Carótidas/inducido químicamente , Fluoxetina/toxicidad , Placa Aterosclerótica , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Animales , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/sangre , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Antígenos CD18/sangre , Permeabilidad Capilar/efectos de los fármacos , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Adhesión Celular/efectos de los fármacos , Quimiocina CCL5/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Esquema de Medicación , Fluoxetina/administración & dosificación , Células HEK293 , Células HL-60 , Humanos , Integrina beta1/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Serotonina/sangre , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Transducción de Señal , Factores de Tiempo
10.
Circulation ; 136(1): 83-97, 2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28450348

RESUMEN

BACKGROUND: The melanocortin 1 receptor (MC1-R) is expressed by monocytes and macrophages, where it exerts anti-inflammatory actions on stimulation with its natural ligand α-melanocyte-stimulating hormone. The present study was designed to investigate the specific role of MC1-R in the context of atherosclerosis and possible regulatory pathways of MC1-R beyond anti-inflammation. METHODS: Human and mouse atherosclerotic samples and primary mouse macrophages were used to study the regulatory functions of MC1-R. The impact of pharmacological MC1-R activation on atherosclerosis was assessed in apolipoprotein E-deficient mice. RESULTS: Characterization of human and mouse atherosclerotic plaques revealed that MC1-R expression localizes in lesional macrophages and is significantly associated with the ATP-binding cassette transporters ABCA1 and ABCG1, which are responsible for initiating reverse cholesterol transport. Using bone marrow-derived macrophages, we observed that α-melanocyte-stimulating hormone and selective MC1-R agonists similarly promoted cholesterol efflux, which is a counterregulatory mechanism against foam cell formation. Mechanistically, MC1-R activation upregulated the levels of ABCA1 and ABCG1. These effects were accompanied by a reduction in cell surface CD36 expression and in cholesterol uptake, further protecting macrophages from excessive lipid accumulation. Conversely, macrophages deficient in functional MC1-R displayed a phenotype with impaired efflux and enhanced uptake of cholesterol. Pharmacological targeting of MC1-R in atherosclerotic apolipoprotein E-deficient mice reduced plasma cholesterol levels and aortic CD36 expression and increased plaque ABCG1 expression and signs of plaque stability. CONCLUSIONS: Our findings identify a novel role for MC1-R in macrophage cholesterol transport. Activation of MC1-R confers protection against macrophage foam cell formation through a dual mechanism: It prevents cholesterol uptake while concomitantly promoting ABCA1- and ABCG1-mediated reverse cholesterol transport.


Asunto(s)
Colesterol/metabolismo , Macrófagos/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Transducción de Señal/fisiología , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Femenino , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Células HEK293 , Humanos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Receptor de Melanocortina Tipo 1/agonistas , Transducción de Señal/efectos de los fármacos , alfa-MSH/metabolismo , alfa-MSH/farmacología
11.
Arterioscler Thromb Vasc Biol ; 37(6): 1022-1028, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28450299

RESUMEN

Physiological cardiovascular functions show daily diurnal variations, which are synchronized by intrinsic molecular clocks and environment-driven cues. The clinical manifestation of cardiovascular disease also exhibits diurnal variation, with an increased incidence in the early morning. This coincides with circadian oscillations of circulating parameters, such as hormones and leukocyte counts. We are just at the beginning of understanding how circadian rhythms of immune functions are related to cardiovascular disease progression and outcome after an acute ischemic event. Here, we briefly summarize clinical data on oscillations of circulating inflammatory parameters, as well as experimental evidences for the role of circadian clocks in atherosclerosis, postmyocardial infarction inflammatory responses, and cardiac healing.


Asunto(s)
Aterosclerosis/fisiopatología , Relojes Circadianos , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Ritmo Circadiano , Mediadores de Inflamación/metabolismo , Inflamación/fisiopatología , Infarto del Miocardio/fisiopatología , Animales , Aterosclerosis/complicaciones , Aterosclerosis/metabolismo , Hematopoyesis , Humanos , Inflamación/complicaciones , Inflamación/metabolismo , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal , Factores de Tiempo , Cicatrización de Heridas
12.
Arterioscler Thromb Vasc Biol ; 37(2): 247-257, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28062509

RESUMEN

OBJECTIVE: ADAM17 (a disintegrin and metalloproteinase 17) is a sheddase releasing different types of membrane-bound proteins, including adhesion molecules, cytokines, and their receptors as well as inflammatory mediators. Because these substrates modulate important mechanisms of atherosclerosis, we hypothesized that ADAM17 might be involved in the pathogenesis of this frequent disease. APPROACH AND RESULTS: Because Adam17-knockout mice are not viable, we studied the effect of Adam17 deficiency on atherosclerosis in Adam17 hypomorphic mice (Adam17ex/ex), which have low residual Adam17 expression. To induce atherosclerosis, mice were crossed onto the low-density lipoprotein receptor (Ldlr)-deficient background. We found that Adam17ex/ex.Ldlr-/- mice developed ≈1.5-fold larger atherosclerotic lesions, which contained more macrophages and vascular smooth muscle cells than wild-type littermate controls (Adam17wt/wt.Ldlr-/-). Reduced Adam17-mediated shedding led to significantly increased protein levels of membrane-resident TNFα (tumor necrosis factor) and TNFR2 (tumor necrosis factor receptor 2), resulting in a constitutive activation of TNFR2 signaling. At the same time, Adam17 deficiency promoted proatherosclerotic cellular functions, such as increased proliferation and reduced apoptosis in cultured macrophages and vascular smooth muscle cells and increased adhesion of macrophages to vascular endothelial cells. Because siRNA (small interfering RNA)-mediated knockdown of Tnfr2 rescued from aberrant proliferation and from misregulation of apoptosis in Adam17-depleted cells, our data indicate that TNFR2 is an important effector of ADAM17 in our mouse model. CONCLUSIONS: Our results provide evidence for an atheroprotective role of ADAM17, which might be mediated by cleaving membrane-bound TNFα and TNFR2, thereby preventing overactivation of endogenous TNFR2 signaling in cells of the vasculature.


Asunto(s)
Proteína ADAM17/deficiencia , Aorta/enzimología , Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Proteína ADAM17/genética , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Adhesión Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Células Endoteliales/patología , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Fenotipo , Placa Aterosclerótica , Interferencia de ARN , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal , Factores de Tiempo , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 37(9): 1640-1645, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28663258

RESUMEN

OBJECTIVE: Circadian regulation of neutrophil homeostasis affects myocardial infarction (MI) healing. It is unknown whether diurnal variations of monocyte counts exist in the heart and whether this affects their cardiac infiltration in response to MI. APPROACH AND RESULTS: Murine blood and organs were harvested at distinct times of day and analyzed by flow cytometry. Ly6Chigh monocyte surface expression levels of chemokine receptors (CCR) were ≈2-fold higher at the beginning of the active phase, Zeitgeber Time (ZT) 13 compared with ZT5. This was because of enhanced receptor surface expression at ZT13, whereas no significant changes in total cellular protein levels were found. Most blood Ly6Chigh monocytes were CCR2high, whereas only a minority was CCR1high and CCR5high. We also found diurnal changes of classical monocyte blood counts in humans, being higher in the evening, while exhibiting enhanced CCR2 surface expression in the morning. In support of monocyte oscillations between blood and tissue, murine cardiac Ly6Chigh monocyte counts were highest at ZT13, accompanied by an upregulation of cardiac CC chemokine ligand 2 mRNA. Mice subjected to MI at ZT13 had an even higher upregulation of CCR2 surface expression on circulating monocytes compared with noninfarcted mice and more elevated cardiac CC chemokine ligand 2 protein expression and more pronounced Ly6Chigh monocyte infiltration compared with ZT5-infarcted mice. Concomitantly, CCR2 antagonism only inhibited the excessive cardiac Ly6Chigh monocyte infiltration after ZT13 MI but not ZT5 MI. CONCLUSIONS: CCR2 surface expression on Ly6Chigh monocytes changes in a time-of-day-dependent manner, which crucially affects cardiac monocyte recruitment after an acute ischemic event.


Asunto(s)
Antígenos Ly/inmunología , Quimiotaxis de Leucocito , Ritmo Circadiano , Monocitos/inmunología , Infarto del Miocardio/inmunología , Miocardio/inmunología , Adulto , Animales , Antígenos Ly/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Inmunofenotipificación/métodos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Monocitos/metabolismo , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Fenotipo , Receptores CCR1/inmunología , Receptores CCR1/metabolismo , Receptores CCR2/genética , Receptores CCR2/inmunología , Receptores CCR2/metabolismo , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Transducción de Señal , Factores de Tiempo , Adulto Joven
14.
Cell Mol Life Sci ; 74(2): 359-372, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27622243

RESUMEN

MicroRNAs (miRNAs) coordinate vascular repair by regulating injury-induced gene expression in vascular smooth muscle cells (SMCs) and promote the transition of SMCs from a contractile to a proliferating phenotype. However, the effect of miRNA expression in SMCs on neointima formation is unclear. Therefore, we studied the role of miRNA biogenesis by Dicer in SMCs in vascular repair. Following wire-induced injury to carotid arteries of Apolipoprotein E knockout (Apoe -/-) mice, miRNA microarray analysis revealed that the most significantly regulated miRNAs, such as miR-222 and miR-21-3p, were upregulated. Conditional deletion of Dicer in SMCs increased neointima formation by reducing SMC proliferation in Apoe -/- mice, and decreased mainly the expression of miRNAs, such as miR-147 and miR-100, which were not upregulated following vascular injury. SMC-specific deletion of Dicer promoted growth factor and inflammatory signaling and regulated a miRNA-target interaction network in injured arteries that was enriched in anti-proliferative miRNAs. The most connected miRNA in this network was miR-27a-3p [e.g., with Rho guanine nucleotide exchange factor 26 (ARHGEF26)], which was expressed in medial and neointimal SMCs in a Dicer-dependent manner. In vitro, miR-27a-3p suppresses ARHGEF26 expression and inhibits SMC proliferation by interacting with a conserved binding site in the 3' untranslated region of ARHGEF26 mRNA. We propose that Dicer expression in SMCs plays an essential role in vascular repair by generating anti-proliferative miRNAs, such as miR-27a-3p, to prevent vessel stenosis due to exaggerated neointima formation.


Asunto(s)
Redes Reguladoras de Genes , MicroARNs/genética , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Neointima/genética , Ribonucleasa III/metabolismo , Cicatrización de Heridas/genética , Animales , Arterias/metabolismo , Arterias/patología , Proliferación Celular , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , MicroARNs/metabolismo , Miocitos del Músculo Liso/patología , Neointima/metabolismo , Especificidad de Órganos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo
15.
Eur Heart J ; 38(3): 187-197, 2017 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-28158426

RESUMEN

Aims: Acute myocardial infarction (MI) is the leading cause of mortality worldwide. Anti-inflammatory strategies to reduce neutrophil-driven acute post-MI injury have been shown to limit acute cardiac tissue damage. On the other hand, whether neutrophils are required for resolving post-MI inflammation and repair is unknown. Methods and Results: We show that neutrophil-depleted mice subjected to MI had worsened cardiac function, increased fibrosis, and progressively developed heart failure. Flow cytometry of blood, lymphoid organs and digested hearts revealed reduced numbers of Ly6Chigh monocytes in infarcts of neutrophil-depleted mice, whereas the number of macrophages increased, which was paralleled by reduced splenic Ly6Chigh monocyte mobilization but enhanced proliferation of cardiac macrophages. Macrophage subtype analysis revealed reduced cardiac expression of M1 markers, whereas M2 markers were increased in neutrophil-depleted mice. Surprisingly, we found reduced expression of phagocytosis receptor myeloid-epithelial-reproductive tyrosine kinase, a marker of reparative M2c macrophages which mediate clearance of apoptotic cells. In agreement with this finding, neutrophil-depleted mice had increased numbers of TUNEL-positive cells within infarcts. We identified neutrophil gelatinase-associated lipocalin (NGAL) in the neutrophil secretome as a key inducer of macrophages with high capacity to engulf apoptotic cells. The cardiac macrophage phenotype in neutrophil-depleted mice was restored by administration of neutrophil secretome or NGAL. Conclusion: Neutrophils are crucially involved in cardiac repair after MI by polarizing macrophages towards a reparative phenotype. Therapeutic strategies to reduce acute neutrophil-driven inflammation after MI should be carefully balanced as they might interfere with the healing response and cardiac remodelling.


Asunto(s)
Macrófagos/fisiología , Infarto del Miocardio/fisiopatología , Neutrófilos/fisiología , Animales , Apoptosis/fisiología , Proliferación Celular/fisiología , Fibrosis Endomiocárdica/etiología , Fibrosis Endomiocárdica/fisiopatología , Femenino , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Ligadura , Lipocalina 2/fisiología , Ratones Endogámicos C57BL , Monocitos/fisiología , Daño por Reperfusión Miocárdica/fisiopatología , Neutropenia/fisiopatología , Fenotipo , Remodelación Ventricular/fisiología , Cicatrización de Heridas/fisiología , Tirosina Quinasa c-Mer/metabolismo
17.
Eur Heart J ; 36(39): 2635-42, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26049157

RESUMEN

Identification of subjects at increased risk for cardiovascular events plays a central role in the worldwide efforts to improve prevention, prediction, diagnosis, and prognosis of cardiovascular disease and to decrease the related costs. Despite their high predictive value on population level, traditional risk factors fail to fully predict individual risk. This position paper provides a summary of current vascular biomarkers other than the traditional risk factors with a special focus on the emerging -omics technologies. The definition of biomarkers and the identification and use of classical biomarkers are introduced, and we discuss the limitations of current biomarkers such as high sensitivity C-reactive protein (hsCRP) or N-terminal pro-brain natriuretic peptide (NT-proBNP). This is complemented by circulating plasma biomarkers, including high-density lipoprotein (HDL), and the conceptual shift from HDL cholesterol levels to HDL composition/function for cardiovascular risk assessment. Novel sources for plasma-derived markers include microparticles, microvesicles, and exosomes and their use for current omics-based analytics. Measurement of circulating micro-RNAs, short RNA sequences regulating gene expression, has attracted major interest in the search for novel biomarkers. Also, mass spectrometry and nuclear magnetic resonance spectroscopy have become key complementary technologies in the search for new biomarkers, such as proteomic searches or identification and quantification of small metabolites including lipids (metabolomics and lipidomics). In particular, pro-inflammatory lipid metabolites have gained much interest in the cardiovascular field. Our consensus statement concludes on leads and needs in biomarker research for the near future to improve individual cardiovascular risk prediction.


Asunto(s)
Aterosclerosis/diagnóstico , Biomarcadores/sangre , Micropartículas Derivadas de Células/metabolismo , Técnicas de Química Analítica/tendencias , Consenso , Enfermedad Coronaria/diagnóstico , Humanos , Lipoproteínas HDL/sangre , Metabolómica/tendencias , MicroARNs/sangre , Proteómica/tendencias , Medición de Riesgo/tendencias , Biología de Sistemas/tendencias
18.
Eur Heart J ; 35(43): 3013-20, 3020a-3020d, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25230814

RESUMEN

Blood vessels are exposed to multiple mechanical forces that are exerted on the vessel wall (radial, circumferential and longitudinal forces) or on the endothelial surface (shear stress). The stresses and strains experienced by arteries influence the initiation of atherosclerotic lesions, which develop at regions of arteries that are exposed to complex blood flow. In addition, plaque progression and eventually plaque rupture is influenced by a complex interaction between biological and mechanical factors-mechanical forces regulate the cellular and molecular composition of plaques and, conversely, the composition of plaques determines their ability to withstand mechanical load. A deeper understanding of these interactions is essential for designing new therapeutic strategies to prevent lesion development and promote plaque stabilization. Moreover, integrating clinical imaging techniques with finite element modelling techniques allows for detailed examination of local morphological and biomechanical characteristics of atherosclerotic lesions that may be of help in prediction of future events. In this ESC Position Paper on biomechanical factors in atherosclerosis, we summarize the current 'state of the art' on the interface between mechanical forces and atherosclerotic plaque biology and identify potential clinical applications and key questions for future research.


Asunto(s)
Arterias/fisiología , Aterosclerosis/fisiopatología , Apoptosis/fisiología , Biomarcadores/metabolismo , Fenómenos Biomecánicos/fisiología , Proliferación Celular/fisiología , Senescencia Celular/fisiología , Progresión de la Enfermedad , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Homeostasis/fisiología , Humanos , Mecanorreceptores/fisiología , Placa Aterosclerótica/fisiopatología , Rotura Espontánea/fisiopatología , Transducción de Señal/fisiología , Estrés Mecánico , Remodelación Vascular/fisiología
19.
Arterioscler Thromb Vasc Biol ; 33(2): 215-23, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23241405

RESUMEN

OBJECTIVE: Endocannabinoid levels are elevated in human and mouse atherosclerosis, but their causal role is not well understood. Therefore, we studied the involvement of fatty acid amide hydrolase (FAAH) deficiency, the major enzyme responsible for endocannabinoid anandamide degradation, in atherosclerotic plaque vulnerability. METHODS AND RESULTS: We assessed atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) and ApoE(-/-)FAAH(-/-) mice. Before and after 5, 10, and 15 weeks on high-cholesterol diet, we analyzed weight, serum cholesterol, and endocannabinoid levels, and atherosclerotic lesions in thoracoabdominal aortas and aortic sinuses. Serum levels of FAAH substrates anandamide, palmitoylethanolamide (PEA), and oleoylethanolamide (OEA) were 1.4- to 2-fold higher in case of FAAH deficiency. ApoE(-/-)FAAH(-/-) mice had smaller plaques with significantly lower content of smooth muscle cells, increased matrix metalloproteinase-9 expression, and neutrophil content. Circulating and bone marrow neutrophil counts were comparable between both genotypes, whereas CXC ligand1 levels were locally elevated in aortas of FAAH-deficient mice. We observed enhanced recruitment of neutrophils, but not monocytes, to large arteries of ApoE(-/-) mice treated with FAAH inhibitor URB597. Spleens of ApoE(-/-)FAAH(-/-) mice had reduced CD4+FoxP3+regulatory T-cell content, and in vitro stimulation of splenocytes revealed significantly elevated interferon-γ and tumor necrosis factor-α production in case of FAAH deficiency. CONCLUSIONS: Increased anandamide and related FAAH substrate levels are associated with the development of smaller atherosclerotic plaques with high neutrophil content, accompanied by an increased proinflammatory immune response.


Asunto(s)
Amidohidrolasas/deficiencia , Aorta/enzimología , Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Infiltración Neutrófila , Neutrófilos/inmunología , Amidas , Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/genética , Animales , Aorta/efectos de los fármacos , Aorta/inmunología , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Ácidos Araquidónicos/sangre , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Benzamidas/farmacología , Carbamatos/farmacología , Células Cultivadas , Quimiocina CXCL1/metabolismo , Colesterol/sangre , Modelos Animales de Enfermedad , Endocannabinoides/sangre , Inhibidores Enzimáticos/farmacología , Etanolaminas/sangre , Genotipo , Mediadores de Inflamación/metabolismo , Interferón gamma/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/patología , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Ácidos Oléicos/sangre , Ácidos Palmíticos/sangre , Fenotipo , Placa Aterosclerótica , Alcamidas Poliinsaturadas/sangre , Bazo/inmunología , Linfocitos T Reguladores/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Cardiovasc Res ; 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38838211

RESUMEN

AIMS: Although the cannabinoid CB1 receptor has been implicated in atherosclerosis, its cell-specific effects in this disease are not well understood. To address this, we generated a transgenic mouse model to study the role of myeloid CB1 signaling in atherosclerosis. METHODS AND RESULTS: Here, we report that male mice with myeloid-specific Cnr1 deficiency on atherogenic background developed smaller lesions and necrotic cores than controls, while only minor genotype differences were observed in females. Male Cnr1 deficient mice showed reduced arterial monocyte recruitment and macrophage proliferation with less inflammatory phenotype. The sex-specific differences in proliferation were dependent on estrogen receptor (ER)α-estradiol signaling. Kinase activity profiling identified a CB1-dependent regulation of p53 and cyclin-dependent kinases. Transcriptomic profiling further revealed chromatin modifications, mRNA processing and mitochondrial respiration among the key processes affected by CB1 signaling, which was supported by metabolic flux assays. Chronic administration of the peripherally-restricted CB1 antagonist JD5037 inhibited plaque progression and macrophage proliferation, but only in male mice. Finally, CNR1 expression was detectable in human carotid endarterectomy plaques and inversely correlated with proliferation, oxidative metabolism and inflammatory markers, suggesting a possible implication of CB1-dependent regulation in human pathophysiology. CONCLUSION: Impaired macrophage CB1 signaling is atheroprotective by limiting their arterial recruitment, proliferation and inflammatory reprogramming in male mice. The importance of macrophage CB1 signaling appears to be sex-dependent.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA