Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Rep ; 11(1): 4126, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33602971

RESUMEN

Individuals with chronic kidney disease are at an increased risk for cardiovascular disease. This risk may partially be explained by a chronic inflammatory state in these patients, reflected by increased arterial wall and cellular inflammation. Statin treatment decreases cardiovascular risk and arterial inflammation in non-CKD subjects. In patients with declining kidney function, cardiovascular benefit resulting from statin therapy is attenuated, possibly due to persisting inflammation. In the current study, we assessed the effect of statin treatment on arterial wall and cellular inflammation. Fourteen patients with chronic kidney disease stage 3 or 4, defined by an estimated Glomerular Filtration Rate between 15 and 60 mL/min/1.73 m2, without cardiovascular disease were included in a single center, open label study to assess the effect of atorvastatin 40 mg once daily for 12 weeks (NTR6896). At baseline and at 12 weeks of treatment, we assessed arterial wall inflammation by 18F-fluoro-deoxyglucose positron-emission tomography computed tomography (18F-FDG PET/CT) and the phenotype of circulating monocytes were assessed. Treatment with atorvastatin resulted in a 46% reduction in LDL-cholesterol, but this was not accompanied by an attenuation in arterial wall inflammation in the aorta or carotid arteries, nor with changes in chemokine receptor expression of circulating monocytes. Statin treatment does not abolish arterial wall or cellular inflammation in subjects with mild to moderate chronic kidney disease. These results imply that CKD-associated inflammatory activity is mediated by factors beyond LDL-cholesterol and specific anti-inflammatory interventions might be necessary to further dampen the inflammatory driven CV risk in these subjects.


Asunto(s)
Antiinflamatorios/uso terapéutico , Atorvastatina/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Insuficiencia Renal Crónica/dietoterapia , Anciano , Aorta/efectos de los fármacos , Aorta/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Enfermedades Cardiovasculares/metabolismo , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , LDL-Colesterol/metabolismo , Femenino , Fluorodesoxiglucosa F18/administración & dosificación , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inflamación/metabolismo , Masculino , Persona de Mediana Edad , Placa Aterosclerótica/tratamiento farmacológico , Placa Aterosclerótica/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Radiofármacos/administración & dosificación , Insuficiencia Renal Crónica/metabolismo , Factores de Riesgo
3.
Eur J Nucl Med Mol Imaging ; 45(11): 1956-1963, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29728748

RESUMEN

PURPOSE: An acute coronary syndrome (ACS) is characterized by a multi-level inflammatory response, comprising activation of bone marrow and spleen accompanied by augmented release of leukocytes into the circulation. The duration of this response after an ACS remains unclear. Here, we assessed the effect of an ACS on the multi-level inflammatory response in patients both acutely and after 3 months. METHODS: We performed 18F-DPA-714 PET/CT acutely and 3 months post-ACS in eight patients and eight matched healthy controls. DPA-714, a PET tracer binding the TSPO receptor and highly expressed in myeloid cells, was used to assess hematopoietic activity. We also characterized circulating monocytes and hematopoietic stem and progenitor cells (HSPCs) by flow cytometry in 20 patients acutely and 3 months post-ACS and in 19 healthy controls. RESULTS: In the acute phase, patients displayed a 1.4-fold and 1.3-fold higher 18F-DPA-714 uptake in, respectively, bone marrow (p = 0.012) and spleen (p = 0.039) compared with healthy controls. This coincided with a 2.4-fold higher number of circulating HSPCs (p = 0.001). Three months post-ACS, 18F-DPA-714 uptake in bone marrow decreased significantly (p = 0.002), but no decrease was observed for 18F-DPA-714 uptake in the spleen (p = 0.67) nor for the number of circulating HSPCs (p = 0.75). CONCLUSIONS: 18F-DPA-714 PET/CT reveals an ACS- triggered hematopoietic organ activation as initiator of a prolonged cellular inflammatory response beyond 3 months, characterized by a higher number of circulating leukocytes and their precursors. This multi-level inflammatory response may provide an attractive target for novel treatment options aimed at reducing the high recurrence rate post-ACS.


Asunto(s)
Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/diagnóstico por imagen , Células Madre Hematopoyéticas/citología , Monocitos/citología , Tomografía Computarizada por Tomografía de Emisión de Positrones , Pirazoles , Pirimidinas , Síndrome Coronario Agudo/metabolismo , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Receptores CCR2/metabolismo , Bazo/inmunología
4.
Arterioscler Thromb Vasc Biol ; 37(5): 969-975, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28336558

RESUMEN

OBJECTIVE: Mendelian randomization studies revealed a causal role for remnant cholesterol in cardiovascular disease. Remnant particles accumulate in the arterial wall, potentially propagating local and systemic inflammation. We evaluated the impact of remnant cholesterol on arterial wall inflammation, circulating monocytes, and bone marrow in patients with familial dysbetalipoproteinemia (FD). APPROACH AND RESULTS: Arterial wall inflammation and bone marrow activity were measured using 18F-FDG PET/CT. Monocyte phenotype was assessed with flow cytometry. The correlation between remnant levels and hematopoietic activity was validated in the CGPS (Copenhagen General Population Study). We found a 1.2-fold increase of 18F-FDG uptake in the arterial wall in patients with FD (n=17, age 60±8 years, remnant cholesterol: 3.26 [2.07-5.71]) compared with controls (n=17, age 61±8 years, remnant cholesterol 0.29 [0.27-0.40]; P<0.001). Monocytes from patients with FD showed increased lipid accumulation (lipid-positive monocytes: Patients with FD 92% [86-95], controls 76% [66-81], P=0.001, with an increase in lipid droplets per monocyte), and a higher expression of surface integrins (CD11b, CD11c, and CD18). Patients with FD also exhibited monocytosis and leukocytosis, accompanied by a 1.2-fold increase of 18F-FDG uptake in bone marrow. In addition, we found a strong correlation between remnant levels and leukocyte counts in the CGPS (n=103 953, P for trend 5×10-276). In vitro experiments substantiated that remnant cholesterol accumulates in human hematopoietic stem and progenitor cells coinciding with myeloid skewing. CONCLUSIONS: Patients with FD have increased arterial wall and cellular inflammation. These findings imply an important inflammatory component to the atherogenicity of remnant cholesterol, contributing to the increased cardiovascular disease risk in patients with FD.


Asunto(s)
Arterias/inmunología , Arteritis/inmunología , Colesterol/inmunología , Hiperlipoproteinemia Tipo III/inmunología , Inmunidad Celular , Lipoproteínas/inmunología , Triglicéridos/inmunología , Anciano , Arterias/diagnóstico por imagen , Arterias/metabolismo , Arteritis/sangre , Arteritis/diagnóstico por imagen , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Colesterol/sangre , Dinamarca , Femenino , Fluorodesoxiglucosa F18 , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hiperlipoproteinemia Tipo III/sangre , Hiperlipoproteinemia Tipo III/diagnóstico por imagen , Integrinas/inmunología , Integrinas/metabolismo , Lipoproteínas/sangre , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Fenotipo , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radiofármacos , Transducción de Señal , Triglicéridos/sangre
5.
Eur Heart J ; 38(6): 425-432, 2017 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-27357356

RESUMEN

Aims: Experimental work posits that acute ischaemic events trigger haematopoietic activity, driving monocytosis, and atherogenesis. Considering the chronic low-grade inflammatory state in atherosclerosis, we hypothesized that haematopoietic hyperactivity is a persistent feature in cardiovascular disease (CVD). Therefore, we aimed to assess the activity of haematopoietic organs and haematopoietic stem and progenitor cells (HSPCs) in humans. Methods and results: First, we performed 18F-fluorodeoxyglucose positron emission tomographic (18F-FDG PET) imaging in 26 patients with stable atherosclerotic CVD (ischaemic event >12 months ago), and 25 matched controls. In splenic tissue, 18F-FDG uptake was 2.68 ± 0.65 in CVD patients vs. 1.75 ± 0.54 in controls (1.6-fold higher; P< 0.001), and in bone marrow 3.20 ± 0.76 vs. 2.72 ± 0.46 (1.2-fold higher; P = 0.003), closely related to LDL cholesterol levels (LDLc, r = 0.72). Subsequently, we determined progenitor potential of HSPCs harvested from 18 patients with known atherosclerotic CVD and 30 matched controls; both groups were selected from a cohort of cancer patients undergoing autologous stem cell transplantation. In CVD patients, the normalized progenitor potential, expressed as the number of colony-forming units-granulocyte/monocyte (CFU-GM) colonies/CD34+ cell, was 1.6-fold higher compared with matched controls (P < 0.001). Finally, we assessed the effects of native and oxidized lipoproteins on HSPCs harvested from healthy donors in vitro. Haematopoietic stem and progenitor cells displayed a 1.5-fold increased CFU-GM capacity in co-culture with oxidized LDL in vitro (P = 0.002), which was inhibited by blocking oxidized phospholipids via E06 (P = 0.001). Conclusion: Collectively, these findings strengthen the case for a chronically affected haematopoietic system, potentially driving the low-grade inflammatory state in patients with atherosclerosis.


Asunto(s)
Enfermedad de la Arteria Coronaria/patología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Calcificación Vascular/patología , Médula Ósea/fisiología , Estudios de Casos y Controles , Células Cultivadas , LDL-Colesterol/fisiología , Femenino , Fluorodesoxiglucosa F18 , Hematopoyesis Extramedular/fisiología , Humanos , Masculino , Persona de Mediana Edad , Tomografía Computarizada por Tomografía de Emisión de Positrones , Radiofármacos , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA