Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 853
Filtrar
1.
Exp Parasitol ; 263-264: 108800, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39043326

RESUMEN

The infectivity of Leishmania is determined by its ability to invade and evade host and its thriving capacity within the macrophage. Our study revealed the role of Leishmania donovani mevalonate kinase (MVK), an enzyme of mevalonate pathway in visceral leishmaniasis pathogenesis. Peritoneal exudate cells (PEC)-derived macrophages from BALB/c mice were infected with wild type (WT), MVK over expressing (MVK OE) and knockdown (KD) parasites and MVK OE parasites were found to be more infective than WT and MVK KD parasites. Incubation of macrophages with MVK OE parasites declined inducible nitric oxide synthase (iNOS) expression as well as nitric oxide (NO) production, both by 2 times in comparison to WT parasites. Moreover, ∼3 fold increase in Arginase1 expression indicated that MVK might induce polarization of macrophage towards M2, favouring the survival of parasite within the macrophages. Post 24 h infection of the macrophages with mutant strains, the levels of different cytokines (TNF-α, IL-12, IL-10 and IFN-γ) were measured. Infection of macrophages with MVK OE parasites showed an increase in the level of anti-inflammatory cytokine: IL-10 while infection with MVK KD parasites exhibited an increase in the level of pro-inflammatory cytokines: TNF-α, IL-12, and IFN-γ. Hence, Leishmania donovani mevalonate kinase (LdMVK) modulates macrophage functions and has a significant role in pathogenesis.


Asunto(s)
Citocinas , Leishmania donovani , Leishmaniasis Visceral , Macrófagos Peritoneales , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico , Fosfotransferasas (Aceptor de Grupo Alcohol) , Leishmania donovani/enzimología , Leishmania donovani/patogenicidad , Leishmania donovani/genética , Leishmania donovani/fisiología , Animales , Ratones , Leishmaniasis Visceral/parasitología , Leishmaniasis Visceral/inmunología , Óxido Nítrico/metabolismo , Macrófagos Peritoneales/parasitología , Macrófagos Peritoneales/enzimología , Citocinas/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Arginasa/metabolismo , Arginasa/genética , Femenino , Técnicas de Silenciamiento del Gen
2.
Microvasc Res ; 140: 104276, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34742813

RESUMEN

PURPOSE: We previously reported that a calpain inhibitor (CAI) prevents the development of atherosclerosis in rats. This study aimed to investigate the effects of CAI (1 mg/kg) on atherosclerosis in apolipoprotein E knockout (ApoE KO) mice that were fed a high-fat diet (HFD) and explore the underlying mechanism by analyzing the expression of genes related to the uptake and efflux of cholesterol. METHODS: Atherosclerotic plaques were evaluated. The activity of calpain in the aorta and that of superoxide dismutase (SOD) in the serum were assessed. Lipid profiles in the serum and liver were examined. Serum oxidized low-density lipoprotein (oxLDL), malondialdehyde (MDA), tumor necrosis factor (TNF-α), and interleukin-6 (IL-6) levels were measured. The mRNA expressions of CD68, TNF-α, IL-6, CD36, scavenger receptor (SR-A), peroxisome proliferator-activated receptor gamma (PPAR-γ), liver-x-receptor alpha (LXR-α), and ATP-binding cassette transporter class A1 (ABCA1) in the aorta and peritoneal macrophages were also evaluated. RESULTS: CAI reduced calpain activity in the aorta. CAI also impeded atherosclerotic lesion formation and mRNA expression of CD68 in the aorta and peritoneal macrophages of ApoE KO mice compared with those of mice receiving HFD. However, CAI had no effect on body weight and lipid levels in both the serum and liver. CAI significantly decreased MDA, oxLDL, TNF-α, and IL-6 levels and increased SOD activity in the serum. Moreover, CAI significantly inhibited the mRNA expression of TNF-α and IL-6 genes in the aorta and peritoneal macrophages. In addition, CAI significantly downregulated the mRNA expression of scavenger receptors CD36 and SR-A and upregulated the expression of genes involved in the cholesterol efflux pathway, i.e., PPAR-γ, LXR-α, and ABCA1 in the aorta and peritoneal macrophages. CONCLUSIONS: CAI inhibited the development of atherosclerotic lesions in ApoE KO mice, and this effect might be related to the reduction of oxidative stress and inflammation and the improvement of cholesterol intake and efflux pathways.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Calpaína/antagonistas & inhibidores , Colesterol/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Leupeptinas/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , ARN Mensajero/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Calpaína/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Metabolismo de los Lípidos/genética , Receptores X del Hígado/genética , Receptores X del Hígado/metabolismo , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , PPAR gamma/genética , PPAR gamma/metabolismo , Placa Aterosclerótica , ARN Mensajero/genética , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/metabolismo
4.
Biomed Pharmacother ; 138: 111543, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34311538

RESUMEN

Acute lung injury (ALI) is a severe lung disease with limited therapeutic strategies. Munronoid I, a limonoid, which is extracted and purified from Munronia sinica, exhibits effective anti-neoplastic activities. In this study, we attempted to determine the anti-inflammatory effects of Munronoid I using both the lipopolysaccharide (LPS)-induced in vivo murine ALI models and in vitro assays. Our results demonstrated that Munronoid I treatment ameliorated LPS-induced ALI and inflammation in mice. Moreover, it also significantly inhibited LPS-induced pathological injuries, infiltration of inflammatory cells, and production of IL-1ß and IL-6. Furthermore, the in vitro assay showed that Munronoid I could inhibit the LPS-induced expression of inflammatory mediators such as iNOS, COX2, and production of pro-inflammatory cytokines by suppressing the activation of NF-κB signaling pathway in mouse peritoneal macrophages. Munronoid I reduced the LPS-, tumor necrosis factor alpha (TNF-α)- or interleukin 1 beta (IL-1ß)-induced transforming growth factor beta-activated kinase 1 (TAK1) phosphorylation and protein expression. Furthermore, the Munronoid I also promoted K48-linked ubiquitination and proteasomal degradation of TAK1. Taken together, these results demonstrated that Munronoid I exhibited anti-inflammatory activities both in vitro and in vivo, which might be a potential therapeutic candidate for the treatment of ALI and pulmonary inflammation.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/farmacología , Limoninas/farmacología , Pulmón/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/patología , Animales , Antiinflamatorios/aislamiento & purificación , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Limoninas/aislamiento & purificación , Lipopolisacáridos , Pulmón/enzimología , Pulmón/patología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosforilación , Proteolisis , Ubiquitinación
6.
Cell Death Dis ; 12(2): 135, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33542207

RESUMEN

Despite the progress to understand inflammatory reactions, mechanisms causing their resolution remain poorly understood. Prostanoids, especially prostaglandin E2 (PGE2), are well-characterized mediators of inflammation. PGE2 is produced in an inducible manner in macrophages (Mϕ) by microsomal PGE2-synthase-1 (mPGES-1), with the notion that it also conveys pro-resolving properties. We aimed to characterize the role of mPGES-1 during resolution of acute, zymosan-induced peritonitis. Experimentally, we applied the mPGES-1 inhibitor compound III (CIII) once the inflammatory response was established and confirmed its potent PGE2-blocking efficacy. mPGES-1 inhibition resulted in an incomplete removal of neutrophils and a concomitant increase in monocytes and Mϕ during the resolution process. The mRNA-seq analysis identified enhanced C-X3-C motif receptor 1 (CX3CR1) expression in resident and infiltrating Mϕ upon mPGES-1 inhibition. Besides elevated Cx3cr1 expression, its ligand CX3CL1 was enriched in the peritoneal lavage of the mice, produced by epithelial cells upon mPGES-1 inhibition. CX3CL1 not only increased adhesion and survival of Mϕ but its neutralization also completely reversed elevated inflammatory cell numbers, thereby normalizing the cellular, peritoneal composition during resolution. Our data suggest that mPGES-1-derived PGE2 contributes to the resolution of inflammation by preventing CX3CL1-mediated retention of activated myeloid cells at sites of injury.


Asunto(s)
Quimiocina CX3CL1/metabolismo , Dinoprostona/metabolismo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Peritonitis/enzimología , Prostaglandina-E Sintasas/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/farmacología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Adhesión Celular , Supervivencia Celular , Células Cultivadas , Quimiocina CX3CL1/antagonistas & inhibidores , Quimiocina CX3CL1/genética , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Femenino , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Ratones Endogámicos C57BL , Infiltración Neutrófila , Peritonitis/genética , Peritonitis/inmunología , Fenotipo , Prostaglandina-E Sintasas/metabolismo , Regulación hacia Arriba
7.
Clin Sci (Lond) ; 135(1): 19-34, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33399849

RESUMEN

Type 1 diabetes mellitus (T1D) is a chronic autoimmune disease characterized by insulin-producing pancreatic ß-cell destruction and hyperglycemia. While monocytes and NOD-like receptor family-pyrin domain containing 3 (NLRP3) are associated with T1D onset and development, the specific receptors and factors involved in NLRP3 inflammasome activation remain unknown. Herein, we evaluated the inflammatory state of resident peritoneal macrophages (PMs) from genetically modified non-obese diabetic (NOD), NLRP3-KO, wild-type (WT) mice and in peripheral blood mononuclear cells (PBMCs) from human T1D patients. We also assessed the effect of docosahexaenoic acid (DHA) on the inflammatory status. Macrophages from STZ-induced T1D mice exhibited increased inflammatory cytokine/chemokine levels, nitric oxide (NO) secretion, NLRP3 and iNOS protein levels, and augmented glycolytic activity compared to control animals. In PMs from NOD and STZ-induced T1D mice, DHA reduced NO production and attenuated the inflammatory state. Furthermore, iNOS and IL-1ß protein expression levels and NO production were lower in the PMs from diabetic NLRP3-KO mice than from WT mice. We also observed increased IL-1ß secretion in PBMCs from T1D patients and immortalized murine macrophages treated with advanced glycation end products and palmitic acid. The present study demonstrated that the resident PMs are in a proinflammatory state characterized by increased NLRP3/iNOS pathway-mediated NO production, up-regulated proinflammatory cytokine/chemokine receptor expression and altered glycolytic activity. Notably, ex vivo treatment with DHA reverted the diabetes-induced changes and attenuated the macrophage inflammatory state. It is plausible that DHA supplementation could be employed as adjuvant therapy for treating individuals with T1D.


Asunto(s)
Antiinflamatorios/farmacología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Ácidos Docosahexaenoicos/farmacología , Inflamación/tratamiento farmacológico , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Adulto , Animales , Células Cultivadas , Citocinas/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Femenino , Humanos , Inflamación/inducido químicamente , Inflamación/enzimología , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Persona de Mediana Edad , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Embarazo , Transducción de Señal , Estreptozocina
8.
Front Immunol ; 11: 1893, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973784

RESUMEN

Excessive nitric oxide (NO) production and NO-mediated nitrative stress contribute to vascular dysfunction, inflammation, and tissue injury in septic shock. New therapeutic targets are urgently needed to provide better control of NO level during septic shock. In the present study, we investigated the role of HDAC6 in the regulation of NO production and nitrative stress in a mouse model of endotoxin-induced septic shock. HDAC6 deficient mice and a specific HDAC6 inhibitor were utilized in our studies. Our data clearly indicate that HDAC6 is an important mediator of NO production in macrophages. HDAC6 mediates NO production through the regulation of iNOS expression in macrophages. HDAC6 up-regulates iNOS expression in macrophages by modulating STAT1 activation and IRF-1 expression. HDAC6 inhibition potently blocked endotoxin-induced STAT1 activation and iNOS expression in macrophages. Furthermore, HDAC6 contributes to excessive NO production and nitrotyrosine level in the blood and promotes iNOS expression in the lung tissues during septic shock. Our data reveal a novel HDAC6/STAT1/iNOS pathway that mediates excessive NO production and nitrative stress in septic shock.


Asunto(s)
Endotoxemia/enzimología , Histona Desacetilasa 6/metabolismo , Macrófagos Peritoneales/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/sangre , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Endotoxemia/sangre , Endotoxemia/genética , Endotoxemia/prevención & control , Histona Desacetilasa 6/antagonistas & inhibidores , Histona Desacetilasa 6/deficiencia , Histona Desacetilasa 6/genética , Inhibidores de Histona Desacetilasas/farmacología , Factor 1 Regulador del Interferón/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Tirosina/análogos & derivados , Tirosina/sangre
9.
J Leukoc Biol ; 108(1): 283-295, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32237257

RESUMEN

1,25-dihydroxyvitamin D3 (1,25(OH)2 D3, VitD3) is the major active ingredient of vitamin D and has anti-inflammatory activity; however, the mechanism for this remains poorly understood. In this study, we found that VitD3 was able to abolish NOD-like receptor protein 3 (NLRP3) inflammasome activation and subsequently inhibit caspase-1 activation and IL-1ß secretion via the vitamin D receptor (VDR). Furthermore, VitD3 specifically prevented NLRP3-mediated apoptosis-associated speck-like protein with a caspase-recruitment domain (ASC) oligomerization. In additional to this, NLRP3 binding to NIMA-related kinase 7 (NEK7) was also inhibited. Notably, VitD3 inhibited autophagy, leading to the inhibition of the NLRP3 inflammasome. Uncoupling protein 2-reactive oxygen species signaling may be involved in inflammasome suppression by VitD3. Importantly, VitD3 had both preventive and therapeutic effects on mouse model of ulcerative colitis, via inhibition of NLRP3 inflammasome activation. Our results reveal a mechanism through which VitD3 represses inflammation and prevents the relevant diseases, and suggest a potential clinical use of VitD3 in autoimmune syndromes or other NLRP3 inflammasome-driven inflammatory diseases.


Asunto(s)
Calcitriol/uso terapéutico , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Autofagia/efectos de los fármacos , Proteínas Adaptadoras de Señalización CARD/metabolismo , Calcitriol/farmacología , Caspasa 1/metabolismo , Polaridad Celular/efectos de los fármacos , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/inmunología , Colon/efectos de los fármacos , Colon/patología , Sulfato de Dextran , Activación Enzimática/efectos de los fármacos , Interleucina-1beta/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Ratones , Quinasas Relacionadas con NIMA/metabolismo , Proteolisis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología , Ubiquitinación/efectos de los fármacos , Proteína Desacopladora 2/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 40(5): 1155-1167, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212851

RESUMEN

OBJECTIVES: During the advancement of atherosclerosis, plaque cellularity is governed by the influx of monocyte-derived macrophages and their turnover via apoptotic and nonapoptotic forms of cell death. Previous reports have demonstrated that programmed necrosis, or necroptosis, of plaque macrophages contribute to necrotic core formation. Knockdown or inhibition of the necrosome components RIPK1 (receptor-interacting protein kinase 1) and RIPK3 (receptor-interacting protein kinase 3) slow atherogenesis, and activation of the terminal step of necroptosis, MLKL (mixed lineage kinase domain-like protein), has been demonstrated in advanced human atherosclerotic plaques. However, whether MLKL directly contributes to lesion development and necrotic core formation has not been investigated. Approaches and Results: MLKL expression was knocked down in atherogenic Apoe-knockout mice via the administration of antisense oligonucleotides. During atherogenesis, Mlkl knockdown decreased both programmed cell death and the necrotic core in the plaque. However, total lesion area remained unchanged. Furthermore, treatment with the MLKL antisense oligonucleotide unexpectedly reduced circulating cholesterol levels compared with control antisense oligonucleotide but increased the accumulation of lipids within the plaque and in vitro in macrophage foam cells. MLKL colocalized with the late endosome and multivesicular bodies in peritoneal macrophages incubated with atherogenic lipoproteins. Transfection with MLKL antisense oligonucleotide increased lipid localization with the multivesicular bodies, suggesting that upon Mlkl knockdown, lipid trafficking becomes defective leading to enhanced lipid accumulation in macrophages. CONCLUSIONS: These studies confirm the requirement for MLKL as the executioner of necroptosis, and as such a significant contributor to the necrotic core during atherogenesis. We also identified a previously unknown role for MLKL in regulating endosomal trafficking to facilitate lipid handling in macrophages during atherogenesis.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Colesterol/metabolismo , Células Espumosas/enzimología , Macrófagos Peritoneales/enzimología , Placa Aterosclerótica , Proteínas Quinasas/deficiencia , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Modelos Animales de Enfermedad , Endosomas/metabolismo , Femenino , Células Espumosas/patología , Macrófagos Peritoneales/patología , Masculino , Ratones Noqueados para ApoE , Necroptosis , Necrosis , Oligonucleótidos Antisentido/administración & dosificación , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal
11.
J Am Heart Assoc ; 9(3): e013933, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-32013704

RESUMEN

Background Recent studies have highlighted a critical role for a group of natriuretic hormones, cardiotonic steroid (CTS), in mediating renal inflammation and fibrosis associated with volume expanded settings, such as chronic kidney disease. Immune cell adhesion is a critical step in the inflammatory response; however, little is currently understood about the potential regulatory role of CTS signaling in this setting. Herein, we tested the hypothesis that CTS signaling through Na+/K+-ATPase α-1 (NKA α-1) enhances immune cell recruitment and adhesion to renal epithelium that ultimately advance renal inflammation. Methods and Results We demonstrate that knockdown of the α-1 isoform of Na/K-ATPase causes a reduction in CTS-induced macrophage infiltration in renal tissue as well reduces the accumulation of immune cells in the peritoneal cavity in vivo. Next, using functional adhesion assay, we demonstrate that CTS-induced increases in the adhesion of macrophages to renal epithelial cells were significantly diminished after reduction of NKA α-1 in either macrophages or renal epithelial cells as well after inhibition of NKA α-1-Src signaling cascade with a specific peptide inhibitor, pNaKtide in vitro. Finally, CTS-induced expression of adhesion markers in both endothelial and immune cells was significantly inhibited in an NKA α-1-Src signaling dependent manner in vitro. Conclusions These findings suggest that CTS potentiates immune cell migration and adhesion to renal epithelium through an NKA α-1-dependent mechanism; our new findings suggest that pharmacological inhibition of this feed-forward loop may be useful in the treatment of renal inflammation associated with renal disease.


Asunto(s)
Bufanólidos/farmacología , Cardiotónicos/farmacología , Adhesión Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Técnicas de Cocultivo , Células Endoteliales/enzimología , Células Epiteliales/enzimología , Humanos , Túbulos Renales Proximales/enzimología , Células LLC-PK1 , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Ratones Noqueados , Ratas Endogámicas Dahl , Transducción de Señal , ATPasa Intercambiadora de Sodio-Potasio/deficiencia , ATPasa Intercambiadora de Sodio-Potasio/genética , Porcinos , Familia-src Quinasas/metabolismo
12.
J Biol Chem ; 294(43): 15836-15849, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31495784

RESUMEN

Cholesterol esters are a key ingredient of foamy cells in atherosclerotic lesions; their formation is catalyzed by two enzymes: acyl-CoA:cholesterol acyltransferases (ACATs; also called sterol O-acyltransferases, or SOATs) ACAT1 and ACAT2. ACAT1 is present in all body cells and is the major isoenzyme in macrophages. Whether blocking ACAT1 benefits atherosclerosis has been under debate for more than a decade. Previously, our laboratory developed a myeloid-specific Acat1 knockout (KO) mouse (Acat1-M/-M), devoid of ACAT1 only in macrophages, microglia, and neutrophils. In previous work using the ApoE KO (ApoE-/-) mouse model for early lesions, Acat1-M/-M significantly reduced lesion macrophage content and suppressed atherosclerosis progression. In advanced lesions, cholesterol crystals become a prominent feature. Here we evaluated the effects of Acat1-M/-M in the ApoE KO mouse model for more advanced lesions and found that mice lacking myeloid Acat1 had significantly reduced lesion cholesterol crystal contents. Acat1-M/-M also significantly reduced lesion size and macrophage content without increasing apoptotic cell death. Cell culture studies showed that inhibiting ACAT1 in macrophages caused cells to produce less proinflammatory responses upon cholesterol loading by acetyl low-density lipoprotein. In advanced lesions, Acat1-M/-M reduced but did not eliminate foamy cells. In advanced plaques isolated from ApoE-/- mice, immunostainings showed that both ACAT1 and ACAT2 are present. In cell culture, both enzymes are present in macrophages and smooth muscle cells and contribute to cholesterol ester biosynthesis. Overall, our results support the notion that targeting ACAT1 or targeting both ACAT1 and ACAT2 in macrophages is a novel strategy to treat advanced lesions.


Asunto(s)
Aterosclerosis/enzimología , Aterosclerosis/prevención & control , Inflamación/patología , Macrófagos Peritoneales/enzimología , Células Mieloides/enzimología , Esterol O-Aciltransferasa/metabolismo , Animales , Apolipoproteínas E , Apoptosis , Aterosclerosis/patología , Colesterol/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Silenciador del Gen , Hidroxicolesteroles/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Células Mieloides/patología , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Células RAW 264.7
13.
PLoS One ; 13(12): e0208279, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30571701

RESUMEN

Multiple organ dysfunction caused by hyperinflammation remains the major cause of mortality during sepsis. Excessive M1-macrophage activation leads to systemic inflammatory responses. Gene related to anergy in lymphocytes (Grail) is regarded as an important regulator of T cells that functions by diminishing cytokine production. However, its role in regulating macrophage activation and organ injury during sepsis remains unclear. Our aim was to examine the effects of Grail on macrophage reactivity and organ injury in endotoxemic animals. Wild-type and Grail knockout mice were injected with vehicle or Escherichia coli lipopolysaccharide and observed for 24 h. Changes in blood pressure, heart rate, blood glucose, and biochemical variables were then examined. Moreover, levels of neutrophil infiltration, MMP-9, and caspase 3 were analyzed in the lungs of animals. The expression of pro-inflammatory cytokines in J774A, RAW264.7, and primary peritoneal macrophages stimulated with LPS were also assessed in the presence or absence of Grail. Results indicated that loss of Grail expression enhances the induction of pro-inflammatory cytokines in J774A, RAW264.7, and primary peritoneal macrophages treated with LPS. Furthermore, LPS-induced macrophage hyperactivation was alleviated by ectopic Grail overexpression. In vivo studies showed that Grail deficiency exacerbates organ damage in endotoxemic animals. Levels of neutrophil infiltration, MMP-9, and caspase 3 were significantly increased in the lungs of Grail-deficient endotoxemic mice. Thus, these results suggest that Grail contributes to the attenuation of hyperinflammation caused by activated macrophages and prevents organ damage in endotoxemic mice. We suggest that Grail signaling could be a therapeutic target for endotoxemia.


Asunto(s)
Inflamación/enzimología , Inflamación/prevención & control , Macrófagos Peritoneales/enzimología , Especificidad de Órganos , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Peso Corporal , Caspasa 3/metabolismo , Creatinina/sangre , Citocinas/metabolismo , Endotoxemia/sangre , Endotoxemia/metabolismo , Endotoxemia/patología , Eliminación de Gen , Hemodinámica , Inflamación/patología , Mediadores de Inflamación/metabolismo , L-Lactato Deshidrogenasa/sangre , Lipopolisacáridos , Pulmón/enzimología , Pulmón/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Células RAW 264.7
14.
Exp Gerontol ; 113: 86-94, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30287187

RESUMEN

The systemic and extra- gonadal levels of 17ß-estradiol (E2) change during aging, and affect the expression of estrogen receptors (ERs) in the immune cells of both females and males. The age-related cessation of ovarian function in females, as well as the tissue-specific expression of enzyme aromatase (estrogen synthase which significantly rises with the advancing age) in both males and females, both determine the concentration of E2 to which immune cells may be exposed. The present study was set up to investigate the direct influence of E2 in vitro on the secretory profile of peritoneal macrophages from young and naturally menopausal female rats, and from young and middle-aged male rats. The involvement of receptor(s) responsible for mediating the effects of E2 in vitro was examined by use of antagonists specific for ERα or ERß. Whereas in macrophages from young female rats E2 treatment diminished interleukin (IL)-1ß secretion, it increased it in young males, and the middle-aged females. The in vitro E2 treatment increased tumor necrosis factor (TNF)-α release by macrophages from young rats of both sexes, while it increased macrophage IL-6 release independently of both sex and age. At the same time, E2 decreased hydrogen peroxide (H2O2) production in macrophages from females, and increased it in male rats of both ages, whereas it diminished nitric oxide (NO) release in all experimental groups. Inspite of the sex- and age-specific effects of E2 on macrophage urea release, E2 did not affect the NO/urea ratio in macrophages from female rats, and diminished it in macrophages from both young and middle-aged male rats. Independently of the sex and age, E2 stimulated the release of inflammatory cytokines predominantly via macrophage ERα, and inhibited the IL-1ß release in young females via ERß. In contrast, E2 increased macrophage H2O2 and urea production by activating ERß, but diminished their release via ERα. Our study may contribute to better understanding of the complex role(s) that E2 may play in innate immunity during aging, and that are dependent of sex.


Asunto(s)
Envejecimiento/metabolismo , Aromatasa/efectos de los fármacos , Estradiol/farmacología , Macrófagos Peritoneales/enzimología , Animales , Células Cultivadas , Citocinas/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Peróxido de Hidrógeno/metabolismo , Inmunidad Innata/efectos de los fármacos , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Óxido Nítrico/metabolismo , Ratas , Receptores de Estrógenos/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 38(11): 2576-2589, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30354239

RESUMEN

Objective- ACAT1 (Acyl-CoA cholesterol acyltransferase 1) esterifies cellular free cholesterol, thereby converting macrophages to cholesteryl ester-laden foam cells in atherosclerotic lesions and cutaneous xanthoma. Paradoxically, however, loss of ACAT1 in bone marrow causes the aggravation of atherosclerosis and the development of severe cutaneous xanthoma in hyperlipidemic mice. Recently, it has been reported that cholesterol crystals activate NLRP3 (NACHT, LRR [leucine-rich repeats], and PYD [pyrin domain] domain-containing protein 3) inflammasomes, thereby contributing to the development of atherosclerosis. The present study aimed to clarify the role of NLRP3 inflammasomes in the worsening of atherosclerosis and cutaneous xanthoma induced by ACAT1 deficiency. Approach and Results- Ldlr-null mice were transplanted with bone marrow from WT (wild type) mice and mice lacking ACAT1, NLRP3, or both. After the 4 types of mice were fed high-cholesterol diets, we compared their atherosclerosis and skin lesions. The mice transplanted with Acat1-null bone marrow developed severe cutaneous xanthoma, which was filled with numerous macrophages and cholesterol clefts and had markedly increased expression of inflammatory cytokines, and increased atherosclerosis. Loss of NLRP3 completely reversed the cutaneous xanthoma, whereas it improved the atherosclerosis only partially. Acat1-null peritoneal macrophages showed enhanced expression of CHOP (C/EBP [CCAAT/enhancer binding protein] homologous protein) and TNF-α (tumor necrosis factor-α) but no evidence of inflammasome activation, after treatment with acetylated LDL (low-density lipoprotein). Conclusions- Elimination of ACAT1 in bone marrow-derived cells aggravates cutaneous xanthoma and atherosclerosis. The development of cutaneous xanthoma is induced mainly via the NLRP3 inflammasome activation.


Asunto(s)
Acetil-CoA C-Acetiltransferasa/metabolismo , Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Médula Ósea/enzimología , Inflamasomas/metabolismo , Macrófagos Peritoneales/enzimología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Placa Aterosclerótica , Enfermedades de la Piel/enzimología , Xantomatosis/enzimología , Acetil-CoA C-Acetiltransferasa/deficiencia , Acetil-CoA C-Acetiltransferasa/genética , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Médula Ósea/patología , Trasplante de Médula Ósea , Células Cultivadas , Colesterol en la Dieta , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Fenotipo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal , Enfermedades de la Piel/genética , Enfermedades de la Piel/patología , Enfermedades de la Piel/prevención & control , Xantomatosis/genética , Xantomatosis/patología , Xantomatosis/prevención & control
16.
Arterioscler Thromb Vasc Biol ; 38(11): 2590-2600, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30354246

RESUMEN

Objective- Inhibition of HMGCR (3-hydroxy-3-methylglutaryl-coenzyme A reductase) is atheroprotective primarily by decreasing plasma LDL (low-density lipoprotein)-cholesterol. However, it is unknown whether inhibition of HMGCR in myeloid cells contributes to this atheroprotection. We sought to determine the role of myeloid HMGCR in the development of atherosclerosis. Approach and Results- We generated mice with genetically reduced Hmgcr in myeloid cells ( Hmgcr m-/m-) using LysM (Cre) and compared various functions of their macrophages to those of Hmgcr fl/fl control mice. We further compared the extent of atherosclerosis in Hmgcr m-/ m- and Hmgcr fl/fl mice in the absence of Ldlr (LDL receptor). Hmgcr m-/ m- macrophages and granulocytes had significantly lower Hmgcr mRNA expression and cholesterol biosynthesis than Hmgcr fl/fl cells. In vitro, Hmgcr m-/ m- monocytes/macrophages had reduced ability to migrate, proliferate, and survive compared with Hmgcr fl/fl monocytes/macrophages. However, there was no difference in ability to adhere, phagocytose, store lipids, or polarize to M1 macrophages between the 2 types of macrophages. The amounts of plasma membrane-associated small GTPase proteins, such as RhoA (RAS homolog family member A), were increased in Hmgcr m-/ m- macrophages. In the setting of Ldlr deficiency, Hmgcr m-/ m- mice developed significantly smaller atherosclerotic lesions than Hmgcr fl/fl mice. However, there were no differences between the 2 types of mice either in plasma lipoprotein profiles or in the numbers of proliferating or apoptotic cells in the lesions in vivo. The in vivo migration of Hmgcr m-/ m- macrophages to the lesions was reduced compared with Hmgcr fl/fl macrophages. Conclusions- Genetic reduction of HMGCR in myeloid cells may exert atheroprotective effects primarily by decreasing the migratory activity of monocytes/macrophages to the lesions.


Asunto(s)
Aorta/enzimología , Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Movimiento Celular , Hidroximetilglutaril-CoA Reductasas/metabolismo , Macrófagos Peritoneales/enzimología , Monocitos/enzimología , Traslado Adoptivo , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/prevención & control , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/prevención & control , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Hidroximetilglutaril-CoA Reductasas/genética , Lípidos/sangre , Macrófagos Peritoneales/patología , Macrófagos Peritoneales/trasplante , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Proteínas de Unión al GTP Monoméricas/metabolismo , Fenotipo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal
17.
Biomed Pharmacother ; 106: 192-199, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29958143

RESUMEN

Aucubin (Ai), a natural compound isolated from plants, including Aucuba japonica and Eucommia ulmoides, shows significant anti-inflammatory and anti-oxidative bioactivities. Here, we attempted to explore the protect effects of Ai on LPS-induced acute lung injury (ALI). Our results indicated that Ai increased the survival rate and ameliorated pathogenic processes in lipopolysaccharide (LPS)-induced mice. However, nuclear factor erythroid 2-related factor 2 (Nrf2) deletion may impede protective effect of Ai. Additionally, Ai reduced oxidative stress by down-regulating malondialdehyde (MDA) and O2· activity, and enhancing Nrf2-targeted signals, including heme oxygenase-1 (HO-1) and quinone oxidoreductase-1 (NQO-1). Also, Ai inhibited pro-inflammatory cytokines and phosphorylated-nuclear factor-κB (NF-κB) expression in LPS-administrated mice. However, these protective effects of Ai were suppressed in Nrf2-knockout mice. Importantly, Nrf2-deficiency showed no effects on phosphorylated AMP-activated protein kinase (p-AMPK) expression in mice treated with LPS and Ai. Similarly, in LPS-induced macrophages, Ai reduced reactive oxygen species (ROS) generation, elevated NQO-1 and HO-1 expression. LPS-stimulated pro-inflammatory cytokines and p-NF-κB were reversed by Ai. Of note, we found that Ai-induced Nrf2 activation was dependent on AMPK activation. Suppression of AMPK levels may inhibit Nrf2 activation, finally leading to up regulation of inflammatory response and oxidative stress. Thus, our findings indicated the crosstalk between Nrf2 and AMPK signaling pathways, and the interaction was essential for the anti-oxidant and anti-inflammatory effects of Ai in LPS-induced macrophages, which might be beneficial for finding new treatments against ALI.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Glucósidos Iridoides/farmacología , Lipopolisacáridos , Pulmón/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/genética , Animales , Citocinas/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Pulmón/enzimología , Pulmón/patología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/deficiencia , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Células THP-1 , Factores de Tiempo
18.
Atherosclerosis ; 275: 156-165, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29908485

RESUMEN

BACKGROUND AND AIMS: Atherosclerosis is a lipid-driven chronic inflammatory disorder of the arteries, and monocytes and macrophages play a central role in this process. Within the atherosclerotic lesion, macrophages can scavenge modified lipids and become the so-called foam cells. We previously reported that the epigenetic enzyme Kdm6b (also known as Jmjd3) controls the pro-fibrotic transcriptional profile of peritoneal foam cells. Given the importance of these cells in atherosclerosis, we now studied the effect of myeloid Kdm6b on disease progression. METHODS: Bone marrow of myeloid Kdm6b deficient (Kdm6bdel) mice or wild type littermates (Kdm6bwt) was transplanted to lethally irradiated Ldlr-/- mice fed a high fat diet for 9 weeks to induce atherosclerosis. RESULTS: Lesion size was similar in Kdm6bwt and Kdm6bdel transplanted mice. However, lesions of Kdm6bdel mice contained more collagen and were more necrotic. Pathway analysis on peritoneal foam cells showed that the pathway involved in leukocyte chemotaxis was most significantly upregulated. Although macrophage and neutrophil content was similar after 9 weeks of high fat diet feeding, the relative increase in collagen content and necrosis revealed that atherosclerotic lesions in Kdm6bdel mice progress faster. CONCLUSION: Myeloid Kdm6b deficiency results in more advanced atherosclerosis.


Asunto(s)
Aorta/enzimología , Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Células Espumosas/enzimología , Histona Demetilasas con Dominio de Jumonji/deficiencia , Macrófagos Peritoneales/enzimología , Placa Aterosclerótica , Animales , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Quimiotaxis de Leucocito , Colágeno/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Fibrosis , Células Espumosas/patología , Histona Demetilasas con Dominio de Jumonji/genética , Macrófagos Peritoneales/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Infiltración Neutrófila , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Tiempo
19.
Biomed Pharmacother ; 103: 1294-1301, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29864911

RESUMEN

Activity, mechanisms of action, and toxicity of natural compounds have been investigated in a context in which knowledge on which pathway is activated remains crucial to understand the action mechanism of these bioactive substances when treating an infected host. Herein, we showed an ability of copaiba oil and kaurenoic acid to eliminate Trypanosoma cruzi forms by infected macrophages through other mechanisms in addition to nitric oxide, reactive oxygen species, iron metabolism, and antioxidant defense. Both compounds induced an anti-inflammatory response with an increase in IL-10 and TGF-ß as well as a decrease in IL-12 production. Despite being able to modulate the immune response in host cells, the antimicrobial activity of copaiba oil and kaurenoic acid seems to be a direct action of the compounds on the parasites, causing their death.


Asunto(s)
Antiprotozoarios/farmacología , Diterpenos/farmacología , Fabaceae/química , Macrófagos Peritoneales/metabolismo , Aceites Volátiles/farmacología , Trypanosoma cruzi/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ferritinas/genética , Ferritinas/metabolismo , Células HeLa , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/parasitología , Masculino , Ratones Endogámicos BALB C , Modelos Biológicos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
20.
Atherosclerosis ; 276: 171-181, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29853191

RESUMEN

BACKGROUND AND AIMS: PDZK1 (Post-synaptic density protein/Drosophila disc-large protein/Zonula occludens protein containing 1) stabilizes the HDL receptor protein, SR-B1, in the liver, and mediates SR-B1 signaling outside of the liver. Complete knockout of pdzk1 increases atherosclerosis in apoE-deficient mice, but the effect of PDZK1 in leukocytes is not known. In this study, we tested the role of leukocyte PDZK1 in atherosclerosis development by using bone marrow transplantation to generate ldlr deficient mice lacking PDZK1 in leukocytes. METHODS: Ldlr-/- mice were transplanted with either pdzk1-/- or pdzk1+/+ bone marrow and fed a high-fat diet to induce atherosclerosis. RESULTS: Bone marrow specific pdzk1 knockout slightly increased atherosclerotic plaque sizes but strikingly increased sizes of necrotic cores and cellular apoptosis in within plaques. PDZK1 deficiency prevented HDL dependent protection of macrophages from apoptosis in vitro and sensitized peritoneal macrophages to apoptosis in situ. PDZK1 deficiency in macrophages also impaired their ability to engulf apoptotic cells, and attenuated the IL-4 dependent induction of mannose receptor in vitro and mannose receptor protein levels in macrophages in atherosclerotic plaques. CONCLUSIONS: PDZK1 is required for anti-atherogenic responses in macrophages including HDL dependent protection against apoptosis and macrophage mediated efferocytosis and limits the accumulation of apoptotic cells within atherosclerotic plaques protecting against necrotic core development.


Asunto(s)
Apoptosis , Aterosclerosis/prevención & control , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucocitos/enzimología , Macrófagos Peritoneales/enzimología , Placa Aterosclerótica , Receptores de LDL/metabolismo , Animales , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Trasplante de Médula Ósea , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Células Jurkat , Leucocitos/patología , Lipoproteínas HDL/metabolismo , Macrófagos Peritoneales/patología , Masculino , Proteínas de la Membrana , Ratones Endogámicos C57BL , Ratones Noqueados , Necrosis , Fenotipo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA