Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Haematol ; 196(4): 1040-1051, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34786709

RESUMEN

Sickle cell disease (SCD) is associated with haemolytic anaemia and secondary activation of leucocytes and platelets, which in turn may further exacerbate haemolysis. As cytokine signalling pathways may participate in this cycle, the present study investigated whether pharmacological blockade of the interleukin-1 receptor (IL-1R) would mitigate anaemia in a murine model of SCD. Within 2 weeks of treatment, reduced markers of haemolysis were observed in anakinra-treated mice compared to vehicle-treated mice. After 4 weeks of anakinra treatment, mice showed increased numbers of erythrocytes, haemoglobin, and haematocrit, along with reduced reticulocytes. Blood from anakinra-treated mice was less susceptible to ex vivo erythrocyte sickling and was resistant to exogenous IL-1ß-mediated sickling. Supernatant generated from IL-1ß-treated platelets was sufficient to promote erythrocyte sickling, an effect not observed with platelet supernatant generated from IL-1R-/- mice. The sickling effect of IL-1ß-treated platelet supernatant was inhibited by a transforming growth factor-ß (TGF-ß) neutralising antibody, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibition, and superoxide scavengers, but replicated by recombinant TGF-ß. In conclusion, pharmacological IL-1R antagonism leads to improved anaemia in a murine SCD model. IL-1ß stimulation of platelets promotes erythrocyte sickling. This effect may be mediated by platelet-derived TGF-ß-induced reactive oxygen species generation though erythrocyte NADPH oxidase.


Asunto(s)
Anemia de Células Falciformes/genética , Anemia/fisiopatología , Plaquetas/metabolismo , Receptores de Interleucina-1/genética , Animales , Modelos Animales de Enfermedad , Eritrocitos Anormales , Humanos , Ratones
2.
Arterioscler Thromb Vasc Biol ; 40(6): 1533-1542, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32268786

RESUMEN

OBJECTIVE: Clopidogrel is a commonly used P2Y12 inhibitor to treat and prevent arterial thrombotic events. Clopidogrel is a prodrug that requires bioactivation by CYP (cytochrome P450) enzymes to exert antiplatelet activity. Diabetes mellitus is associated with an increased risk of ischemic events, and impaired ability to generate the active metabolite (AM) from clopidogrel. The objective of this study is to identify the mechanism of clopidogrel resistance in a murine model of diet-induced obesity (DIO). Approach and Results: C57BL/6J mice and IL-1R-/- mice were given high-fat diet for 10 weeks to generate a murine model of diet-induced obesity. Platelet aggregation and carotid arterial thrombosis were assessed in response to clopidogrel treatment. Wild-type DIO mice exhibited resistance to antiplatelet and antithrombotic effects of clopidogrel that was associated with reduced hepatic expression of CYP genes and reduced generation of the AM. IL (Interleukin)-1 receptor-deficient DIO (IL1R-/- DIO) mice showed no resistance to clopidogrel. Lack of resistance was accompanied by increased exposure of the clopidogrel AM. This resistance was also absent when wild-type DIO mice were treated with the conjugate of the clopidogrel AM, DT-678. CONCLUSIONS: These findings indicate that antiplatelet effects of clopidogrel may be impaired in the setting of diabetes mellitus due to reduced prodrug bioactivation related to IL-1 receptor signaling. Therapeutic targeting of P2Y12 in patients with diabetes mellitus using the conjugate of clopidogrel AM may lead to improved outcomes.


Asunto(s)
Clopidogrel/farmacocinética , Clopidogrel/uso terapéutico , Resistencia a Medicamentos , Obesidad/complicaciones , Receptores de Interleucina-1/fisiología , Animales , Trombosis de las Arterias Carótidas/prevención & control , Clopidogrel/farmacología , Sistema Enzimático del Citocromo P-450/genética , Diabetes Mellitus , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrinolíticos , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/enzimología , Obesidad/etiología , Obesidad/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria , Profármacos/farmacocinética , Profármacos/uso terapéutico , Receptores de Interleucina-1/deficiencia
3.
Br J Haematol ; 174(3): 461-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27030515

RESUMEN

Mechanistic target of rapamycin (mTOR) has been shown to play an important role in red blood cell physiology, with inhibition of mTOR signalling leading to alterations in erythropoiesis. To determine if mTOR inhibition would improve anaemia in sickle cell disease (SCD), mice with SCD were treated with the dual mTORC1/2 inhibitor, INK128. One week after daily oral drug treatment, erythrocyte count, haemoglobin, and haematocrit were all significantly increased while reticulocyte counts were reduced. These parameters remained stable during 3 weeks of treatment. Similar effects were observed following oral treatment with the mTORC1 inhibitor, sirolimus. Sirolimus treatment prolonged the lifespan of sickle cell erythrocytes in circulation, reduced spleen size, and reduced renal and hepatic iron accumulation in SCD mice. Following middle cerebral artery occlusion, stroke size was reduced in SCD mice treated with sirolimus. In conclusion, mTOR inhibition is protective against anaemia and organ damage in a murine model of SCD.


Asunto(s)
Anemia de Células Falciformes/tratamiento farmacológico , Benzoxazoles/farmacología , Pirimidinas/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Anemia/prevención & control , Anemia de Células Falciformes/complicaciones , Animales , Benzoxazoles/administración & dosificación , Modelos Animales de Enfermedad , Eritrocitos/efectos de los fármacos , Hematócrito , Hemoglobinas , Ratones , Pirimidinas/administración & dosificación , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/prevención & control
4.
J Pharmacol Exp Ther ; 359(1): 11-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27511819

RESUMEN

Clopidogrel is a prodrug that requires bioactivation by cytochrome P450 (P450) enzymes to a pharmacologically active metabolite for antiplatelet action. The clinical limitations of clopidogrel are in large part due to its poor pharmacokinetics resulting from inefficient bioactivation by P450s. In this study, we determined the pharmacokinetics and pharmacodynamics of a novel conjugate of clopidogrel, referred to as ClopNPT, in animal models and we evaluated its potential to overcome the limitations of clopidogrel. Results from pharmacokinetic (PK) studies showed that ClopNPT released the active metabolite with a time to maximal plasma concentration of <5 minutes in C57BL/6 mice after either oral or intravenous administration, and plasma concentrations of the active metabolite reached Cmax values of 1242 and 1100 ng/ml after a 10-mg/kg oral dose and a 5-mg/kg intravenous dose, respectively. Furthermore, ClopNPT was highly effective in preventing arterial thrombosis in rabbits and mice after vascular injuries. Formation of occlusive thrombi was prevented by ClopNPT at the 1-mg/kg dose with no significant increase in tongue bleeding time, whereas clopidogrel was ineffective at the same dose. These results suggest that ClopNPT has favorable PK/pharmacodynamic properties that can potentially overcome the attenuated PK properties of clopidogrel and thus significantly improve the efficacy of antiplatelet therapy.


Asunto(s)
Arterias/efectos de los fármacos , Fibrinolíticos/química , Fibrinolíticos/farmacología , Trombosis/tratamiento farmacológico , Ticlopidina/análogos & derivados , Animales , Arterias/fisiopatología , Clopidogrel , Modelos Animales de Enfermedad , Fibrinolíticos/farmacocinética , Fibrinolíticos/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Agregación Plaquetaria/efectos de los fármacos , Piridinas/química , Conejos , Trombosis/fisiopatología , Ticlopidina/química , Ticlopidina/farmacocinética , Ticlopidina/farmacología , Ticlopidina/uso terapéutico
5.
Circ Res ; 114(6): 947-56, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24425713

RESUMEN

RATIONALE: Neutrophil extracellular trap (NET) formation promotes vascular damage, thrombosis, and activation of interferon-α-producing plasmacytoid dendritic cells in diseased arteries. Peptidylarginine deiminase inhibition is a strategy that can decrease in vivo NET formation. OBJECTIVE: To test whether peptidylarginine deiminase inhibition, a novel approach to targeting arterial disease, can reduce vascular damage and inhibit innate immune responses in murine models of atherosclerosis. METHODS AND RESULTS: Apolipoprotein-E (Apoe)(-/-) mice demonstrated enhanced NET formation, developed autoantibodies to NETs, and expressed high levels of interferon-α in diseased arteries. Apoe(-/-) mice were treated for 11 weeks with daily injections of Cl-amidine, a peptidylarginine deiminase inhibitor. Peptidylarginine deiminase inhibition blocked NET formation, reduced atherosclerotic lesion area, and delayed time to carotid artery thrombosis in a photochemical injury model. Decreases in atherosclerosis burden were accompanied by reduced recruitment of netting neutrophils and macrophages to arteries, as well as by reduced arterial interferon-α expression. CONCLUSIONS: Pharmacological interventions that block NET formation can reduce atherosclerosis burden and arterial thrombosis in murine systems. These results support a role for aberrant NET formation in the pathogenesis of atherosclerosis through modulation of innate immune responses.


Asunto(s)
Aterosclerosis/prevención & control , Inhibidores Enzimáticos/uso terapéutico , Hidrolasas/antagonistas & inhibidores , Inmunidad Innata/efectos de los fármacos , Ornitina/análogos & derivados , Animales , Enfermedades de la Aorta/tratamiento farmacológico , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Apolipoproteínas E/deficiencia , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/enzimología , Aterosclerosis/etiología , Aterosclerosis/inmunología , Aterosclerosis/patología , Autoanticuerpos/biosíntesis , Autoanticuerpos/inmunología , Citrulina/análisis , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Espacio Extracelular , Histonas/metabolismo , Hidrolasas/fisiología , Interferón-alfa/biosíntesis , Interferón-alfa/genética , Selectina L/análisis , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutropenia/inmunología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/ultraestructura , Ornitina/farmacología , Ornitina/uso terapéutico , Procesos Fotoquímicos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Arginina Deiminasa Proteína-Tipo 4 , Receptor de Interferón alfa y beta/deficiencia , Seno Aórtico/patología , Túnica Íntima/patología
6.
Arterioscler Thromb Vasc Biol ; 34(10): 2217-23, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25147343

RESUMEN

Obesity is a worldwide epidemic and is associated with multiple comorbidities. The mechanisms underlying the relationship between obesity and adverse health outcomes remain poorly understood. This may be because of several factors including the crude measures used to estimate adiposity, the striking heterogeneity between adipose tissue depots, and the influence of fat accumulation in multiple organs. To advance our understanding of fat stores and associated comorbidities in humans, it will be necessary to image adiposity throughout the body and ultimately also assess its functionality. Large clinical studies are demonstrating the prognostic importance of adipose tissue imaging. Newer techniques capable of imaging fat metabolism and other functions of adipose tissue may provide additional prognostic use and may be useful in guiding therapeutic interventions.


Asunto(s)
Tejido Adiposo , Adiposidad , Enfermedades Cardiovasculares/epidemiología , Diagnóstico por Imagen , Obesidad/diagnóstico , Absorciometría de Fotón , Tejido Adiposo/diagnóstico por imagen , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Animales , Enfermedades Cardiovasculares/diagnóstico , Comorbilidad , Diagnóstico por Imagen/métodos , Humanos , Metabolismo de los Lípidos , Imagen por Resonancia Magnética , Obesidad/epidemiología , Obesidad/metabolismo , Obesidad/fisiopatología , Tomografía de Emisión de Positrones , Valor Predictivo de las Pruebas , Factores de Riesgo , Tomografía Computarizada por Rayos X , Ultrasonografía , Imagen de Cuerpo Entero
7.
Arterioscler Thromb Vasc Biol ; 34(8): 1621-30, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24833795

RESUMEN

Perivascular adipose tissue (PVAT), long assumed to be nothing more than vessel-supporting connective tissue, is now understood to be an important, active component of the vasculature, with integral roles in vascular health and disease. PVAT is an adipose tissue with similarities to both brown and white adipose tissue, although recent evidence suggests that PVAT develops from its own precursors. Like other adipose tissue depots, PVAT secretes numerous biologically active substances that can act in both autocrine and paracrine fashion. PVAT has also proven to be involved in vascular inflammation. Although PVAT can support inflammation during atherosclerosis via macrophage accumulation, emerging evidence suggests that PVAT also has antiatherosclerotic properties related to its abilities to induce nonshivering thermogenesis and metabolize fatty acids. We here discuss the accumulated knowledge of PVAT biology and related research on models of hypertension and atherosclerosis.


Asunto(s)
Tejido Adiposo/metabolismo , Aterosclerosis/metabolismo , Vasos Sanguíneos/metabolismo , Hipertensión/metabolismo , Tejido Adiposo/fisiopatología , Adiposidad , Animales , Aterosclerosis/fisiopatología , Comunicación Autocrina , Vasos Sanguíneos/fisiopatología , Regulación de la Temperatura Corporal , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Hemodinámica , Humanos , Hipertensión/fisiopatología , Inflamación/metabolismo , Inflamación/fisiopatología , Comunicación Paracrina , Transducción de Señal
9.
Br J Haematol ; 162(1): 120-9, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23590132

RESUMEN

Sickle cell disease (SCD) is associated with vascular complications including premature stroke. The role of atherothrombosis in these vascular complications is unclear. To determine the effect of SCD on atherosclerosis and thrombosis, mice with SCD along with controls were generated by transplantation of bone marrow from mice carrying the homozygous sickle cell mutation (Hbb(hßs/hßs) ) or wild-type mice (Hbb(+/+) ) into C57BL6/J or apolipoprotein E deficient (Apoe(-/-) ) recipient mice. At the time of sacrifice, 23-28 weeks following bone marrow transplantation, anaemia, reticulocytosis, and splenomegaly were present in mice receiving Hbb(hßs/hßs) bone marrow compared with control mice. Analysis of atherosclerosis involving the aortic root revealed reduced atherosclerotic lesion area with reduced macrophage content and increased collagen content in Apoe(-/-) , Hbb(hßs/hßs) mice compared to Apoe(-/-) , Hbb(+/+) mice. In a carotid thrombosis model, the time to thrombosis was prolonged in Hbb(hßs/hßs) mice compared to Hbb(+/+) mice. This apparent protective effect of SCD on atherosclerosis and thrombosis was diminished by inhibition of heme oxygenase-1 (HMOX1) using zinc protoporphyrin IX. We conclude that SCD in mice is paradoxically protective against atherosclerosis and thrombosis, highlighting the complexity of vascular events in SCD. This protective effect is at least partially mediated by induction of HMOX1.


Asunto(s)
Anemia de Células Falciformes/genética , Anemia de Células Falciformes/metabolismo , Aterosclerosis/prevención & control , Trombosis/prevención & control , Anemia de Células Falciformes/complicaciones , Animales , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/etiología , Arterias Carótidas/patología , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/metabolismo , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Protoporfirinas/farmacología , Trombosis/etiología
10.
Circ J ; 77(1): 11-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23207957

RESUMEN

Perivascular adipose tissue (PVAT) is the fat tissue surrounding most of the vasculature and it has long been considered solely as vessel-supporting connective tissue. There are 2 major types of adipose tissue widely distributed throughout the body: white (WAT) and brown (BAT). PVAT is similar to BAT in rodents, but it was believed that only WAT existed in adult humans and BAT was present only in infants. However, the presence of functional BAT in adult humans is now accepted. The main function of BAT is to generate heat, and it is essential for adaptive thermogenesis and energy expenditure, whereas the main function of WAT is to store lipids. Besides the different functions of WAT and BAT, growing evidence suggests that different depots of adipose tissue have different functions. Similar to other fat depots, PVAT produces various adipokines, growth factors and inhibitors that affect functions of adjacent layers of the vasculature. Pathophysiological conditions such as obesity, vascular injury, aging and infection could cause PVAT dysfunction, leading to vascular endothelial and smooth muscle cell dysfunctions. In this review, we discuss the function and dysfunction of PVAT on atherosclerosis and hypertension.


Asunto(s)
Tejido Adiposo/metabolismo , Tejido Adiposo/fisiopatología , Aterosclerosis/metabolismo , Aterosclerosis/fisiopatología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Adipoquinas/metabolismo , Tejido Adiposo/patología , Animales , Aterosclerosis/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Metabolismo Energético , Humanos , Hipertensión/patología , Metabolismo de los Lípidos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Obesidad/metabolismo , Obesidad/fisiopatología , Termogénesis
11.
Arthritis Rheum ; 64(9): 2975-85, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22549550

RESUMEN

OBJECTIVE: Patients with systemic lupus erythematosus (SLE) have a notable increase in atherothrombotic cardiovascular disease (CVD) which is not explained by the Framingham risk equation. In vitro studies indicate that type I interferons (IFNs) may play prominent roles in increased CV risk in SLE. However, the in vivo relevance of these findings, with regard to the development of CVD, has not been characterized. This study was undertaken to examine the role of type I IFNs in endothelial dysfunction, aberrant vascular repair, and atherothrombosis in murine models of lupus and atherosclerosis. METHODS: Lupus-prone New Zealand mixed 2328 (NZM) mice and atherosclerosis-prone apolipoprotein E- knockout (apoE(-/-) ) mice were compared to mice lacking type I IFN receptor (INZM and apoE(-/-) IFNAR(-/-) mice, respectively) with regard to endothelial vasodilatory function, endothelial progenitor cell (EPC) function, in vivo neoangiogenesis, plaque development, and occlusive thrombosis. Similar experiments were performed using NZM and apoE(-/-) mice exposed to an IFNα-containing or empty adenovirus. RESULTS: Loss of type I IFN receptor signaling improved endothelium-dependent vasorelaxation, lipoprotein parameters, EPC numbers and function, and neoangiogenesis in lupus-prone mice, independent of disease activity or sex. Further, acute exposure to IFNα impaired endothelial vasorelaxation and EPC function in lupus-prone and non-lupus-prone mice. Decreased atherosclerosis severity and arterial inflammatory infiltrates and increased neoangiogenesis were observed in apoE(-/-) IFNAR(-/-) mice, compared to apoE(-/-) mice, while NZM and apoE(-/-) mice exposed to IFNα developed accelerated thrombosis and platelet activation. CONCLUSION: These results support the hypothesis that type I IFNs play key roles in the development of premature CVD in SLE and, potentially, in the general population, through pleiotropic deleterious effects on the vasculature.


Asunto(s)
Aterosclerosis/metabolismo , Interferón Tipo I/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Placa Aterosclerótica/metabolismo , Trombosis/metabolismo , Cicatrización de Heridas/fisiología , Animales , Aterosclerosis/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Femenino , Lupus Eritematoso Sistémico/fisiopatología , Masculino , Ratones , Vasodilatación/fisiología
12.
Eur J Case Rep Intern Med ; 10(6): 003879, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37305002

RESUMEN

Sickle cell disease is a prevalent hematologic condition, but some of the factors that lead to erythrocyte sickling are not fully known. A 58-year-old male patient with a history of sickle cell disease (SCD) and paroxysmal atrial fibrillation was transferred from an outside hospital for further management of refractory sickle cell crisis with acute chest syndrome. Before transfer, the patient received antibiotics and multiple packed red blood cell (pRBC) transfusions, with minimal effect on symptoms or anemia. After transfer, the patient developed rapid supraventricular tachycardia and atrial fibrillation (rates >160) with a drop in blood pressure. He was started on IV amiodarone. His heart rate was subsequently better controlled and converted to sinus rhythm the following day. Three days following initiation of amiodarone, the patient, with a hemoglobin count of 6.4 g/dl, required one additional unit of pRBC. On the fourth day, the patient's hemoglobin count rose to 9.4 g/dl, and he reported a marked improvement in symptoms. The improvements in symptoms and hemoglobin count were sustained, and the patient was discharged two days later. This remarkable improvement in anemia and symptoms triggered a search for potential causes. Amiodarone is a complex drug shown to have effects on multiple cell types, including erythrocytes. A recent preclinical study demonstrated reduced sickling and improved anemia in a murine model of SCD. This case report raises the possibility that amiodarone may have contributed to the rapid improvement in anemia and should be further explored in clinical trials. LEARNING POINTS: Prior studies support a link between erythrocyte sickling and membrane lipid composition.Amiodarone may impact erythrocyte pathophysiology by increasing cellular lipids including bis(mono)acylglycerol phosphate (BMP).Drugs with effects on erythrocyte lipid fractions may be beneficial during sickle cell crises.

13.
PLoS One ; 18(5): e0285499, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37235594

RESUMEN

Traumatic brain injury (TBI) is associated with cardiovascular mortality in humans. Enhanced sympathetic activity following TBI may contribute to accelerated atherosclerosis. The effect of beta1-adrenergic receptor blockade on atherosclerosis progression induced by TBI was studied in apolipoprotein E deficient mice. Mice were treated with metoprolol or vehicle following TBI or sham operation. Mice treated with metoprolol experienced a reduced heart rate with no difference in blood pressure. Six weeks following TBI, mice were sacrificed for analysis of atherosclerosis. Total surface area and lesion thickness, analyzed at the level of the aortic valve, was found to be increased in mice receiving TBI with vehicle treatment but this effect was ameliorated in TBI mice receiving metoprolol. No effect of metoprolol on atherosclerosis was observed in mice receiving only sham operation. In conclusion, accelerated atherosclerosis following TBI is reduced with beta-adrenergic receptor antagonism. Beta blockers may be useful to reduce vascular risk associated with TBI.


Asunto(s)
Aterosclerosis , Lesiones Traumáticas del Encéfalo , Animales , Ratones , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Aterosclerosis/patología , Presión Sanguínea , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Modelos Animales de Enfermedad , Metoprolol/farmacología , Metoprolol/uso terapéutico , Ratones Endogámicos C57BL
14.
Circ Res ; 107(3): 388-97, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20558823

RESUMEN

RATIONALE: Adhesive interactions between endothelial cells and leukocytes affect leukocyte trafficking in adipose tissue. The role of P-selectin glycoprotein ligand-1 (Psgl-1) in this process is unclear. OBJECTIVE: The goal of this study was to determine the effect of Psgl-1 deficiency on adhesive properties of the endothelium and on leukocyte recruitment into obese adipose depots. METHODS AND RESULTS: A genetic model of obesity was generated to study the effects of Psgl-1 deficiency on leukocyte trafficking. Leukocyte-endothelial interactions were increased in obese leptin receptor mutant mice (Lepr(db/db),Psgl-1(+/+)) but not obese Psgl-1-deficient mice (Lepr(db/db),Psgl-1(-/-)), when compared with lean mice (Lepr(+/+),Psgl-1(+/+)). This effect of Psgl-1 deficiency was due to indirect effects of Psgl-1, because Psgl-1(+/+) adoptively transferred leukocytes did not exhibit enhanced rolling in Lepr (db/db),Psgl-1(-/-) mice. Additionally, circulating levels of P-selectin, E-selectin, monocyte chemoattractant protein-1, and macrophage content of visceral adipose tissue were reduced in Lepr(db/db),Psgl-1(-/-) compared with Lepr(db/db),Psgl-1(+/+) mice. Reduced leukocyte-endothelial interactions and macrophage content of visceral adipose tissue due to Psgl-1 deficiency was also observed in a diet-induced obese mouse model. Psgl-1(-/-) mice were resistant to the endothelial effects of exogenous IL-1beta, suggesting that defective cytokine signaling contributes to the effect of Psgl-1 deficiency on leukocyte-endothelial interactions. Mice deficient in the IL-1 receptor also had reduced levels of circulating P-selectin, similar to those observed in Psgl-1(-/-) mice. CONCLUSIONS: Deficiency of Psgl-1 is associated with reduced IL-1 receptor-mediated adhesive properties of the endothelium and is protective against visceral fat inflammation in obese mice.


Asunto(s)
Tejido Adiposo/fisiología , Endotelio/fisiología , Leucocitos/fisiología , Glicoproteínas de Membrana/fisiología , Obesidad/genética , Alimentación Animal , Animales , Trasplante de Médula Ósea , Adhesión Celular , Quimiocina CCL2/sangre , Cruzamientos Genéticos , Selectina E/sangre , Femenino , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Selectina-P/sangre , Selectina-P/genética , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Receptores de Leptina/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Sci Rep ; 12(1): 16437, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36180774

RESUMEN

Sickle cell disease (SCD) is associated with altered plasma and erythrocyte lipid profiles. In a previous study, SCD mice with deficiency of proprotein convertase subtilisin/kexin type 9 (PCSK9) were observed to have more severe anemia and increased sickling compared to control SCD mice. Although PCSK9 affects circulating low density lipoprotein (LDL) by regulation of the LDL receptor, the effect of PCSK9 on anemia was independent of LDL receptor expression. In the current study, erythrocyte metabolomics were performed and revealed altered erythrocyte lipid species between SCD mice with and without PCSK9. Of particular interest, the late endosome-specific lipid bis(mono)acylglycerol phosphate (BMP) 44:12 was markedly decreased in erythrocytes from SCD mice deficient in PCSK9 mice relative to control SCD mice. Incubation of sickle erythrocytes with a neutralizing antibody to BMP increased erythrocyte sickling in vitro. In vitro treatment of SCD erythrocytes with amiodarone (1.5 µM) or medroxyprogesterone (6.75 µM), two pharmacologic compounds known to increase BMP, resulted in reduced erythrocyte sickling. Treatment of SCD mice with amiodarone (10 mg/kg) for 2 weeks resulted in increased BMP, improvement in anemia with reduced reticulocytosis, and decreased ex vivo sickling. In conclusion, severity of anemia in SCD is improved with amiodarone treatment, an effect which may be mediated through increased erythrocyte BMP.


Asunto(s)
Amiodarona , Anemia de Células Falciformes , Amiodarona/farmacología , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/tratamiento farmacológico , Animales , Anticuerpos Neutralizantes/farmacología , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Lipoproteínas LDL/metabolismo , Medroxiprogesterona/farmacología , Ratones , Monoglicéridos/metabolismo , Fosfatos/metabolismo , Proproteína Convertasa 9/metabolismo , Receptores de LDL/metabolismo , Subtilisinas/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 30(6): 1151-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20299683

RESUMEN

OBJECTIVE: To determine the role of monocyte chemoattractant protein-1 (Mcp-1) on the progression of visceral fat-induced atherosclerosis. METHODS AND RESULTS: Visceral fat inflammation was induced by transplantation of perigonadal fat. To determine whether recipient Mcp-1 status affected atherosclerosis induced by inflammatory fat, apolipoprotein E-deficient (ApoE(-/-)) and ApoE(-/-) and Mcp-1-deficient (Mcp-1(-/-)) mice underwent visceral fat transplantation. Intravital microscopy was used to study leukocyte-endothelial interactions. To study the primary tissue source of circulating Mcp-1, both fat and bone marrow transplantation experiments were used. Transplantation of visceral fat increased atherosclerosis in ApoE(-/-) mice but had no effect on atherosclerosis in ApoE(-/-),Mcp-1(-/-) mice. Intravital microscopy revealed increased leukocyte attachment to the endothelium in ApoE(-/-) mice compared with ApoE(-/-),Mcp-1(-/-) mice after receiving visceral fat transplants. Transplantation of visceral fat increased plasma Mcp-1, although donor adipocytes were not the source of circulating Mcp-1 because no Mcp-1 was detected in plasma from ApoE(-/-),Mcp-1(-/-) mice transplanted with Wt fat, indicating that recipient Mcp-1-producing cells were affecting the atherogenic response to the fat transplantation. Consistently, transplantation of Mcp-1(-/-) fat to ApoE(-/-) mice did not lead to atheroprotection in recipient mice. Bone marrow transplantation between Wt and Mcp-1(-/-) mice indicated that the primary tissue source of circulating Mcp-1 was the endothelium. CONCLUSIONS: Recipient Mcp-1 deficiency protects against atherosclerosis induced by transplanted visceral adipose tissue.


Asunto(s)
Aterosclerosis/prevención & control , Quimiocina CCL2/deficiencia , Células Endoteliales/inmunología , Grasa Intraabdominal/inmunología , Leucocitos/inmunología , Paniculitis/inmunología , Adipocitos/inmunología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Trasplante de Médula Ósea , Adhesión Celular , Quimiocina CCL2/sangre , Quimiocina CCL2/genética , Modelos Animales de Enfermedad , Grasa Intraabdominal/trasplante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía por Video , Paniculitis/complicaciones , Paniculitis/metabolismo , Factores de Tiempo
18.
Curr Opin Hematol ; 16(5): 342-6, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19550319

RESUMEN

PURPOSE OF REVIEW: The thrombotic response to vascular injury is an important clinical problem that mediates most vascular disease complications. Thrombus formation involves an integrated response that is influenced by blood flow, multiple cell types, and numerous circulating factors. As a result, modeling of this complex response using in-vitro or in-silico strategies is insufficient. The use of animal models of thrombosis provides a critical tool for the discovery and initial testing of novel therapies for vascular thrombosis. RECENT FINDINGS: The literature from 2008 to the present provides significant advances in regard to novel models of arterial thrombosis, novel mechanisms underlying thrombus formation, new models and mechanisms related to thrombotic stroke, and preclinical advances in therapeutics for vascular thrombosis. SUMMARY: The formation of occlusive thrombi is complex, involving the integration of many molecular interactions and cell types at the site of vascular injury. The identification of strategies to suppress occlusive thrombus formation without undermining normal hemostatic function is the primary goal of this area of study.


Asunto(s)
Modelos Animales de Enfermedad , Trombosis , Animales , Trombosis/diagnóstico , Trombosis/etiología , Trombosis/terapia
19.
J Am Heart Assoc ; 9(20): e019307, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32941090

RESUMEN

We, the Editors of the Journal of the American Heart Association, sincerely regret the publication of the article "Diversity, Inclusion, and Equity: Evolution of Race and Ethnicity Considerations for the Cardiology Workforce in the United States of America From 1969 to 2019".1 We are aware that the publication of this flawed and biased article has caused a great deal of unnecessary pain and anguish to a number of parties, and reflects extremely poorly on us. We fully support the retraction of this article.

20.
Circulation ; 117(6): 798-805, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18212290

RESUMEN

BACKGROUND: Fat inflammation may play an important role in comorbidities associated with obesity such as atherosclerosis. METHODS AND RESULTS: To first establish feasibility of fat transplantation, epididymal fat pads were harvested from wild-type C57BL/6J mice and transplanted into leptin-deficient (Lep(ob/ob)) mice. Fat transplantation produced physiological leptin levels and prevented obesity and infertility in Lep(ob/ob) mice. However, the transplanted fat depots were associated with chronically increased macrophage infiltration with characteristics identical to those observed in fat harvested from obese animals. The inflammation in transplanted adipose depots was regulated by the same factors that have been implicated in endogenous fat inflammation such as monocyte chemoattractant protein-1. To determine whether this inflamed adipose depot could affect vascular disease in mice, epididymal fat depots were transplanted into atherosclerosis-prone apolipoprotein E-deficient ApoE(-/-) mice. Plasma from ApoE(-/-) mice receiving fat transplants contained increased leptin, resistin, and monocyte chemoattractant protein-1 compared with plasma from sham-operated ApoE(-/-) mice. Furthermore, mice transplanted with visceral fat developed significantly more atherosclerosis compared with sham-operated animals, whereas transplants with subcutaneous fat did not affect atherosclerosis despite a similar degree of fat inflammation. Treatment of transplanted ApoE(-/-) mice with pioglitazone decreased macrophage content of the transplanted visceral fat pad and reduced plasma monocyte chemoattractant protein-1. Importantly, pioglitazone also reduced atherosclerosis triggered by inflammatory visceral fat but had no protective effect on atherosclerosis in the absence of the visceral fat transplantation. CONCLUSIONS: Our results indicate that visceral adipose-related inflammation accelerates atherosclerosis in mice. Drugs such as thiazolidinediones might be a useful strategy to specifically attenuate the vascular disease induced by visceral inflammatory fat.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/etiología , Inflamación/fisiopatología , Grasa Intraabdominal/fisiología , Tiazolidinedionas/uso terapéutico , Adiponectina/sangre , Animales , Aterosclerosis/tratamiento farmacológico , Inflamación/complicaciones , Inflamación/patología , Grasa Intraabdominal/patología , Grasa Intraabdominal/trasplante , Leptina/deficiencia , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Pioglitazona , Tiazolidinedionas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA