Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 51(5): 899-914.e7, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31732166

RESUMEN

Myocardial infarction, stroke, and sepsis trigger systemic inflammation and organism-wide complications that are difficult to manage. Here, we examined the contribution of macrophages residing in vital organs to the systemic response after these injuries. We generated a comprehensive catalog of changes in macrophage number, origin, and gene expression in the heart, brain, liver, kidney, and lung of mice with myocardial infarction, stroke, or sepsis. Predominantly fueled by heightened local proliferation, tissue macrophage numbers increased systemically. Macrophages in the same organ responded similarly to different injuries by altering expression of tissue-specific gene sets. Preceding myocardial infarction improved survival of subsequent pneumonia due to enhanced bacterial clearance, which was caused by IFNÉ£ priming of alveolar macrophages. Conversely, EGF receptor signaling in macrophages exacerbated inflammatory lung injury. Our data suggest that local injury activates macrophages in remote organs and that targeting macrophages could improve resilience against systemic complications following myocardial infarction, stroke, and sepsis.


Asunto(s)
Susceptibilidad a Enfermedades , Macrófagos/inmunología , Macrófagos/metabolismo , Animales , Biomarcadores , Recuento de Células , Susceptibilidad a Enfermedades/inmunología , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Isquemia/etiología , Isquemia/metabolismo , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Células Musculares/inmunología , Células Musculares/metabolismo , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Neumonía/etiología , Neumonía/metabolismo , Neumonía/patología
2.
Basic Res Cardiol ; 118(1): 36, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37656254

RESUMEN

Cardiotoxicity is a major complication of anthracycline therapy that negatively impacts prognosis. Effective pharmacotherapies for prevention of anthracycline-induced cardiomyopathy (AICM) are currently lacking. Increased plasma levels of the neutrophil-derived enzyme myeloperoxidase (MPO) predict occurrence of AICM in humans. We hypothesized that MPO release causally contributes to AICM. Mice intravenously injected with the anthracycline doxorubicin (DOX) exhibited higher neutrophil counts and MPO levels in the circulation and cardiac tissue compared to saline (NaCl)-treated controls. Neutrophil-like HL-60 cells exhibited increased MPO release upon exposition to DOX. DOX induced extensive nitrosative stress in cardiac tissue alongside with increased carbonylation of sarcomeric proteins in wildtype but not in Mpo-/- mice. Accordingly, co-treatment of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with DOX and MPO aggravated loss of hiPSC-CM-contractility compared to DOX treatment alone. DOX-treated animals exhibited pronounced cardiac apoptosis and inflammation, which was attenuated in MPO-deficient animals. Finally, genetic MPO deficiency and pharmacological MPO inhibition protected mice from the development of AICM. The anticancer efficacy of DOX was unaffected by MPO deficiency. Herein we identify MPO as a critical mediator of AICM. We demonstrate that DOX induces cardiac neutrophil infiltration and release of MPO, which directly impairs cardiac contractility through promoting oxidation of sarcomeric proteins, cardiac inflammation and cardiomyocyte apoptosis. MPO thus emerges as a promising pharmacological target for prevention of AICM.


Asunto(s)
Cardiomiopatías , Células Madre Pluripotentes Inducidas , Peroxidasa , Animales , Humanos , Ratones , Antraciclinas/toxicidad , Cardiomiopatías/inducido químicamente , Cardiomiopatías/prevención & control , Doxorrubicina/toxicidad , Inflamación , Peroxidasa/genética
3.
Basic Res Cardiol ; 117(1): 38, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35896846

RESUMEN

Chronic kidney disease's prevalence rises globally. Whereas dialysis treatment replaces the kidney's filtering function and prolongs life, dreaded consequences in remote organs develop inevitably over time. Even milder reductions in kidney function not requiring replacement therapy associate with bacterial infections, cardiovascular and heart valve disease, which markedly limit prognosis in these patients. The array of complications is diverse and engages a wide gamut of cellular and molecular mechanisms. The innate immune system is profoundly and systemically altered in chronic kidney disease and, as a unifying element, partakes in many of the disease's complications. As such, a derailed immune system fuels cardiovascular disease progression but also elevates the propensity for serious bacterial infections. Recent data further point towards a role in developing calcific aortic valve stenosis. Here, we delineate the current state of knowledge on how chronic kidney disease affects innate immunity in cardiovascular organs and on a systemic level. We review the role of circulating myeloid cells, monocytes and neutrophils, resident macrophages, dendritic cells, ligands, and cellular pathways that are activated or suppressed when renal function is chronically impaired. Finally, we discuss myeloid cells' varying responses to uremia from a systems immunology perspective.


Asunto(s)
Insuficiencia Renal Crónica , Uremia , Humanos , Inflamación , Leucocitos , Macrófagos , Insuficiencia Renal Crónica/complicaciones , Uremia/complicaciones
4.
Circulation ; 142(3): 244-258, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32316750

RESUMEN

BACKGROUND: Diabetes mellitus is a prevalent public health problem that affects about one-third of the US population and leads to serious vascular complications with increased risk for coronary artery disease. How bone marrow hematopoiesis contributes to diabetes mellitus complications is incompletely understood. We investigated the role of bone marrow endothelial cells in diabetic regulation of inflammatory myeloid cell production. METHODS: In 3 types of mouse diabetes mellitus, including streptozotocin, high-fat diet, and genetic induction using leptin-receptor-deficient db/db mice, we assayed leukocytes, hematopoietic stem and progenitor cells (HSPC). In addition, we investigated bone marrow endothelial cells with flow cytometry and expression profiling. RESULTS: In diabetes mellitus, we observed enhanced proliferation of HSPC leading to augmented circulating myeloid cell numbers. Analysis of bone marrow niche cells revealed that endothelial cells in diabetic mice expressed less Cxcl12, a retention factor promoting HSPC quiescence. Transcriptome-wide analysis of bone marrow endothelial cells demonstrated enrichment of genes involved in epithelial growth factor receptor (Egfr) signaling in mice with diet-induced diabetes mellitus. To explore whether endothelial Egfr plays a functional role in myelopoiesis, we generated mice with endothelial-specific deletion of Egfr (Cdh5CreEgfrfl/fl). We found enhanced HSPC proliferation and increased myeloid cell production in Cdh5CreEgfrfl/fl mice compared with wild-type mice with diabetes mellitus. Disrupted Egfr signaling in endothelial cells decreased their expression of the HSPC retention factor angiopoietin-1. We tested the functional relevance of these findings for wound healing and atherosclerosis, both implicated in complications of diabetes mellitus. Inflammatory myeloid cells accumulated more in skin wounds of diabetic Cdh5CreEgfrfl/fl mice, significantly delaying wound closure. Atherosclerosis was accelerated in Cdh5CreEgfrfl/fl mice, leading to larger and more inflamed atherosclerotic lesions in the aorta. CONCLUSIONS: In diabetes mellitus, bone marrow endothelial cells participate in the dysregulation of bone marrow hematopoiesis. Diabetes mellitus reduces endothelial production of Cxcl12, a quiescence-promoting niche factor that reduces stem cell proliferation. We describe a previously unknown counterregulatory pathway, in which protective endothelial Egfr signaling curbs HSPC proliferation and myeloid cell production.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Endoteliales/metabolismo , Mielopoyesis , Animales , Diabetes Mellitus Experimental , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Masculino , Ratones , Modelos Biológicos , Células Mieloides/metabolismo , Mielopoyesis/genética , Transducción de Señal , Transcriptoma
5.
Circ Res ; 123(4): 415-427, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-29980569

RESUMEN

RATIONALE: Inflammatory stress induced by exposure to bacterial lipopolysaccharide causes hematopoietic stem cell expansion in the bone marrow niche, generating a cellular immune response. As an integral component of the hematopoietic stem cell niche, the bone marrow vasculature regulates the production and release of blood leukocytes, which protect the host against infection but also fuel inflammatory diseases. OBJECTIVE: We aimed to develop imaging tools to explore vascular changes in the bone marrow niche during acute inflammation. METHODS AND RESULTS: Using the TLR (Toll-like receptor) ligand lipopolysaccharide as a prototypical danger signal, we applied multiparametric, multimodality and multiscale imaging to characterize how the bone marrow vasculature adapts when hematopoiesis boosts leukocyte supply. In response to lipopolysaccharide, ex vivo flow cytometry and histology showed vascular changes to the bone marrow niche. Specifically, proliferating endothelial cells gave rise to new vasculature in the bone marrow during hypoxic conditions. We studied these vascular changes with complementary intravital microscopy and positron emission tomography/magnetic resonance imaging. Fluorescence and positron emission tomography integrin αVß3 imaging signal increased during lipopolysaccharide-induced vascular remodeling. Vascular leakiness, quantified by albumin-based in vivo microscopy and magnetic resonance imaging, rose when neutrophils departed and hematopoietic stem and progenitor cells proliferated more vigorously. CONCLUSIONS: Introducing a tool set to image bone marrow either with cellular resolution or noninvasively within the entire skeleton, this work sheds light on angiogenic responses that accompany emergency hematopoiesis. Understanding and monitoring bone marrow vasculature may provide a key to unlock therapeutic targets regulating systemic inflammation.


Asunto(s)
Médula Ósea/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Tomografía de Emisión de Positrones/métodos , Nicho de Células Madre , Estrés Fisiológico , Animales , Médula Ósea/patología , Células Progenitoras Endoteliales/citología , Femenino , Inflamación/diagnóstico por imagen , Integrina alfaVbeta3/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Imagen Multimodal/métodos
6.
Arterioscler Thromb Vasc Biol ; 36(9): 1802-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27470513

RESUMEN

OBJECTIVE: Atherosclerosis is a chronic disease characterized by lipid accumulation in the arterial wall. After myocardial infarction (MI), atherosclerotic plaques are infiltrated by inflammatory myeloid cells that aggravate the disease and increase the risk of secondary myocardial ischemia. Splenic myelopoiesis provides a steady flow of myeloid cells to inflamed atherosclerotic lesions after MI. Therefore, targeting myeloid cell production in the spleen could ameliorate increased atherosclerotic plaque inflammation after MI. APPROACH AND RESULTS: Here we show that MI increases splenic myelopoiesis by driving hematopoietic stem and progenitor cells into the cell cycle. In an atherosclerotic mouse model, E-selectin inhibition decreased hematopoietic stem and progenitor cell proliferation in the spleen after MI. This led to reduced extramedullary myelopoiesis and decreased myeloid cell accumulation in atherosclerotic lesions. Finally, we observed stable atherosclerotic plaque features, including smaller plaque size, reduced necrotic core area, and thicker fibrous cap after E-selectin inhibition. CONCLUSIONS: Inhibiting E-selectin attenuated inflammation in atherosclerotic plaques, likely by reducing leukocyte recruitment into plaques and by mitigating hematopoietic stem and progenitor cell activation in the spleen of mice with MI.


Asunto(s)
Enfermedades de la Aorta/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Selectina E/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Hipercolesterolemia/metabolismo , Mielopoyesis/efectos de los fármacos , Infarto del Miocardio/tratamiento farmacológico , Bazo/efectos de los fármacos , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proliferación Celular/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrosis , Células Madre Hematopoyéticas/metabolismo , Hipercolesterolemia/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Necrosis , Placa Aterosclerótica , Transducción de Señal/efectos de los fármacos , Bazo/metabolismo
7.
J Mol Cell Cardiol ; 66: 126-32, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24286707

RESUMEN

The role of endocannabinoids such as anandamide during atherogenesis remains largely unknown. Fatty acid amide hydrolase (FAAH) represents the key enzyme in anandamide degradation, and its inhibition is associated with subsequent higher levels of anandamide. Here, we tested whether selective inhibition of FAAH influences the progression of atherosclerosis in mice. Selective inhibition of FAAH using URB597 resulted in significantly increased plasma levels of anandamide compared to control, as assessed by mass spectrometry experiments in mice. Apolipoprotein E-deficient (ApoE(-/-)) mice were fed a high-fat, cholesterol-rich diet to induce atherosclerotic conditions. Simultaneously, mice received either the pharmacological FAAH inhibitor URB597 1mg/kg body weight (n=28) or vehicle (n=25) via intraperitoneal injection three times a week. After eight weeks, mice were sacrificed, and experiments were performed. Vascular superoxide generation did not differ between both groups, as measured by L012 assay. To determine whether selective inhibition of FAAH affects atherosclerotic plaque inflammation, immunohistochemical staining of the aortic root was performed. Atherosclerotic plaque formation, vascular macrophage accumulation, as well as vascular T cell infiltration did not differ between both groups. Interestingly, neutrophil cell accumulation was significantly increased in mice receiving URB597 compared to control. Vascular collagen structures in atherosclerotic plaques were significantly diminished in mice treated with URB597 compared to control, as assessed by picro-sirius-red staining. This was accompanied by an increased aortic expression of matrix metalloproteinase-9, as determined by quantitative RT-PCR and western blot analysis. Inhibition of fatty acid amide hydrolase does not influence plaque size but increases plaque vulnerability in mice.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Benzamidas/farmacología , Carbamatos/farmacología , Inhibidores Enzimáticos/farmacología , Placa Aterosclerótica/enzimología , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Ácidos Araquidónicos/sangre , Movimiento Celular/efectos de los fármacos , Dieta Alta en Grasa , Grasas de la Dieta/efectos adversos , Endocannabinoides/sangre , Expresión Génica , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/patología , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/etiología , Placa Aterosclerótica/patología , Alcamidas Poliinsaturadas/sangre , Superóxidos/metabolismo
8.
Front Immunol ; 15: 1360700, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736886

RESUMEN

Introduction: Myocardial infarction (MI) is a significant contributor to morbidity and mortality worldwide. Many individuals who survive the acute event continue to experience heart failure (HF), with inflammatory and healing processes post-MI playing a pivotal role. Polymorphonuclear neutrophils (PMN) and monocytes infiltrate the infarcted area, where PMN release high amounts of the heme enzyme myeloperoxidase (MPO). MPO has numerous inflammatory properties and MPO plasma levels are correlated with prognosis and severity of MI. While studies have focused on MPO inhibition and controlling PMN infiltration into the infarcted tissue, less is known on MPO's role in monocyte function. Methods and results: Here, we combined human data with mouse and cell studies to examine the role of MPO on monocyte activation and migration. We revealed a correlation between plasma MPO levels and monocyte activation in a patient study. Using a mouse model of MI, we demonstrated that MPO deficiency led to an increase in splenic monocytes and a decrease in cardiac monocytes compared to wildtype mice (WT). In vitro studies further showed that MPO induces monocyte migration, with upregulation of the chemokine receptor CCR2 and upregulation of inflammatory pathways identified as underlying mechanisms. Conclusion: Taken together, we identify MPO as a pro-inflammatory mediator of splenic monocyte recruitment and activation post-MI and provide mechanistic insight for novel therapeutic strategies after ischemic injury.


Asunto(s)
Monocitos , Infarto del Miocardio , Peroxidasa , Animales , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Peroxidasa/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Humanos , Ratones , Masculino , Movimiento Celular , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Femenino , Neutrófilos/inmunología , Neutrófilos/metabolismo , Ratones Noqueados , Receptores CCR2/metabolismo , Persona de Mediana Edad
9.
Arterioscler Thromb Vasc Biol ; 32(8): 1925-35, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22701020

RESUMEN

OBJECTIVE: Endothelial microparticles (EMP) are released from activated or apoptotic cells, but their effect on target cells and the exact way of incorporation are largely unknown. We sought to determine the uptake mechanism and the biological effect of EMP on endothelial and endothelial-regenerating cells. METHODS AND RESULTS: EMP were generated from starved endothelial cells and isolated by ultracentrifugation. Caspase 3 activity assay and terminal deoxynucleotidyl transferase dUTP nick end labeling assay showed that EMP protect target endothelial cells against apoptosis in a dose-dependent manner. Proteomic analysis was performed to identify molecules contained in EMP, which might be involved in EMP uptake. Expression of annexin I in EMP was found and confirmed by Western blot, whereas the corresponding receptor phosphatidylserine receptor was present on endothelial target cells. Silencing either annexin I on EMP or phosphatidylserine receptor on target cells using small interfering RNA showed that the uptake of EMP by human coronary artery endothelial cells is annexin I/phosphatidylserine receptor dependent. Annexin I-downregulated EMP abrogated the EMP-mediated protection against apoptosis of endothelial target cells. p38 activation was found to mediate camptothecin-induced apoptosis. Finally, human coronary artery endothelial cells pretreated with EMP inhibited camptothecin-induced p38 activation. CONCLUSIONS: EMP are incorporated by endothelial cells in an annexin I/phosphatidylserine receptor-dependent manner and protect target cells against apoptosis. Inhibition of p38 activity is involved in EMP-mediated protection against apoptosis.


Asunto(s)
Anexina A1/fisiología , Apoptosis , Micropartículas Derivadas de Células/fisiología , Células Endoteliales/fisiología , Receptores de Superficie Celular/fisiología , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Células Cultivadas , Humanos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
11.
J Cell Mol Med ; 16(11): 2777-88, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22697268

RESUMEN

Microparticles (MP) are generated during a vast number of biological processes such as inflammation, cell activation and apoptosis. Increasing evidence points towards an important role of MP as intercellular messengers of biological information. During atherogenesis, monocytes infiltrate the vascular wall and foster inflammation, accompanied by the release of monocytic MP (mono-MP). To date, only little is known about the biological function of mono-MP in the vascular wall. Here, we investigated the role of mono-MP during atherogenesis. Mono-MP were generated by starvation of THP-1 monocytes and isolated by ultracentrifugation. To investigate whether mono-MP influence atherogenesis, ApoE(-/-) mice were fed a high-fat, cholesterol-rich diet for 8 weeks and simultaneously treated with mono-MP or vehicle twice a week. Mice treated with mono-MP showed significantly increased monocyte and T-cell infiltration into the vessel wall, as assessed by Moma-2 and CD3 staining, and enhanced plaque formation, as assessed by oil-red-O staining. However, atherosclerotic plaque composition was not influenced by mono-MP application. In vitro, incubation of mono-MP with murine macrophages and endothelial cells resulted in the uptake of calcein-labelled mono-MP. Mono-MP uptake initiated the generation of intracellular reactive oxygen species. Murine macrophages pre-treated with mono-MP showed significantly enhanced expression of CCR2, migration to MCP-1 and increased release of pro-inflammatory interleukin-6. Co-incubation of mono-MP with endothelial cells resulted in significantly increased expression of ICAM-1, as assessed by RT-PCR and ELISA. Mono-MP act as paracrine messengers that intensify inflammation during atherogenesis by stimulating vascular-bound and inflammatory cells in their vicinity.


Asunto(s)
Aterosclerosis/fisiopatología , Monocitos/fisiología , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Presión Sanguínea , Movimiento Celular , Células Cultivadas , Colesterol/sangre , Colágeno/metabolismo , Dieta Alta en Grasa/efectos adversos , Células Endoteliales/metabolismo , Frecuencia Cardíaca , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-6/metabolismo , Macrófagos/fisiología , Ratones , Ratones Mutantes , Especies Reactivas de Oxígeno/metabolismo , Receptores CCR2/metabolismo , Linfocitos T/fisiología , Vasculitis/metabolismo , Vasculitis/patología
12.
Mol Cell Biochem ; 359(1-2): 25-31, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21755457

RESUMEN

Atherosclerosis is a chronic inflammatory disease and represents the main cause of death in the industrialized world. Metabolites of the arachidonic acid derived from the 5-lipoxygenase pathway are known as leukotrienes that mediate various inflammatory processes during atherogenesis. Leukotriene B4 elicits the overexpression of several proinflammatory proteins, promotes chemotaxis and foam cell formation via BLT receptors. Currently, little is known about the implications of the BLT2 receptor in atherogenesis. Here, we tested whether selective inhibition of this receptor influences the progression of atherosclerosis in mice. Apolipoprotein-E deficient mice were fed a high-fat, cholesterol-rich diet to create atherosclerotic conditions (each group n=9). Simultaneously, mice received the pharmacologic BLT2 inhibition (Ly) by intraperitoneal injection every second day 5 mg/kg bw or vehicle. After 8 weeks, mice were killed and experiments were performed. Vascular superoxide release was diminished in mice treated with Ly compared with the control group (68±15 vs 131±20 RLU, P=0.01), as measured by L012 assay. Next, endothelial function was assessed by organ chamber experiments. Endothelial-dependent relaxation was improved in mice treated with the BLT2 receptor antagonist. To determine whether selective inhibition of the BLT2 receptor affects the atherosclerotic plaque growth, immunohistochemical stainings of the aortic root were performed. Oil red O staining revealed no significant differences between both groups (36±3% vs 38±3%). Monocyte infiltration into the vessel wall was analyzed using Moma-2 staining. No significant differences were observed between both groups (31±3% vs 34±2%). Selective inhibition of the BLT2 receptor in mice reduces the release of vascular reactive oxygen species and improves endothelial function in mice. Further experiments are necessary in order to obtain tissue-specific and mechanistical insights.


Asunto(s)
Endotelio Vascular/metabolismo , Antagonistas de Leucotrieno/farmacología , Estrés Oxidativo/efectos de los fármacos , Receptores de Leucotrieno B4/antagonistas & inhibidores , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Movimiento Celular , Endotelio Vascular/fisiología , Ratones , Ratones Noqueados , Monocitos , Especies Reactivas de Oxígeno/metabolismo
13.
J Mol Cell Cardiol ; 51(6): 1007-14, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21884703

RESUMEN

Low-dose oral tetrahydrocannabinol (THC) reduces progression of atherosclerosis in mice. THC activates central cannabinoid-1 receptors (CB1) with subsequent psychoactive effects as well as peripheral cannabinoid-2 receptors (CB2). In order to dissect the underlying mechanisms, we performed experiments under selective CB2 stimulation as well as after genetic disruption of the CB2 receptor. Atherosclerosis prone apolipoprotein E-deficient mice were crossed with cannabinoid receptor-2 deficient mice to obtain ApoE -/- CB2 -/- double knockout mice. After 8weeks of a high-cholesterol diet, immunohistochemical stainings of the aortic root revealed that vascular leukocyte infiltration in atherosclerotic plaques was accelerated in ApoE -/- CB2 -/- mice compared with ApoE -/- mice. This was accompanied by increased release of reactive oxygen species as measured using L012-enhanced chemiluminescence, and by decreased endothelial function as assessed in isolated aortic rings in organ chamber experiments. ApoE -/- mice treated with the selective CB2 agonist JWH 133 during a high-cholesterol diet showed decreased atherosclerotic lesion formation, improved endothelial function and reduced levels of reactive oxygen species. To assess whether CB2 expression in circulating cells influences atherosclerosis, irradiated ApoE -/- mice were repopulated with bone marrow-derived cells from ApoE -/- and ApoE -/- CB2 -/- mice and were fed a high-cholesterol diet for 8weeks. CB2 deficiency in bone marrow-derived cells increased leukocyte infiltration into the vessel wall, but had no impact on plaque formation. Cell culture experiments revealed that CB2 activation diminishes ROS generation in vascular cells. Selective CB2 receptor stimulation modulates atherogenesis via impact on both circulating proinflammatory and vascular cells.


Asunto(s)
Aterosclerosis/metabolismo , Receptor Cannabinoide CB2/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Presión Sanguínea , Trasplante de Médula Ósea , Moduladores de Receptores de Cannabinoides/metabolismo , Colesterol/sangre , Células Endoteliales/metabolismo , Endotelio/metabolismo , Frecuencia Cardíaca , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Receptor Cannabinoide CB2/genética
14.
Front Cardiovasc Med ; 8: 812702, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35097027

RESUMEN

The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.

15.
J Cell Mol Med ; 14(9): 2250-6, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20597998

RESUMEN

Endothelial cell apoptosis is a pivotal step in the development of atherosclerotic disease. Regeneration of the damaged endothelium is an attractive therapy option in the prevention and treatment of atherosclerotic disease. Apoptosis is associated with the release of microparticles (MP). Besides their role as marker of cell damage, recent reports have underlined their role as signalling elements in cell-cell communication. In this review, we focus on the emerging role of circulating MP as transmitters of biological information in cardiovascular disease.


Asunto(s)
Biomarcadores/metabolismo , Micropartículas Derivadas de Células/metabolismo , Animales , Comunicación Celular , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
16.
Nat Biomed Eng ; 4(11): 1076-1089, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33020600

RESUMEN

Bone-marrow endothelial cells in the haematopoietic stem-cell niche form a network of blood vessels that regulates blood-cell traffic as well as the maintenance and function of haematopoietic stem and progenitor cells. Here, we report the design and in vivo performance of systemically injected lipid-polymer nanoparticles encapsulating small interfering RNA (siRNA), for the silencing of genes in bone-marrow endothelial cells. In mice, nanoparticles encapsulating siRNA sequences targeting the proteins stromal-derived factor 1 (Sdf1) or monocyte chemotactic protein 1 (Mcp1) enhanced (when silencing Sdf1) or inhibited (when silencing Mcp1) the release of stem and progenitor cells and of leukocytes from the bone marrow. In a mouse model of myocardial infarction, nanoparticle-mediated inhibition of cell release from the haematopoietic niche via Mcp1 silencing reduced leukocytes in the diseased heart, improved healing after infarction and attenuated heart failure. Nanoparticle-mediated RNA interference in the haematopoietic niche could be used to investigate haematopoietic processes for therapeutic applications in cancer, infection and cardiovascular disease.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Silenciador del Gen/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , Nicho de Células Madre/genética , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Infarto del Miocardio/prevención & control
17.
Circ Cardiovasc Imaging ; 13(10): e010586, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33076700

RESUMEN

BACKGROUND: Macrophages, innate immune cells that reside in all organs, defend the host against infection and injury. In the heart and vasculature, inflammatory macrophages also enhance tissue damage and propel cardiovascular diseases. METHODS: We here use in vivo positron emission tomography (PET) imaging, flow cytometry, and confocal microscopy to evaluate quantitative noninvasive assessment of cardiac, arterial, and pulmonary macrophages using the nanotracer 64Cu-Macrin-a 20-nm spherical dextran nanoparticle assembled from nontoxic polyglucose. RESULTS: PET imaging using 64Cu-Macrin faithfully reported accumulation of macrophages in the heart and lung of mice with myocardial infarction, sepsis, or pneumonia. Flow cytometry and confocal microscopy detected the near-infrared fluorescent version of the nanoparticle (VT680Macrin) primarily in tissue macrophages. In 5-day-old mice, 64Cu-Macrin PET imaging quantified physiologically more numerous cardiac macrophages. Upon intravenous administration of 64Cu-Macrin in rabbits and pigs, we detected heightened macrophage numbers in the infarcted myocardium, inflamed lung regions, and atherosclerotic plaques using a clinical PET/magnetic resonance imaging scanner. Toxicity studies in rats and human dosimetry estimates suggest that 64Cu-Macrin is safe for use in humans. CONCLUSIONS: Taken together, these results indicate 64Cu-Macrin could serve as a facile PET nanotracer to survey spatiotemporal macrophage dynamics during various physiological and pathological conditions. 64Cu-Macrin PET imaging could stage inflammatory cardiovascular disease activity, assist disease management, and serve as an imaging biomarker for emerging macrophage-targeted therapeutics.


Asunto(s)
Radioisótopos de Cobre , Dextranos , Corazón/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Macrófagos/patología , Imagen Molecular , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radiofármacos , Animales , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/patología , Radioisótopos de Cobre/administración & dosificación , Radioisótopos de Cobre/farmacocinética , Dextranos/administración & dosificación , Dextranos/farmacocinética , Modelos Animales de Enfermedad , Inyecciones Intravenosas , Pulmón/patología , Macrófagos Alveolares/patología , Ratones , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Nanopartículas , Neumonía/diagnóstico por imagen , Neumonía/patología , Valor Predictivo de las Pruebas , Conejos , Radiofármacos/administración & dosificación , Radiofármacos/farmacocinética , Porcinos , Porcinos Enanos , Factores de Tiempo
18.
PLoS One ; 11(1): e0146267, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26731274

RESUMEN

BACKGROUND: The endocannabinoid 2-arachidonoylglycerol (2-AG) is a known modulator of inflammation. Despite its high concentration in vascular tissue, the role of 2-AG in atherogenesis has not yet been examined. METHODS: ApoE-deficient mice were sublethally irradiated and reconstituted with bone marrow from mice with a myeloid-specific knockout of the 2-AG synthesising enzyme diacylglycerol lipase α (Dagla) or control bone marrow with an intact 2-AG biosynthesis. After a cholesterol-rich diet for 8 weeks, plaque size and plaque morphology were examined in chimeric mice. Circulating inflammatory cells were assessed by flow cytometry. Aortic tissue and plasma levels of endocannabinoids were measured using liquid chromatography-multiple reaction monitoring. RESULTS: Mice with Dagla-deficient bone marrow and circulating myeloid cells showed a significantly reduced plaque burden compared to controls. The reduction in plaque size was accompanied by a significantly diminished accumulation of both neutrophil granulocytes and macrophages in atherosclerotic lesions of Dagla-deficient mice. Moreover, CB2 expression and the amount of oxidised LDL within atherosclerotic lesions was significantly reduced. FACS analyses revealed that levels of circulating inflammatory cells were unaltered in Dagla-deficient mice. CONCLUSIONS: Myeloid synthesis of the endocannabinoid 2-AG appears to promote vascular inflammation and atherogenesis. Thus, myeloid-specific disruption of 2-AG synthesis may represent a potential novel therapeutic strategy against atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Lipoproteína Lipasa/genética , Células Mieloides/metabolismo , Animales , Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Presión Sanguínea/genética , Frecuencia Cardíaca/genética , Lipoproteína Lipasa/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Células Mieloides/patología , Placa Aterosclerótica/patología , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Superóxidos/metabolismo
19.
J Exp Med ; 212(4): 497-512, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25800955

RESUMEN

Splenic myelopoiesis provides a steady flow of leukocytes to inflamed tissues, and leukocytosis correlates with cardiovascular mortality. Yet regulation of hematopoietic stem cell (HSC) activity in the spleen is incompletely understood. Here, we show that red pulp vascular cell adhesion molecule 1 (VCAM-1)(+) macrophages are essential to extramedullary myelopoiesis because these macrophages use the adhesion molecule VCAM-1 to retain HSCs in the spleen. Nanoparticle-enabled in vivo RNAi silencing of the receptor for macrophage colony stimulation factor (M-CSFR) blocked splenic macrophage maturation, reduced splenic VCAM-1 expression and compromised splenic HSC retention. Both, depleting macrophages in CD169 iDTR mice or silencing VCAM-1 in macrophages released HSCs from the spleen. When we silenced either VCAM-1 or M-CSFR in mice with myocardial infarction or in ApoE(-/-) mice with atherosclerosis, nanoparticle-enabled in vivo RNAi mitigated blood leukocytosis, limited inflammation in the ischemic heart, and reduced myeloid cell numbers in atherosclerotic plaques.


Asunto(s)
Hematopoyesis Extramedular/inmunología , Células Madre Hematopoyéticas/inmunología , Macrófagos/inmunología , Mielopoyesis/inmunología , Bazo/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/inmunología , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Hematopoyesis Extramedular/genética , Células Madre Hematopoyéticas/patología , Macrófagos/patología , Ratones , Ratones Noqueados , Mielopoyesis/genética , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Nanopartículas , Placa Aterosclerótica/genética , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Interferencia de ARN , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/inmunología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Lectina 1 Similar a Ig de Unión al Ácido Siálico/inmunología , Bazo/patología , Molécula 1 de Adhesión Celular Vascular/genética
20.
Cardiol J ; 21(4): 392-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23990193

RESUMEN

BACKGROUND: Obstructive sleep apnea (OSA) has been identified as associated with the onset and propagation of atrial fibrillation (AF) and predicts recurrences of AF after pulmonary vein isolation (PVI). Vice versa, it has never been investigated whether PVI influences OSA. However, it has been controversial whether a restored atrial function can affect the course of OSA. There-fore, we have assessed whether PVI procedure modulates the prevalence and severity of OSA. METHODS AND RESULTS: We included 23 individuals with AF that were assigned to undergo PVI into this study. Patients were 65 ± 7 years old, obese (BMI 29.9 ± 5.4 kg/m²), white (100%) and had a normal left ventricular function (LVEF 64 ± 9%). Polygraphic assessment was carried out before and 6 months after PVI. The prevalence of OSA, defined as an apnea-hypopnea index (AHI) ≥ 5 per hour of sleep, was 74% before PVI compared to 70% 6 months after the procedure (p > 0.05). Severity of OSA did not differ (AHI before vs. after: 18 ± 18/h vs. 15 ± 17/h, p > 0.05) as well as further polygraphic parameters did not differ before and after the procedure. CONCLUSIONS: Prevalence and severity of OSA are not affected by PVI in patients suffering from AF.


Asunto(s)
Fibrilación Atrial/cirugía , Ablación por Catéter , Venas Pulmonares/cirugía , Síndromes de la Apnea del Sueño/epidemiología , Anciano , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/epidemiología , Fibrilación Atrial/fisiopatología , Ablación por Catéter/efectos adversos , Femenino , Alemania/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Prevalencia , Venas Pulmonares/fisiopatología , Índice de Severidad de la Enfermedad , Síndromes de la Apnea del Sueño/diagnóstico , Síndromes de la Apnea del Sueño/fisiopatología , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA