Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.395
Filtrar
1.
Cells ; 13(18)2024 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-39329770

RESUMEN

Atherosclerosis is an autoimmune disease characterized by lipid imbalances and chronic inflammation within blood vessels, with limited preventive and treatment options currently available. In this study, a vaccine prepared with COL6A6 peptide (named the Pep_A6 vaccine) was administered to immunize Apoe-/- mice, and the immune mechanism of the Pep_A6 vaccine against atherosclerosis was first investigated. The results of arterial oil red O staining demonstrated that the Pep_A6 vaccine significantly reduced the atherosclerotic plaque area in Apoe-/- mice fed with a high-fat diet for 20 weeks. A flow cytometry analysis revealed that the Pep_A6 vaccine inhibited Th1 cell differentiation and increased the proportion of Treg cells. Furthermore, there was a significant increase in Ly6Clow monocytes observed in the vaccinated group. The ELISA results showed that the Pep_A6 vaccine induced a significant expression of Pep_A6-specific antibody IgG and IgG1 in mouse serum. Additionally, we found that the Pep_A6 vaccine significantly decreased serum LDL-C content and regulated the expression of genes related to liver lipid metabolism. Together, our findings suggest that the Pep_A6 vaccine alleviates atherosclerosis by inducing a positive immune response and regulating lipid metabolism, providing new insights into potential prevention strategies for atherosclerosis as an innovative vaccine.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Colágeno Tipo VI , Metabolismo de los Lípidos , Vacunas de Subunidad , Animales , Aterosclerosis/inmunología , Aterosclerosis/prevención & control , Ratones , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Vacunas de Subunidad/inmunología , Colágeno Tipo VI/metabolismo , Ratones Endogámicos C57BL , Placa Aterosclerótica/inmunología , Masculino , Inmunidad , Ratones Noqueados , Dieta Alta en Grasa , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Modelos Animales de Enfermedad , Células TH1/inmunología , Vacunas de Subunidades Proteicas
2.
Sci Rep ; 14(1): 20648, 2024 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-39232217

RESUMEN

Atherosclerosis is a chronic inflammatory condition of the arteries and represents the primary cause of various cardiovascular diseases. Despite ongoing progress, finding effective anti-inflammatory therapeutic strategies for atherosclerosis remains a challenge. Here, we assessed the potential of molecular magnetic resonance imaging (MRI) to visualize the effects of 01BSUR, an anti-interleukin-1ß monoclonal antibody, for treating atherosclerosis in a murine model. Male apolipoprotein E-deficient mice were divided into a therapy group (01BSUR, 2 × 0.3 mg/kg subcutaneously, n = 10) and control group (no treatment, n = 10) and received a high-fat diet for eight weeks. The plaque burden was assessed using an elastin-targeted gadolinium-based contrast probe (0.2 mmol/kg intravenously) on a 3 T MRI scanner. T1-weighted imaging showed a significantly lower contrast-to-noise (CNR) ratio in the 01BSUR group (pre: 3.93042664; post: 8.4007067) compared to the control group (pre: 3.70679168; post: 13.2982156) following administration of the elastin-specific MRI probe (p < 0.05). Histological examinations demonstrated a significant reduction in plaque size (p < 0.05) and a significant decrease in plaque elastin content (p < 0.05) in the treatment group compared to control animals. This study demonstrated that 01BSUR hinders the progression of atherosclerosis in a mouse model. Using an elastin-targeted MRI probe, we could quantify these therapeutic effects in MRI.


Asunto(s)
Aterosclerosis , Elastina , Interleucina-1beta , Animales , Masculino , Ratones , Anticuerpos Monoclonales , Apolipoproteínas E/deficiencia , Aterosclerosis/diagnóstico por imagen , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Medios de Contraste/química , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Elastina/metabolismo , Gadolinio/química , Gadolinio/farmacología , Interleucina-1beta/metabolismo , Imagen por Resonancia Magnética/métodos , Placa Aterosclerótica/diagnóstico por imagen , Placa Aterosclerótica/tratamiento farmacológico
3.
Atherosclerosis ; 397: 118556, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39222595

RESUMEN

BACKGROUND AND AIMS: Atherosclerotic cardiovascular disease complicated by diabetes mellitus (DM) is the leading cause of death in diabetic patients, and it is strongly associated with macrophages and inflammasomes. It has been found that activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is closely associated with phosphatidylinositol 4-phosphate (PI4P) on the trans-Golgi. However, how PI4P and NLRP3 regulate macrophage function and its role in diabetic atherosclerotic plaques is unclear. METHODS: The expression of Pi4p and Nlrp3-inflammasome-related proteins in atherosclerosis in apolipoprotein E-deficient (Apoe-/-) and Apoe-/- DM mice was investigated. Then, Pi4p levels were affected by shRNA-Pi4kb or cDNA-Sac1 plasmid to investigate the effects of changes in Pi4p-related metabolic enzymes on macrophage function. Finally, genetically modified macrophages were injected into diabetic Apoe-/- mice to explore the effects on atherosclerosis. RESULTS: DM promoted plaque progression in atherosclerotic mice and increased expression of Pi4p and Nlrp3 in plaques. In addition, impaired macrophage function induced by high glucose was reversed by transfected shRNA-Pi4kb or cDNA-Sac1 plasmid. Furthermore, decreased levels of Pi4p reduced plaque area in diabetic Apoe-/- mice. CONCLUSIONS: Our data suggests that Pi4p/Nlrp3 in macrophages play an important role in the exacerbation of atherosclerosis in diabetic mice. Pi4p-related metabolizing enzymes (PI4KB and SAC1) may be a potential therapeutic strategy for diabetic atherosclerosis, and macrophage therapy is also a potential treatment.


Asunto(s)
Aterosclerosis , Diabetes Mellitus Experimental , Progresión de la Enfermedad , Macrófagos , Placa Aterosclerótica , Transducción de Señal , Animales , Masculino , Ratones , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Diabetes Mellitus Experimental/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética
4.
Nature ; 634(8033): 447-456, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39232165

RESUMEN

Systemic immune responses caused by chronic hypercholesterolaemia contribute to atherosclerosis initiation, progression and complications1. However, individuals often change their dietary habits over time2, and the effects of an alternating high-fat diet (HFD) on atherosclerosis remain unclear. Here, to address this relevant issue, we developed a protocol using atherosclerosis-prone mice to compare an alternating versus continuous HFD while maintaining similar overall exposure periods. We found that an alternating HFD accelerated atherosclerosis in Ldlr-/- and Apoe-/- mice compared with a continuous HFD. This pro-atherogenic effect of the alternating HFD was also observed in Apoe-/-Rag2-/- mice lacking T, B and natural killer T cells, ruling out the role of the adaptive immune system in the observed phenotype. Discontinuing the HFD in the alternating HFD group downregulated RUNX13, promoting inflammatory signalling in bone marrow myeloid progenitors. After re-exposure to an HFD, these cells produced IL-1ß, leading to emergency myelopoiesis and increased neutrophil levels in blood. Neutrophils infiltrated plaques and released neutrophil extracellular traps, exacerbating atherosclerosis. Specific depletion of neutrophils or inhibition of IL-1ß pathways abolished emergency myelopoiesis and reversed the pro-atherogenic effects of the alternating HFD. This study highlights the role of IL-1ß-dependent neutrophil progenitor reprogramming in accelerated atherosclerosis induced by alternating HFD.


Asunto(s)
Aterosclerosis , Reprogramación Celular , Dieta Alta en Grasa , Neutrófilos , Animales , Femenino , Masculino , Ratones , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células de la Médula Ósea/citología , Dieta Alta en Grasa/efectos adversos , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Trampas Extracelulares , Inflamación/patología , Interleucina-1beta/metabolismo , Ratones Endogámicos C57BL , Mielopoyesis , Neutrófilos/metabolismo , Neutrófilos/patología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal
5.
Atherosclerosis ; 397: 118570, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39276419

RESUMEN

BACKGROUND AND AIMS: CCN4/WISP-1 regulates various cell behaviours that contribute to atherosclerosis progression, including cell adhesion, migration, proliferation and survival. We therefore hypothesised that CCN4 regulates the development and progression of atherosclerotic plaques. METHODS: We used a high fat fed ApoE-/- mouse model to study atherosclerotic plaque progression in the brachiocephalic artery and aortic root. In protocol 1, male ApoE-/- mice with established plaques were given a CCN4 helper-dependent adenovirus to see the effect of treatment with CCN4, while in protocol 2 male CCN4-/-ApoE-/- were compared to CCN4+/+ApoE-/- mice to assess the effect of CCN4 deletion on plaque progression. RESULTS: CCN4 overexpression resulted in reduced occlusion of the brachiocephalic artery with less apoptosis, fewer macrophages, and attenuated lipid core size. The amount of plaque found on the aortic root was also reduced. CCN4 deficiency resulted in increased apoptosis and occlusion of the brachiocephalic artery as well as increased plaque in the aortic root. Additionally, in vitro cells from CCN4-/-ApoE-/- mice had higher apoptotic levels. CCN4 deficiency did not significantly affect blood cholesterol levels or circulating myeloid cell populations. CONCLUSIONS: We conclude that in an atherosclerosis model the most important action of CCN4 is the effect on cell apoptosis. CCN4 provides pro-survival signals and leads to reduced cell death, lower macrophage number, smaller lipid core size and reduced atherosclerotic plaque burden. As such, the pro-survival effect of CCN4 is worthy of further investigation, in a bid to find a therapeutic for atherosclerosis.


Asunto(s)
Apoptosis , Aterosclerosis , Placa Aterosclerótica , Animales , Masculino , Ratones , Aorta/patología , Aorta/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/patología , Aterosclerosis/metabolismo , Aterosclerosis/genética , Tronco Braquiocefálico/patología , Tronco Braquiocefálico/metabolismo , Proteínas CCN de Señalización Intercelular/metabolismo , Proteínas CCN de Señalización Intercelular/genética , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE
6.
Cell Biochem Biophys ; 82(3): 2297-2319, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39174865

RESUMEN

This study established an LPS-induced RAW264.7 macrophage inflammatory injury model and an AS mouse vulnerable plaque model to observe the effect of JPHYP on macrophage inflammation, plaque formation, blood lipids, inflammation levels, intestinal flora and the influence of TLR4/MyD88/MAPK pathway, and explore the anti-AS effect and molecular mechanism of JPHYP, and detected 16S rRNA of mice intestinal microbes. The difference of intestinal flora in different groups of mice was compared to further explore the intervention effect of JPHYP and clarify the molecular biological mechanism of JPHYP in preventing and treating AS by regulating TLR4/MyD88/MAPK inflammatory signaling pathway and improving intestinal flora.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Inflamación , Factor 88 de Diferenciación Mieloide , Receptor Toll-Like 4 , Animales , Ratones , Microbioma Gastrointestinal/efectos de los fármacos , Aterosclerosis/prevención & control , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/patología , Receptor Toll-Like 4/metabolismo , Células RAW 264.7 , Factor 88 de Diferenciación Mieloide/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Lipopolisacáridos , Modelos Animales de Enfermedad , Masculino , Transducción de Señal/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ratones Noqueados
7.
Proc Natl Acad Sci U S A ; 121(35): e2405845121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39178231

RESUMEN

Atherosclerosis is a chronic inflammatory disease of the arterial wall characterized by the accumulation of cholesterol-rich lipoproteins in macrophages. How macrophages commit to proinflammatory polarization under atherosclerosis conditions is not clear. Report here that the level of a circulating protein, leucine-rich alpha-2 glycoprotein 1 (LRG1), is elevated in the atherosclerotic tissue and serum samples from patients with coronary artery disease (CAD). LRG1 stimulated macrophages to proinflammatory M1-like polarization through the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) pathways. The LRG1 knockout mice showed significantly delayed atherogenesis progression and reduced levels of macrophage-related proinflammatory cytokines in a high-fat diet-induced Apoe-/- mouse atherosclerosis model. An anti-LRG1 neutralizing antibody also effectively blocked LRG1-induced macrophage M1-like polarization in vitro and conferred therapeutic benefits to animals with ApoE deficiency-induced atherosclerosis. LRG1 may therefore serve as an additional biomarker for CAD and targeting LRG1 could offer a potential therapeutic strategy for CAD patients by mitigating the proinflammatory response of macrophages.


Asunto(s)
Aterosclerosis , Glicoproteínas , Macrófagos , Animales , Aterosclerosis/patología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/inmunología , Macrófagos/metabolismo , Macrófagos/inmunología , Ratones , Humanos , Glicoproteínas/metabolismo , Glicoproteínas/genética , Ratones Noqueados , Masculino , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Modelos Animales de Enfermedad , Citocinas/metabolismo , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Enfermedad de la Arteria Coronaria/patología , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/inmunología , Femenino , Ratones Noqueados para ApoE , Activación de Macrófagos
8.
Lipids Health Dis ; 23(1): 247, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39138561

RESUMEN

BACKGROUND: Dysferlin-deficient limb-girdle muscular dystrophy type 2B (Dysf) mice are notorious for their mild phenotype. Raising plasma total cholesterol (CHOL) via apolipoprotein E (ApoE) knockout (KO) drastically exacerbates muscle wasting in Dysf mice. However, dysferlinopathic patients have abnormally reduced plasma high-density lipoprotein cholesterol (HDL-C) levels. The current study aimed to determine whether HDL-C lowering can exacerbate the mild phenotype of dysferlin-null mice. METHODS: Human cholesteryl ester transfer protein (CETP), a plasma lipid transfer protein not found in mice that reduces HDL-C, and/or its optimal adapter protein human apolipoprotein B (ApoB), were overexpressed in Dysf mice. Mice received a 2% cholesterol diet from 2 months of age and characterized through ambulatory and hanging functional tests, plasma analyses, and muscle histology. RESULTS: CETP/ApoB expression in Dysf mice caused reduced HDL-C (54.5%) and elevated ratio of CHOL/HDL-C (181.3%) compared to control Dysf mice in plasma, but without raising CHOL. Compared to the severe muscle pathology found in high CHOL Dysf/ApoE double knockout mice, Dysf/CETP/ApoB mice did not show significant changes in ambulation, hanging capacity, increases in damaged area, collagen deposition, or decreases in cross-sectional area and healthy myofibre coverage. CONCLUSIONS: CETP/ApoB over-expression in Dysf mice decreases HDL-C without increasing CHOL or exacerbating muscle pathology. High CHOL or nonHDL-C caused by ApoE KO, rather than low HDL-C, likely lead to rodent muscular dystrophy phenotype humanization.


Asunto(s)
Apolipoproteínas E , Proteínas de Transferencia de Ésteres de Colesterol , HDL-Colesterol , Disferlina , Ratones Noqueados , Distrofia Muscular de Cinturas , Animales , Humanos , Masculino , Ratones , Apolipoproteínas B/sangre , Apolipoproteínas B/genética , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Proteínas de Transferencia de Ésteres de Colesterol/genética , Proteínas de Transferencia de Ésteres de Colesterol/deficiencia , HDL-Colesterol/sangre , Modelos Animales de Enfermedad , Disferlina/genética , Disferlina/deficiencia , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Atrofia Muscular/metabolismo , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología
9.
Genes Immun ; 25(5): 381-388, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39103538

RESUMEN

Apolipoprotein E (ApoE) plays a crucial role in iron homeostasis in the body, while macrophages are the principal cells responsible for handling iron in mammals. However, it is unknown whether ApoE can affect the functional subtypes and the iron handling capacity of splenic macrophages (SM). Here, we investigated the effects of ApoE deficiency (ApoE-/-) on the polarization and iron content of SM and its potential mechanisms. ApoE-/- was found to induce a significant increase in the expressions of M1 marker genes CD86, IL-1ß, IL-6, IL-12, TNF-α and iNOS and a reduction in M2 marker genes CD206, Arg-1, IL-10 and Ym-1 in SM of mice aged 28 weeks, Meanwhile, ApoE-/- caused a significant increase in iron content and expression of ferritin, transferrin receptor 1 (TfR1), iron regulatory protein 1 (IRP1) and heme oxygenase-1 (HO-1) and a reduction in ferroportin1 (Fpn1) in spleen and/or SM of mice aged 28 weeks. It was concluded that ApoE-/- can increase iron content through increased iron uptake mediated by TfR/ IRPs and decreased iron release mediated by Fpn1, leading to polarization of the SM to M1 phenotype.


Asunto(s)
Apolipoproteínas E , Proteínas de Transporte de Catión , Hierro , Macrófagos , Receptores de Transferrina , Bazo , Animales , Hierro/metabolismo , Bazo/metabolismo , Bazo/citología , Ratones , Macrófagos/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/deficiencia , Receptores de Transferrina/metabolismo , Receptores de Transferrina/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Proteína 1 Reguladora de Hierro/metabolismo , Proteína 1 Reguladora de Hierro/genética , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Ratones Endogámicos C57BL , Fenotipo , Ferritinas/metabolismo , Ferritinas/genética
10.
Sci Rep ; 14(1): 18377, 2024 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-39112593

RESUMEN

The effects of calcitonin gene-related peptide (CGRP) on atherosclerosis remain unclear. We used apolipoprotein E-deficient (ApoE-/-) mice to generate double-knockout ApoE-/-:CGRP-/- mice lacking alpha CGRP. ApoE-/-:CGRP-/- mice exhibited larger atherosclerotic plaque areas, peritoneal macrophages with enhanced migration functions, and elevated levels of the inflammatory cytokine tumor necrosis factor (TNF)-⍺. Thus, we also explored whether inhibiting TNF-⍺ could improve atherosclerosis in ApoE-/-:CGRP-/- mice by administering etanercept intraperitoneally once a week (5 mg/kg) alongside a high-fat diet for 2 weeks. This treatment led to significant reductions in aortic root lesion size, atherosclerotic plaque area and macrophage migration in ApoE-/-:CGRP-/- mice compared with mice treated with human IgG (5 mg/kg). We further examined whether results observed in ApoE-/-:CGRP-/- mice could similarly be obtained by administering a humanized monoclonal CGRP antibody, galcanezumab, to ApoE-/- mice. ApoE-/- mice were subcutaneously administered galcanezumab at an initial dose of 50 mg/kg, followed by a dose of 30 mg/kg in the second week. Galcanezumab administration did not affect systolic blood pressure, serum lipid levels, or macrophage migration but led to a significant increase in lipid deposition at the aortic root. These findings suggest that alpha CGRP plays a critical role in inhibiting the progression of atherosclerosis.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Péptido Relacionado con Gen de Calcitonina , Ratones Noqueados , Placa Aterosclerótica , Animales , Aterosclerosis/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Ratones , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Placa Aterosclerótica/patología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/genética , Dieta Alta en Grasa/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo , Masculino , Ratones Noqueados para ApoE , Modelos Animales de Enfermedad , Humanos , Anticuerpos Monoclonales Humanizados/farmacología , Etanercept/farmacología , Ratones Endogámicos C57BL , Movimiento Celular/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aorta/efectos de los fármacos
11.
Atherosclerosis ; 396: 118524, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38972156

RESUMEN

BACKGROUND AND AIMS: In advanced atherosclerotic lesions, macrophage deaths result in necrotic core formation and plaque vulnerability. Cyclophilin D (CypD) is a mitochondria-specific cyclophilin involved in the process of cell death after organ ischemia-reperfusion. However, the role of CypD in atherosclerosis, especially in necrotic core formation, is unknown. Therefore, this experiment aims to clarify the role of CypD in necrotic core formation. METHODS: To clarify the specific role of CypD, encoded by Ppif in mice, apolipoprotein-E/CypD-double knockout (Apoe-/-Ppif-/-) mice were generated. These mice were fed a high-fat diet containing 0.15 % cholesterol for 24 weeks to accelerate atherosclerotic lesion development. RESULTS: Deletion of CypD decreased the necrotic core size, accompanied by a reduction of macrophage apoptosis compared to control Apoe-/- mice. In RAW264.7 cells, siRNA-mediated knockdown of CypD attenuated the release of cytochrome c from the mitochondria to the cytosol induced by endoplasmic reticulum stress inducer thapsigargin. In addition, necroptosis, induced by TNF-α and caspase inhibitor, was attenuated by knockdown of CypD. Ly-6Chigh inflammatory monocytes in peripheral blood leukocytes and mRNA expression of Il1b in the aorta were decreased by deletion of CypD. In contrast, siRNA-mediated knockdown of CypD did not significantly decrease Il1b nor Ccl2 mRNA expression in RAW264.7 cells treated with LPS and IFN-γ, suggesting that inhibition of inflammation in vivo is likely due to decreased cell death in the atherosclerotic lesions rather than a direct action of CypD deletion on the macrophage. CONCLUSIONS: These results indicate that CypD induces macrophage death and mediates necrotic core formation in advanced atherosclerotic lesions. CypD could be a novel therapeutic target for treating atherosclerotic vascular diseases.


Asunto(s)
Aterosclerosis , Macrófagos , Mitocondrias , Necrosis , Peptidil-Prolil Isomerasa F , Placa Aterosclerótica , Animales , Peptidil-Prolil Isomerasa F/metabolismo , Peptidil-Prolil Isomerasa F/genética , Macrófagos/metabolismo , Aterosclerosis/patología , Aterosclerosis/metabolismo , Aterosclerosis/genética , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Células RAW 264.7 , Modelos Animales de Enfermedad , Apoptosis , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Necroptosis , Masculino , Ratones Noqueados , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Ciclofilinas/metabolismo , Ciclofilinas/genética , Ciclofilinas/deficiencia , Dieta Alta en Grasa , Interleucina-1beta/metabolismo , Antígenos Ly
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(6): 527-531, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-38952092

RESUMEN

Objective To explore a simple and feasible method for whole-mount immunofluorescence staining of lymphatic vessels in the ApoE-/- mouse model of atherosclerosis. Methods Aortic specimens were carefully excised from the ApoE-/- mouse model. Following immunostaining with specific antibodies against smooth muscle actin (SMA) and lymphatic vessel endothelial receptor 1 (LYVE1), the aortas, including the aortic root, were subjected to a 30-minute treatment with 5 g/L Sudan Black B solution. This step was instrumental in minimizing the autofluorescent background of the tissue. Thereafter, the aortas were processed through a clearing protocol and imaged within a purpose-built chamber under a fluorescence microscope. Results The pretreatment with 5 g/L Sudan Black B effectively suppressed the autofluorescent signals emanating from the vascular structures, thereby enhancing the contrast and clarity of the specific fluorescence signals associated with the lymphatic vessels. This enhancement in signal quality did not compromise the integrity or specificity of the immunofluorescent markers. Conclusion A facile, highly specific, and effective approach for the visualization of lymphatic vessels in whole-mount aortic preparations from ApoE-/- mice is established.


Asunto(s)
Aorta , Apolipoproteínas E , Técnica del Anticuerpo Fluorescente , Vasos Linfáticos , Animales , Vasos Linfáticos/metabolismo , Vasos Linfáticos/diagnóstico por imagen , Ratones , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Adventicia/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Coloración y Etiquetado/métodos , Microscopía Fluorescente/métodos
13.
J Pineal Res ; 76(5): e12988, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38982751

RESUMEN

Vulnerable atherosclerotic plaque rupture, the leading cause of fatal atherothrombotic events, is associated with an increased risk of mortality worldwide. Peroxisome proliferator-activated receptor delta (PPARδ) has been shown to modulate vascular smooth muscle cell (SMC) phenotypic switching, and, hence, atherosclerotic plaque stability. Melatonin reportedly plays a beneficial role in cardiovascular diseases; however, the mechanisms underlying improvements in atherosclerotic plaque vulnerability remain unknown. In this study, we assessed the role of melatonin in regulating SMC phenotypic switching and its consequential contribution to the amelioration of atherosclerotic plaque vulnerability and explored the mechanisms underlying this process. We analyzed features of atherosclerotic plaque vulnerability and markers of SMC phenotypic transition in high-cholesterol diet (HCD)-fed apolipoprotein E knockout (ApoE-/-) mice and human aortic SMCs (HASMCs). Melatonin reduced atherosclerotic plaque size and necrotic core area while enhancing collagen content, fibrous cap thickness, and smooth muscle alpha-actin positive cell coverage on the plaque cap, which are all known phenotypic characteristics of vulnerable plaques. In atherosclerotic lesions, melatonin significantly decreased the synthetic SMC phenotype and KLF4 expression and increased the expression of PPARδ, but not PPARα and PPARγ, in HCD-fed ApoE-/- mice. These results were subsequently confirmed in the melatonin-treated HASMCs. Further analysis using PPARδ silencing and immunoprecipitation assays revealed that PPARδ plays a role in the melatonin-induced SMC phenotype switching from synthetic to contractile. Collectively, we provided the first evidence that melatonin mediates its protective effect against plaque destabilization by enhancing PPARδ-mediated SMC phenotypic switching, thereby indicating the potential of melatonin in treating atherosclerosis.


Asunto(s)
Factor 4 Similar a Kruppel , Melatonina , Miocitos del Músculo Liso , PPAR delta , Placa Aterosclerótica , Animales , Melatonina/farmacología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Ratones , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Factor 4 Similar a Kruppel/metabolismo , Humanos , PPAR delta/metabolismo , PPAR delta/genética , Ratones Noqueados , Masculino , Ratones Noqueados para ApoE , Fenotipo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/deficiencia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/efectos de los fármacos , Ratones Endogámicos C57BL
14.
J Cell Mol Med ; 28(10): e18402, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-39008328

RESUMEN

Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.


Asunto(s)
Aterosclerosis , Autofagia , Células Endoteliales de la Vena Umbilical Humana , Inflamación , Lipoproteínas LDL , Proteínas Qa-SNARE , Autofagia/genética , Animales , Humanos , Aterosclerosis/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/genética , Ratones , Lipoproteínas LDL/metabolismo , Técnicas de Silenciamiento del Gen , Lisosomas/metabolismo , Ratones Noqueados , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Dieta Alta en Grasa/efectos adversos , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/deficiencia
15.
Int J Mol Sci ; 25(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39000363

RESUMEN

Foods enriched with insects can potentially prevent several health disorders, including cardiovascular diseases, by reducing inflammation and improving antioxidant status. In this study, Tenebrio molitor and Gryllus assimilis were selected to determine the effect on the development of atherosclerosis in ApoE/LDLR-/- mice. Animals were fed AIN-93G-based diets (control) with 10% Tenebrio molitor (TM) and 10% Gryllus assimilis (GA) for 8 weeks. The nutritional value as well as antioxidant activity of selected insects were determined. The lipid profile, liver enzyme activity, and the fatty acid composition of liver and adipose tissue of model mice were evaluated. Quantitative analysis of atherosclerotic lesions in the entire aorta was performed using the en face method, and for aortic roots, the cross-section method was used. The antioxidant status of the GA cricket was significantly higher compared to the TM larvae. The results showed that the area of atherosclerosis (en face method) was not significantly different between groups. Dietary GA reduced plaque formation in the aortic root; additionally, significant differences were observed in sections at 200 and 300 µm compared to other groups. Furthermore, liver enzyme ALT activity was lower in insect-fed groups compared to the control group. The finding suggests that a diet containing edible insect GA potentially prevents atherosclerotic plaque development in the aortic root, due to its high antioxidant activity.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Receptores de LDL , Animales , Aterosclerosis/patología , Aterosclerosis/metabolismo , Ratones , Receptores de LDL/genética , Receptores de LDL/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Insectos Comestibles , Ratones Noqueados , Hígado/metabolismo , Hígado/patología , Antioxidantes/metabolismo , Masculino , Tenebrio , Dieta , Aorta/patología , Aorta/metabolismo , Modelos Animales de Enfermedad , Alimentación Animal , Placa Aterosclerótica/patología , Placa Aterosclerótica/metabolismo , Gryllidae
16.
J Am Heart Assoc ; 13(14): e034066, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38979792

RESUMEN

BACKGROUND: Atherosclerosis is highly prevalent in people with chronic kidney disease (CKD), including those receiving peritoneal dialysis (PD). Although it is lifesaving, PD induces profound systemic inflammation, which may aggravate atherosclerosis. Therefore, the hypothesis is that this PD-induced inflammation aggravates atherosclerosis via immune cell activation. METHODS AND RESULTS: ApoE-/- mice were subjected to a 5/6 nephrectomy to induce CKD. Three weeks later, mice were fed a high-cholesterol diet. Half of the nephrectomized mice then received daily peritoneal infusions of 3.86% Physioneal for 67 further days (CKD+PD) until the end of the experiment, and were compared with mice without CKD. Sham operated and PD-only mice were additional controls. CKD+PD mice displayed more severe atherosclerotic disease than control mice. Plaque area increased, and plaques were more advanced with a vulnerable phenotype typified by decreased collagen content and decreased fibrous cap thickness. Increased CD3+ T-cell numbers were present in plaques and perivascular adipose tissue of CKD and CKD+PD mice. Plaques of CKD+PD mice contained more iNOS+ immune cells. Spleens of CKD+PD mice showed more CD4+ central memory, terminally differentiated type 1 T-helper (Th1), Th17, and CX3C motif chemokine receptor 1+ (CX3CR1) CD4+ T-cells with less regulatory and effector T-cells. CONCLUSIONS: PD-fluid exposure in uremic mice potentiates systemic and vascular T-cell-driven inflammation and aggravates atherosclerosis. PD polarized CD4+ T-cells toward an inflammatory Th1/Th17 phenotype, and increased CX3CR1+ CD4+ T-cells, which are associated with vascular homing in CKD-associated atherosclerosis. Targeting CD4+ T-cell activation and CX3CR1+ polarization has the potential to attenuate atherosclerosis in PD patients.


Asunto(s)
Aterosclerosis , Modelos Animales de Enfermedad , Diálisis Peritoneal , Insuficiencia Renal Crónica , Uremia , Animales , Aterosclerosis/patología , Aterosclerosis/etiología , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/genética , Uremia/inmunología , Uremia/metabolismo , Diálisis Peritoneal/efectos adversos , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/metabolismo , Ratones Noqueados para ApoE , Ratones , Placa Aterosclerótica , Masculino , Ratones Endogámicos C57BL , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Nefrectomía
17.
Front Immunol ; 15: 1410832, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975335

RESUMEN

Introduction: Aging increases the risk of atherosclerotic vascular disease and its complications. Macrophages are pivotal in the pathogenesis of vascular aging, driving inflammation and atherosclerosis progression. NOX4 (NADPH oxidase 4) expression increases with age, correlating with mitochondrial dysfunction, inflammation, and atherosclerosis. We hypothesized that the NOX4-dependent mitochondrial oxidative stress promotes aging-associated atherosclerosis progression by causing metabolic dysfunction and inflammatory phenotype switch in macrophages. Methods: We studied atherosclerotic lesion morphology and macrophage phenotype in young (5-month-old) and aged (16-month-old) Nox4 -/-/Apoe -/- and Apoe -/- mice fed Western diet. Results: Young Nox4-/-/Apoe-/- and Apoe-/- mice had comparable aortic and brachiocephalic artery atherosclerotic lesion cross-sectional areas. Aged mice showed significantly increased lesion area compared with young mice. Aged Nox4-/-/Apoe-/- had significantly lower lesion areas than Apoe-/- mice. Compared with Apoe-/- mice, atherosclerotic lesions in aged Nox4-/-/Apoe-/- showed reduced cellular and mitochondrial ROS and oxidative DNA damage, lower necrotic core area, higher collagen content, and decreased inflammatory cytokine expression. Immunofluorescence and flow cytometry analysis revealed that aged Apoe-/- mice had a higher percentage of classically activated pro-inflammatory macrophages (CD38+CD80+) in the lesions. Aged Nox4-/-/Apoe-/- mice had a significantly higher proportion of alternatively activated pro-resolving macrophages (EGR2+/CD163+CD206+) in the lesions, with an increased CD38+/EGR2+ cell ratio compared with Apoe-/- mice. Mitochondrial respiration assessment revealed impaired oxidative phosphorylation and increased glycolytic ATP production in macrophages from aged Apoe-/- mice. In contrast, macrophages from Nox4-/-/Apoe-/- mice were less glycolytic and more aerobic, with preserved basal and maximal respiration and mitochondrial ATP production. Macrophages from Nox4-/-/Apoe-/- mice also had lower mitochondrial ROS levels and reduced IL1ß secretion; flow cytometry analysis showed fewer CD38+ cells after IFNγ+LPS treatment and more EGR2+ cells after IL4 treatment than in Apoe-/- macrophages. In aged Apoe-/- mice, inhibition of NOX4 activity using GKT137831 significantly reduced macrophage mitochondrial ROS and improved mitochondrial function, resulting in decreased CD68+CD80+ and increased CD163+CD206+ lesion macrophage proportion and attenuated atherosclerosis. Discussion: Our findings suggest that increased NOX4 in aging drives macrophage mitochondrial dysfunction, glycolytic metabolic switch, and pro-inflammatory phenotype, advancing atherosclerosis. Inhibiting NOX4 or mitochondrial dysfunction could alleviate vascular inflammation and atherosclerosis, preserving plaque integrity.


Asunto(s)
Envejecimiento , Aterosclerosis , Macrófagos , Mitocondrias , NADPH Oxidasa 4 , Fenotipo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/etiología , Aterosclerosis/inmunología , Mitocondrias/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Envejecimiento/inmunología , NADPH Oxidasa 4/metabolismo , NADPH Oxidasa 4/genética , Progresión de la Enfermedad , Ratones Noqueados , Estrés Oxidativo , Inflamación/inmunología , Inflamación/metabolismo , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Masculino , Modelos Animales de Enfermedad , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Ratones Noqueados para ApoE , Reprogramación Metabólica
18.
Redox Biol ; 74: 103229, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38870781

RESUMEN

BACKGROUND: Nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor, is critically involved in the regulation of oxidative stress and inflammation. However, the role of endothelial Nrf2 in atherogenesis has yet to be defined. In addition, how endothelial Nrf2 is activated and whether Nrf2 can be targeted for the prevention and treatment of atherosclerosis is not explored. METHODS: RNA-sequencing and single-cell RNA sequencing analysis of mouse atherosclerotic aortas were used to identify the differentially expressed genes. In vivo endothelial cell (EC)-specific activation of Nrf2 was achieved by injecting adeno-associated viruses into ApoE-/- mice, while EC-specific knockdown of Nrf2 was generated in Cdh5CreCas9floxed-stopApoE-/- mice. RESULTS: Endothelial inflammation appeared as early as on day 3 after feeding of a high cholesterol diet (HCD) in ApoE-/- mice, as reflected by mRNA levels, immunostaining and global mRNA profiling, while the immunosignal of the end-product of lipid peroxidation (LPO), 4-hydroxynonenal (4-HNE), started to increase on day 10. TNF-α, 4-HNE, and erastin (LPO inducer), activated Nrf2 signaling in human ECs by increasing the mRNA and protein expression of Nrf2 target genes. Knockdown of endothelial Nrf2 resulted in augmented endothelial inflammation and LPO, and accelerated atherosclerosis in Cdh5CreCas9floxed-stopApoE-/- mice. By contrast, both EC-specific and pharmacological activation of Nrf2 inhibited endothelial inflammation, LPO, and atherogenesis. CONCLUSIONS: Upon HCD feeding in ApoE-/- mice, endothelial inflammation is an earliest event, followed by the appearance of LPO. EC-specific activation of Nrf2 inhibits atherosclerosis while EC-specific knockdown of Nrf2 results in the opposite effect. Pharmacological activators of endothelial Nrf2 may represent a novel therapeutic strategy for the treatment of atherosclerosis.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Células Endoteliales , Inflamación , Peroxidación de Lípido , Factor 2 Relacionado con NF-E2 , Animales , Humanos , Masculino , Ratones , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Inflamación/metabolismo , Inflamación/genética , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo
19.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38892314

RESUMEN

GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.


Asunto(s)
Apolipoproteínas E , Aterosclerosis , Enfermedades Periodontales , Porphyromonas gingivalis , Animales , Ratones , Apolipoproteínas E/deficiencia , Enfermedades Periodontales/microbiología , Enfermedades Periodontales/prevención & control , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Aterosclerosis/microbiología , Telomerasa/metabolismo , Fragmentos de Péptidos/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/microbiología , Periodontitis/microbiología , Periodontitis/prevención & control , Infecciones por Bacteroidaceae/microbiología , Infecciones por Bacteroidaceae/complicaciones , Infecciones por Bacteroidaceae/prevención & control , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino , Humanos
20.
Biomolecules ; 14(6)2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38927075

RESUMEN

Atherosclerosis (AS) has become the leading cause of cardiovascular disease worldwide. Our previous study had observed that Nippostrongylus brasiliensis (Nb) infection or its derived products could inhibit AS development by inducing an anti-inflammatory response. We performed a metabolic analysis to screen Nb-derived metabolites with anti-inflammation activity and evaluated the AS-prevention effect. We observed that the metabolite uridine had higher expression levels in mice infected with the Nb and ES (excretory-secretory) products and could be selected as a key metabolite. ES and uridine interventions could reduce the pro-inflammatory responses and increase the anti-inflammatory responses in vitro and in vivo. The apolipoprotein E gene knockout (ApoE-/-) mice were fed with a high-fat diet for the AS modeling. Following the in vivo intervention, ES products or uridine significantly reduced serum and liver lipid levels, alleviated the formation of atherosclerosis, and reduced the pro-inflammatory responses in serum or plaques, while the anti-inflammatory responses showed opposite trends. After blocking with 5-HD (5-hydroxydecanoate sodium) in vitro, the mRNA levels of M2 markers were significantly reduced. When blocked with 5-HD in vivo, the degree of atherosclerosis was worsened, the pro-inflammatory responses were increased compared to the uridine group, while the anti-inflammatory responses decreased accordingly. Uridine, a key metabolite from Nippostrongylus brasiliensis, showed anti-inflammatory and anti-atherosclerotic effects in vitro and in vivo, which depend on the activation of the mitochondrial ATP-sensitive potassium channel.


Asunto(s)
Antiinflamatorios , Aterosclerosis , Nippostrongylus , Uridina , Animales , Masculino , Ratones , Antiinflamatorios/farmacología , Apolipoproteínas E/genética , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Aterosclerosis/genética , Modelos Animales de Enfermedad , Canales KATP/metabolismo , Canales KATP/genética , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Uridina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA