Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 323
Filtrar
1.
Br J Pharmacol ; 181(12): 1843-1856, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38378175

RESUMEN

BACKGROUND AND PURPOSE: Our previous studies have found that andrographolide (AGP) alleviates calcific aortic valve disease (CAVD), but the underlying mechanism is unclear. This study explores the molecular target and signal mechanisms of AGP in inhibiting CAVD. EXPERIMENTAL APPROACH: The anti-calcification effects of the aortic valve with AGP treatment were evaluated by alizarin red staining in vitro and ultrasound and histopathological assessment of a high-fat (HF)-fed ApoE-/- mouse valve calcification model. A correlation between the H3 histone lactylation (H3Kla) and calcification was detected. Molecular docking and surface plasmon resonance (SPR) experiments were further used to confirm p300 as a target for AGP. Overexpression (oe) and silencing (si) of p300 were used to verify the inhibitory effect of AGP targeting p300 on the H3Kla in vitro and ex vivo. KEY RESULTS: AGP significantly inhibited calcium deposition in valve interstitial cells (VICs) and ameliorated aortic valve calcification. The multi-omics analysis revealed the glycolysis pathway involved in CAVD, indicating that AGP interfered with lactate production by regulating lactate dehydrogenase A (LDHA). In addition, lactylation, a new post-translational modification, was shown to have a role in promoting aortic valve calcification. Furthermore, H3Kla and H3K9la site were shown to correlate with Runx2 expression inhibition by AGP treatment. Importantly, we found that p300 transferase was the molecular target of AGP in inhibiting H3Kla. CONCLUSIONS AND IMPLICATIONS: Our findings, for the first time, demonstrated that AGP alleviates calcification by interfering with H3Kla via p300, which might be a powerful drug to prevent CAVD.


Asunto(s)
Estenosis de la Válvula Aórtica , Válvula Aórtica , Calcinosis , Diterpenos , Histonas , Animales , Humanos , Masculino , Ratones , Válvula Aórtica/patología , Válvula Aórtica/metabolismo , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Calcinosis/metabolismo , Calcinosis/tratamiento farmacológico , Calcinosis/patología , Diterpenos/farmacología , Diterpenos/química , Proteína p300 Asociada a E1A/metabolismo , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Histonas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Factores de Transcripción p300-CBP/antagonistas & inhibidores
2.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35181609

RESUMEN

Aortic valve stenosis (AVS) patients experience pathogenic valve leaflet stiffening due to excessive extracellular matrix (ECM) remodeling. Numerous microenvironmental cues influence pathogenic expression of ECM remodeling genes in tissue-resident valvular myofibroblasts, and the regulation of complex myofibroblast signaling networks depends on patient-specific extracellular factors. Here, we combined a manually curated myofibroblast signaling network with a data-driven transcription factor network to predict patient-specific myofibroblast gene expression signatures and drug responses. Using transcriptomic data from myofibroblasts cultured with AVS patient sera, we produced a large-scale, logic-gated differential equation model in which 11 biochemical and biomechanical signals were transduced via a network of 334 signaling and transcription reactions to accurately predict the expression of 27 fibrosis-related genes. Correlations were found between personalized model-predicted gene expression and AVS patient echocardiography data, suggesting links between fibrosis-related signaling and patient-specific AVS severity. Further, global network perturbation analyses revealed signaling molecules with the most influence over network-wide activity, including endothelin 1 (ET1), interleukin 6 (IL6), and transforming growth factor ß (TGFß), along with downstream mediators c-Jun N-terminal kinase (JNK), signal transducer and activator of transcription (STAT), and reactive oxygen species (ROS). Lastly, we performed virtual drug screening to identify patient-specific drug responses, which were experimentally validated via fibrotic gene expression measurements in valvular interstitial cells cultured with AVS patient sera and treated with or without bosentan-a clinically approved ET1 receptor inhibitor. In sum, our work advances the ability of computational approaches to provide a mechanistic basis for clinical decisions including patient stratification and personalized drug screening.


Asunto(s)
Válvula Aórtica/metabolismo , Perfilación de la Expresión Génica/métodos , Medicina de Precisión/métodos , Actinas/metabolismo , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/fisiología , Estenosis de la Válvula Aórtica/metabolismo , Biomarcadores Farmacológicos , Calcinosis/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Cicatriz/metabolismo , Biología Computacional/métodos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Modelos Genéticos , Miofibroblastos/metabolismo , Miofibroblastos/fisiología , Suero/metabolismo , Transducción de Señal , Transcriptoma/genética
3.
J Cardiovasc Pharmacol ; 79(1): e103-e115, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654784

RESUMEN

ABSTRACT: Aortic valve replacement for severe stenosis is a standard procedure in cardiovascular medicine. However, the use of biological prostheses has limitations especially in young patients because of calcifying degeneration, resulting in implant failure. Pioglitazone, a peroxisome proliferator-activated receptor gamma (PPAR-gamma) agonist, was shown to decrease the degeneration of native aortic valves. In this study, we aim to examine the impact of pioglitazone on inflammation and calcification of aortic valve conduits (AoC) in a rat model. Cryopreserved AoC (n = 40) were infrarenally implanted into Wistar rats treated with pioglitazone (75 mg/kg chow; n = 20, PIO) or untreated (n = 20, controls). After 4 or 12 weeks, AoC were explanted and analyzed by histology, immunohistology, and polymerase chain reaction. Pioglitazone significantly decreased the expression of inflammatory markers and reduced the macrophage-mediated inflammation in PIO compared with controls after 4 (P = 0.03) and 12 weeks (P = 0.012). Chondrogenic transformation was significantly decreased in PIO after 12 weeks (P = 0.001). Calcification of the intima and media was significantly reduced after 12 weeks in PIO versus controls (intima: P = 0.008; media: P = 0.025). Moreover, echocardiography revealed significantly better functional outcome of the AoC in PIO after 12 weeks compared with control. Interestingly, significantly increased intima hyperplasia could be observed in PIO compared with controls after 12 weeks (P = 0.017). Systemic PPAR-gamma activation prevents inflammation as well as intima and media calcification in AoC and seems to inhibit functional impairment of the implanted aortic valve. To further elucidate the therapeutic role of PPAR-gamma regulation for graft durability, translational studies and long-term follow-up data should be striven for.


Asunto(s)
Insuficiencia de la Válvula Aórtica/cirugía , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/trasplante , Bioprótesis , Implantación de Prótesis de Válvulas Cardíacas/instrumentación , Prótesis Valvulares Cardíacas , PPAR gamma/agonistas , Pioglitazona/farmacología , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Insuficiencia de la Válvula Aórtica/metabolismo , Insuficiencia de la Válvula Aórtica/patología , Calcinosis/metabolismo , Calcinosis/patología , Calcinosis/prevención & control , Condrogénesis/efectos de los fármacos , Criopreservación , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Implantación de Prótesis de Válvulas Cardíacas/efectos adversos , Humanos , Mediadores de Inflamación/metabolismo , Osteogénesis/efectos de los fármacos , PPAR gamma/metabolismo , Ratas Sprague-Dawley , Ratas Wistar , Transducción de Señal
4.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638942

RESUMEN

Calcific aortic valve disease (CAVD) is an athero-inflammatory process. Growing evidence supports the inflammation-driven calcification model, mediated by cytokines such as interferons (IFNs) and tumor necrosis factor (TNF)-α. Our goal was investigating IFNs' effects in human aortic valve endothelial cells (VEC) and the potential differences between aortic (aVEC) and ventricular (vVEC) side cells. The endothelial phenotype was analyzed by Western blot, qPCR, ELISA, monocyte adhesion, and migration assays. In mixed VEC populations, IFNs promoted the activation of signal transducers and activators of transcription-1 and nuclear factor-κB, and the subsequent up-regulation of pro-inflammatory molecules. Side-specific VEC were activated with IFN-γ and TNF-α in an orbital shaker flow system. TNF-α, but not IFN-γ, induced hypoxia-inducible factor (HIF)-1α stabilization or endothelial nitric oxide synthase downregulation. Additionally, IFN-γ inhibited TNF-α-induced migration of aVEC. Also, IFN-γ triggered cytokine secretion and adhesion molecule expression in aVEC and vVEC. Finally, aVEC were more prone to cytokine-mediated monocyte adhesion under multiaxial flow conditions as compared with uniaxial flow. In conclusion, IFNs promote inflammation and reduce TNF-α-mediated migration in human VEC. Moreover, monocyte adhesion was higher in inflamed aVEC sheared under multiaxial flow, which may be relevant to understanding the initial stages of CAVD.


Asunto(s)
Válvula Aórtica/metabolismo , Células Endoteliales/metabolismo , Interferón-alfa/farmacología , Interferón gamma/farmacología , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/inmunología , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/inmunología , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/inmunología , Calcinosis/inmunología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Trasplante de Corazón , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Monocitos/metabolismo , FN-kappa B/metabolismo , Fenotipo , Factor de Transcripción STAT1/metabolismo , Células THP-1 , Receptores de Trasplantes , Factor de Necrosis Tumoral alfa/farmacología
5.
Ann Biomed Eng ; 49(12): 3550-3562, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34704164

RESUMEN

The renin-angiotensin system (RAS) is activated in aortic valve disease, yet little is understood about how it affects the acute functional response of valve interstitial cells (VICs). Herein, we developed a gelatin-based valve thin film (vTF) platform to investigate whether the contractile response of VICs can be regulated via RAS mediators and inhibitors. First, the impact of culture medium (quiescent, activated, and osteogenic medium) on VIC phenotype and function was assessed. Contractility of VICs was measured upon treatment with angiotensin I (Ang I), angiotensin II (Ang II), angiotensin-converting enzyme (ACE) inhibitor, and Angiotensin II type 1 receptor (AT1R) inhibitor. Anisotropic cell alignment on gelatin vTF was achieved independent of culture conditions. Cells cultured in activated and osteogenic conditions were found to be more elongated than in quiescent medium. Increased α-SMA expression was observed in activated medium and no RUNX2 expression were observed in cells. VIC contractile stress increased with increasing concentrations (from 10-10 to 10-6 M) of Ang I and Ang II. Moreover, cell contraction was significantly reduced in all ACE and AT1R inhibitor-treated groups. Together, these findings suggest that local RAS is active in VICs, and our vTF may provide a powerful platform for valve drug screening and development.


Asunto(s)
Válvula Aórtica/citología , Sistema Renina-Angiotensina/fisiología , Angiotensina I/farmacología , Angiotensina I/fisiología , Angiotensina II/farmacología , Angiotensina II/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Válvula Aórtica/efectos de los fármacos , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Losartán/farmacología , Miofibroblastos/fisiología , Peptidil-Dipeptidasa A/fisiología , Receptor de Angiotensina Tipo 1/fisiología , Porcinos , Tetrahidroisoquinolinas/farmacología
6.
Biomed Pharmacother ; 139: 111674, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34243610

RESUMEN

OBJECTIVES: In calcific aortic valve disease (CAVD), the valve interstitial cells (VIC) osteogenic phenotype changes can lead to thickening and calcification of the valve leaflets,eventually lead to restricted valve movement and life-threatening. This study aims to investigate the effect and mechanism of dihydrotanshinone I (DHI) on osteogenic medium (OM) induced osteogenic phenotypic transition of porcine valve interstitial cells (PVICs), which can provide theoretical and scientific basis for clinical intervention in CAVD. METHODS AND RESULTS: Immunohistochemical methods were used to detect the expression of osteogenic indicators Runx2, OPN and inflammation indicators IL-1ß and p-NF-κB in valve specimens of CAVD patients(N = 3) and normal controls(N = 1). PVICs stimulated by osteoblastic medium (OM) were treated with or without DHI. CCK8, ALP and Alizarin Red S staining were used to detect cell growth and calcification, respectively. The results showed that under the treated with DHI, compared with OM, the formation of calcium nodules was reduced, and the expression of calcification-related markers Runx2 and OPN were down-regulated, which quantified by qRT-PCR and western blot. In addition, on the basis of OM induction, DHI also inhibited the phosphorylation of the NF-κB/ERK1/2 and SMAD1/5/8 signaling pathway. CONCLUSION: DHI (10 µM) treatment can reverse the osteogenic phenotypic transition of PVICs induced by osteogenic medium, and the mechanism may be related to NF-κB、ERK 1/2 and Smad1/5/8 pathways.


Asunto(s)
Estenosis de la Válvula Aórtica/tratamiento farmacológico , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/patología , Calcinosis/tratamiento farmacológico , Furanos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , FN-kappa B/metabolismo , Fenantrenos/farmacología , Quinonas/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Animales , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Porcinos
7.
J Cardiovasc Pharmacol ; 78(3): 411-421, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34132687

RESUMEN

ABSTRACT: Calcific aortic valve disease is a common heart disease that contributes to increased cardiovascular morbidity and mortality. There is a lack of effective pharmaceutical therapy because its mechanisms are not yet fully known. Ginkgo biloba extract (EGB761) is reported to alleviate vascular calcification. However, whether EGB761 protects against aortic valve calcification, a disease whose pathogenesis shares many similarities with vascular calcification, and potential molecular mechanisms remain unknown. In this study, porcine aortic valve interstitial cell (pAVIC) calcification was induced by warfarin with or without the presence of EGB761. Immunostaining was performed to establish and characterize the pAVIC phenotype. Calcium deposition and calcium content were examined by Alizarin Red S staining and an intracellular calcium content assay. Alkaline phosphatase activity was detected by the p-nitrophenyl phosphate method. The expression levels of bone morphogenetic protein-2 (BMP2), Runt-related transcription factor 2 (Runx2), homeobox protein MSX-2, and phosphorylated (p)-Smad1/5 were detected by reverse transcription-quantitative polymerase chain reaction (PCR) and Western blot analysis. Consistent with these in vitro data, we also confirmed the suppression of in vivo calcification by EGB761 in the warfarin-induced C57/Bl6 mice. The results indicated that both pAVICs and aortic valves tissue of mice stimulated with warfarin showed increased calcium deposition and expression of osteogenic markers (alkaline phosphatase, BMP2, homeobox protein MSX-2, and Runx2) and promoted p-Smad1/5 translocation from the cytoplasm to the nucleus. The addition of EGB761 significantly inhibited p-Smad1/5 translocation from the cytoplasm to the nucleus, thus suppressing calcification. In conclusion, EGB761 could ameliorate warfarin-induced aortic valve calcification through the inhibition of the BMP2-medicated Smad1/5/Runx2 signaling pathway.


Asunto(s)
Válvula Aórtica/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Calcinosis/prevención & control , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Enfermedades de las Válvulas Cardíacas/prevención & control , Extractos Vegetales/farmacología , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Transporte Activo de Núcleo Celular , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Calcinosis/inducido químicamente , Calcinosis/metabolismo , Calcinosis/patología , Calcio/metabolismo , Células Cultivadas , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Ginkgo biloba , Enfermedades de las Válvulas Cardíacas/inducido químicamente , Enfermedades de las Válvulas Cardíacas/metabolismo , Enfermedades de las Válvulas Cardíacas/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Fosforilación , Transducción de Señal , Sus scrofa , Warfarina
8.
FEBS J ; 288(22): 6528-6542, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34009721

RESUMEN

Calcific aortic valve disease (CAVD) is the most prevalent valvulopathy worldwide. Growing evidence supports a role for viral and cell-derived double-stranded (ds)-RNA in cardiovascular pathophysiology. Poly(I:C), a dsRNA surrogate, has been shown to induce inflammation, type I interferon (IFN) responses, and osteogenesis through Toll-like receptor 3 in aortic valve interstitial cells (VIC). Here, we aimed to determine whether IFN signaling via Janus kinase (JAK)/Signal transducers and activators of transcription (STAT) mediates dsRNA-induced responses in primary human VIC. Western blot, ELISA, qPCR, calcification, flow cytometry, and enzymatic assays were performed to evaluate the mechanisms of dsRNA-induced inflammation and calcification. Poly(I:C) triggered a type I IFN response characterized by IFN-regulatory factors gene upregulation, IFN-ß secretion, and STAT1 activation. Additionally, Poly(I:C) promoted VIC inflammation via NF-κB and subsequent adhesion molecule expression, and cytokine secretion. Pretreatment with ruxolitinib, a clinically used JAK inhibitor, abrogated these responses. Moreover, Poly(I:C) promoted a pro-osteogenic phenotype and increased VIC calcification to a higher extent in cells from males. Inhibition of JAK with ruxolitinib or a type I IFN receptor blocking antibody blunted Poly(I:C)-induced calcification. Mechanistically, Poly(I:C) promoted VIC apoptosis in calcification medium, which was inhibited by ruxolitinib. Moreover, Poly(I:C) co-operated with IFN-γ to increase VIC calcification by synergistically activating extracellular signal-regulated kinases and hypoxia-inducible factor-1α pathways. In conclusion, JAK/STAT signaling mediates dsRNA-triggered inflammation, apoptosis, and calcification and may contribute to a positive autocrine loop in human VIC in the presence of IFN-γ. Blockade of dsRNA responses with JAK inhibitors may be a promising therapeutic avenue for CAVD.


Asunto(s)
Estenosis de la Válvula Aórtica/tratamiento farmacológico , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/patología , Calcinosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inhibidores de las Cinasas Janus/farmacología , Nitrilos/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Bicatenario/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Calcinosis/metabolismo , Calcinosis/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inhibidores de las Cinasas Janus/química , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Masculino , Persona de Mediana Edad , Nitrilos/química , Pirazoles/química , Pirimidinas/química , ARN Bicatenario/metabolismo , Adulto Joven
9.
Cells ; 10(1)2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33401457

RESUMEN

Calcific aortic valve disease (CAVD) accompanies inflammatory cell infiltration, fibrosis, and ultimately calcification of the valve leaflets. We previously demonstrated that dipeptidyl peptidase-4 (DPP-4) is responsible for the progression of aortic valvular calcification in CAVD animal models. As evogliptin, one of the DPP-4 inhibitors displays high specific accumulation in cardiac tissue, we here evaluated its therapeutic potency for attenuating valvular calcification in CAVD animal models. Evogliptin administration markedly reduced calcific deposition accompanied by a reduction in proinflammatory cytokine expression in endothelial nitric oxide synthase-deficient mice in vivo, and significantly ameliorated the mineralization of the primary human valvular interstitial cells (VICs), with a reduction in the mRNA expression of bone-associated and fibrosis-related genes in vitro. In addition, evogliptin ameliorated the rate of change in the transaortic peak velocity and mean pressure gradients in our rabbit model as assessed by echocardiography. Importantly, evogliptin administration in a rabbit model was found to suppress the effects of a high-cholesterol diet and of vitamin D2-driven fibrosis in association with a reduction in macrophage infiltration and calcific deposition in aortic valves. These results have indicated that evogliptin prohibits inflammatory cytokine expression, fibrosis, and calcification in a CAVD animal model, suggesting its potential as a selective therapeutic agent for the inhibition of valvular calcification during CAVD progression.


Asunto(s)
Estenosis de la Válvula Aórtica/tratamiento farmacológico , Válvula Aórtica/patología , Calcinosis/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Piperazinas/uso terapéutico , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/complicaciones , Estenosis de la Válvula Aórtica/genética , Calcinosis/complicaciones , Calcinosis/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/complicaciones , Inflamación/genética , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Piperazinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos
10.
J Thorac Cardiovasc Surg ; 161(4): e261-e271, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-31924353

RESUMEN

BACKGROUND: Aortic valve interstitial cells have been implicated in the pathogenesis of aortic stenosis. In response to proinflammatory stimuli, aortic valve interstitial cells undergo an osteogenic phenotypic change. The purpose of this study was to determine whether the anti-inflammatory effects of statins prevent osteogenic activity in cultured aortic valve interstitial cells. METHODS: Human aortic valve interstitial cells were isolated from hearts explanted for cardiac transplantation. To test whether simvastatin down-regulates TLR4-induced osteogenic response, aortic valve interstitial cells were treated with simvastatin with and without TLR4 agonist lipopolysaccharide (LPS), and osteogenic markers were measured. Simvastatin's influence on in vitro calcium deposition was assessed by alizarin red staining. Knockdown of postreceptor signaling proteins (MyD88 and TRIF) was performed to determine which of 2 TLR4-associated pathways mediates the osteogenic response. Expression levels of TLR4-induced nuclear factor kappa light chain enhancer of activated B cells (NF-κB) and TLR4 expression were assessed after treatment with simvastatin. Statistical testing was done by analysis of variance (P < .05). RESULTS: Simvastatin decreased LPS-induced ALP and Runx2 expression and inhibited in vitro calcium deposition in aortic valve interstitial cells. Knockdown of MyD88 and TRIF attenuated the osteogenic response. Simvastatin attenuated TLR4-dependent NF-κB signaling and down-regulated TLR4 levels. CONCLUSIONS: Simvastatin prevented TLR4-induced osteogenic phenotypic changes in isolated aortic valve interstitial cells via down-regulation of TLR4 and inhibition of NF-κB signaling. These data offer mechanistic insight into a possible therapeutic role for simvastatin in the prevention of aortic stenosis.


Asunto(s)
Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Osteogénesis/efectos de los fármacos , Simvastatina/farmacología , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Fosfatasa Alcalina/metabolismo , Válvula Aórtica/metabolismo , Técnicas de Cultivo de Célula , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Humanos , Lipopolisacáridos/fisiología , Factor 88 de Diferenciación Mieloide/fisiología , FN-kappa B/metabolismo , Transducción de Señal , Receptor Toll-Like 4/fisiología
11.
J Mol Cell Cardiol ; 150: 54-64, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33045251

RESUMEN

AIMS: Calcific aortic valve disease (CAVD) is a primary cause of cardiovascular mortality; however, its mechanisms are unknown. Currently, no effective pharmacotherapy is available for CAVD. Aldo-keto reductase family 1 member B (Akr1B1) has been identified as a potential therapeutic target for valve interstitial cell calcification. Herein, we hypothesized that inhibition of Akr1B1 can attenuate aortic valve calcification. METHODS AND RESULTS: Normal and degenerative tricuspid calcific valves from human samples were analyzed by immunoblotting and immunohistochemistry. The results showed significant upregulation of Akr1B1 in CAVD leaflets. Akr1B1 inhibition attenuated calcification of aortic valve interstitial cells in osteogenic medium. In contrast, overexpression of Akr1B1 aggravated calcification in osteogenic medium. Mechanistically, using RNA sequencing (RNAseq), we revealed that Hippo-YAP signaling functions downstream of Akr1B1. Furthermore, we established that the protein level of the Hippo-YAP signaling effector active-YAP had a positive correlation with Akr1B1. Suppression of YAP reversed Akr1B1 overexpression-induced Runx2 upregulation. Moreover, YAP activated the Runx2 promoter through TEAD1 in a manner mediated by ChIP and luciferase reporter systems. Animal experiments showed that the Akr1B1 inhibitor epalrestat attenuated aortic valve calcification induced by a Western diet in LDLR-/- mice. CONCLUSION: This study demonstrates that inhibition of Akr1B1 can attenuate the degree of calcification both in vitro and in vivo. The Akr1B1 inhibitor epalrestat may be a potential treatment option for CAVD.


Asunto(s)
Aldehído Reductasa/metabolismo , Aldo-Ceto Reductasas/metabolismo , Estenosis de la Válvula Aórtica/enzimología , Estenosis de la Válvula Aórtica/patología , Válvula Aórtica/enzimología , Válvula Aórtica/patología , Calcinosis/enzimología , Calcinosis/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aldehído Reductasa/antagonistas & inhibidores , Animales , Válvula Aórtica/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Inhibidores Enzimáticos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Lentivirus/metabolismo , Ratones , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
12.
Arterioscler Thromb Vasc Biol ; 41(1): 117-127, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33115271

RESUMEN

Cardiovascular calcification is an insidious form of ectopic tissue mineralization that presents as a frequent comorbidity of atherosclerosis, aortic valve stenosis, diabetes, renal failure, and chronic inflammation. Calcification of the vasculature and heart valves contributes to mortality in these diseases. An inability to clinically image or detect early microcalcification coupled with an utter lack of pharmaceutical therapies capable of inhibiting or regressing entrenched and detectable macrocalcification has led to a prominent and deadly gap in care for a growing portion of our rapidly aging population. Recognition of this mounting concern has arisen over the past decade and led to a series of revolutionary works that has begun to pull back the curtain on the pathogenesis, mechanistic basis, and causative drivers of cardiovascular calcification. Central to this progress is the discovery that calcifying extracellular vesicles act as active precursors of cardiovascular microcalcification in diverse vascular beds. More recently, the omics revolution has resulted in the collection and quantification of vast amounts of molecular-level data. As the field has become poised to leverage these resources for drug discovery, new means of deriving relevant biological insights from these rich and complex datasets have come into focus through the careful application of systems biology and network medicine approaches. As we look onward toward the next decade, we envision a growing need to standardize approaches to study this complex and multifaceted clinical problem and expect that a push to translate mechanistic findings into therapeutics will begin to finally provide relief for those impacted by this disease.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Arterias/metabolismo , Calcinosis/metabolismo , Vesículas Extracelulares/metabolismo , Osteogénesis , Calcificación Vascular/metabolismo , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/patología , Arterias/efectos de los fármacos , Arterias/patología , Calcinosis/tratamiento farmacológico , Calcinosis/genética , Calcinosis/patología , Desarrollo de Medicamentos , Descubrimiento de Drogas , Vesículas Extracelulares/patología , Regulación de la Expresión Génica , Genómica , Humanos , Osteogénesis/efectos de los fármacos , Transducción de Señal , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/genética , Calcificación Vascular/patología
13.
Cardiovasc Res ; 117(3): 820-835, 2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-32259211

RESUMEN

AIMS: Calcific aortic valve disease (CAVD) is the most common heart valve disease in the Western world. It has been reported that zinc is accumulated in calcified human aortic valves. However, whether zinc directly regulates CAVD is yet to be elucidated. The present study sought to determine the potential role of zinc in the pathogenesis of CAVD. METHODS AND RESULTS: Using a combination of a human valve interstitial cell (hVIC) calcification model, human aortic valve tissues, and blood samples, we report that 20 µM zinc supplementation attenuates hVIC in vitro calcification, and that this is mediated through inhibition of apoptosis and osteogenic differentiation via the zinc-sensing receptor GPR39-dependent ERK1/2 signalling pathway. Furthermore, we report that GPR39 protein expression is dramatically reduced in calcified human aortic valves, and there is a significant reduction in zinc serum levels in patients with CAVD. Moreover, we reveal that 20 µM zinc treatment prevents the reduction of GPR39 observed in calcified hVICs. We also show that the zinc transporter ZIP13 and ZIP14 are significantly increased in hVICs in response to zinc treatment. Knockdown of ZIP13 or ZIP14 significantly inhibited hVIC in vitro calcification and osteogenic differentiation. CONCLUSIONS: Together, these findings suggest that zinc is a novel inhibitor of CAVD, and report that zinc transporter ZIP13 and ZIP14 are important regulators of hVIC in vitro calcification and osteogenic differentiation. Zinc supplementation may offer a potential therapeutic strategy for CAVD.


Asunto(s)
Válvula Aórtica/efectos de los fármacos , Calcinosis/tratamiento farmacológico , Enfermedades de las Válvulas Cardíacas/tratamiento farmacológico , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sulfato de Zinc/farmacología , Válvula Aórtica/enzimología , Válvula Aórtica/patología , Apoptosis/efectos de los fármacos , Calcinosis/enzimología , Calcinosis/patología , Estudios de Casos y Controles , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Femenino , Enfermedades de las Válvulas Cardíacas/enzimología , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Osteogénesis/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Sulfato de Zinc/metabolismo
14.
Trends Cardiovasc Med ; 31(5): 305-311, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32525013

RESUMEN

Elevated levels of lipoprotein(a) [Lp(a)] have been identified as an independent and causal risk factor for atherosclerotic cardiovascular disease (ASCVD) and, more recently, calcific aortic valve disease (CAVD). CAVD is a slow, progressive disorder presenting as severe trileaflet calcification known as aortic valve stenosis (AS) that impairs valve motion and restricts ventricular outflow. AS afflicts 2% of the aging population (≥ 65 years) and tends to be quite advanced by the time it presents clinical symptoms of exertional angina, syncope, or heart failure. Currently, the only effective clinical therapy for AS patients is surgical or transcatheter aortic valve replacement. Evidence is accumulating that Lp(a) can exacerbate pathophysiological processes in CAVD, specifically, endothelial dysfunction, formation of foam cells, and promotion of a pro-inflammatory state. In the valve milieu, the pro-inflammatory effects of Lp(a) are manifested in valve thickening and mineralization through pro-osteogenic signaling and changes in gene expression in valve interstitial cells that is primarily facilitated by the oxidized phospholipid content of Lp(a). In AS pathogenesis, an incomplete understanding of the role of Lp(a) at the molecular level and the absence of appropriate animal models are barriers for the development of specific and effective clinical interventions designed to mitigate the role of Lp(a) in AS. However, the advent of effective therapies that dramatically lower Lp(a) provides the possibility of the first medical treatment to halt AS progression.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Calcinosis/metabolismo , Lipoproteína(a)/metabolismo , Anciano , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/diagnóstico , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/genética , Calcinosis/diagnóstico , Calcinosis/tratamiento farmacológico , Calcinosis/genética , Modelos Animales de Enfermedad , Femenino , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Hipolipemiantes/uso terapéutico , Lipoproteína(a)/genética , Masculino , Oligonucleótidos/uso terapéutico , Oxidación-Reducción , Fosfolípidos/metabolismo
15.
Science ; 371(6530)2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33303684

RESUMEN

Mapping the gene-regulatory networks dysregulated in human disease would allow the design of network-correcting therapies that treat the core disease mechanism. However, small molecules are traditionally screened for their effects on one to several outputs at most, biasing discovery and limiting the likelihood of true disease-modifying drug candidates. Here, we developed a machine-learning approach to identify small molecules that broadly correct gene networks dysregulated in a human induced pluripotent stem cell (iPSC) disease model of a common form of heart disease involving the aortic valve (AV). Gene network correction by the most efficacious therapeutic candidate, XCT790, generalized to patient-derived primary AV cells and was sufficient to prevent and treat AV disease in vivo in a mouse model. This strategy, made feasible by human iPSC technology, network analysis, and machine learning, may represent an effective path for drug discovery.


Asunto(s)
Enfermedad de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Válvula Aórtica/patología , Calcinosis/tratamiento farmacológico , Redes Reguladoras de Genes/efectos de los fármacos , Aprendizaje Automático , Nitrilos/farmacología , Nitrilos/uso terapéutico , Tiazoles/farmacología , Tiazoles/uso terapéutico , Algoritmos , Animales , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/metabolismo , Válvula Aórtica/fisiopatología , Enfermedad de la Válvula Aórtica/genética , Enfermedad de la Válvula Aórtica/fisiopatología , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/fisiopatología , Calcinosis/genética , Calcinosis/fisiopatología , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Haploinsuficiencia , Humanos , Células Madre Pluripotentes Inducidas , Ratones Endogámicos C57BL , RNA-Seq , Receptor Notch1/genética , Bibliotecas de Moléculas Pequeñas
16.
Arterioscler Thromb Vasc Biol ; 41(1): 11-19, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33232199

RESUMEN

Mineralization of cardiovascular structures including blood vessels and heart valves is a common feature. We postulate that ectopic mineralization is a response-to-injury in which signals delivered to cells trigger a chain of events to restore and repair tissues. Maladaptive response to external or internal signals promote the expression of danger-associated molecular patterns, which, in turn, promote, when expressed chronically, a procalcifying gene program. Growing evidence suggest that danger-associated molecular patterns such as oxyphospholipids and small lipid mediators, generated by enzyme activity, are involved in the transition of vascular smooth muscle cells and valve interstitial cells to an osteoblast-like phenotype. Understanding the regulation and the molecular processes underpinning the mineralization of atherosclerotic plaques and cardiac valves are providing valuable mechanistic insights, which could lead to the development of novel therapies. Herein, we provide a focus account on the role oxyphospholipids and their mediators in the development of mineralization in plaques and calcific aortic valve disease.


Asunto(s)
Estenosis de la Válvula Aórtica/metabolismo , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Arterias/metabolismo , Calcinosis/metabolismo , Fosfolípidos/metabolismo , Calcificación Vascular/metabolismo , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/tratamiento farmacológico , Estenosis de la Válvula Aórtica/patología , Arterias/efectos de los fármacos , Arterias/patología , Calcinosis/tratamiento farmacológico , Calcinosis/patología , Plasticidad de la Célula , Humanos , Oxidación-Reducción , Placa Aterosclerótica , Transducción de Señal , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/patología
17.
Int J Mol Sci ; 21(23)2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255450

RESUMEN

Calcific aortic valve disease (CAVD) is the result of maladaptive fibrocalcific processes leading to a progressive thickening and stiffening of aortic valve (AV) leaflets. CAVD is the most common cause of aortic stenosis (AS). At present, there is no effective pharmacotherapy in reducing CAVD progression; when CAVD becomes symptomatic it can only be treated with valve replacement. Inflammation has a key role in AV pathological remodeling; hence, anti-inflammatory therapy has been proposed as a strategy to prevent CAVD. Cyclooxygenase 2 (COX-2) is a key mediator of the inflammation and it is the target of widely used anti-inflammatory drugs. COX-2-inhibitor celecoxib was initially shown to reduce AV calcification in a murine model. However, in contrast to these findings, a recent retrospective clinical analysis found an association between AS and celecoxib use. In the present study, we investigated whether variations in COX-2 expression levels in human AVs may be linked to CAVD. We extracted total RNA from surgically explanted AVs from patients without CAVD or with CAVD. We found that COX-2 mRNA was higher in non-calcific AVs compared to calcific AVs (0.013 ± 0.002 vs. 0.006 ± 0.0004; p < 0.0001). Moreover, we isolated human aortic valve interstitial cells (AVICs) from AVs and found that COX-2 expression is decreased in AVICs from calcific valves compared to AVICs from non-calcific AVs. Furthermore, we observed that COX-2 inhibition with celecoxib induces AVICs trans-differentiation towards a myofibroblast phenotype, and increases the levels of TGF-ß-induced apoptosis, both processes able to promote the formation of calcific nodules. We conclude that reduced COX-2 expression is a characteristic of human AVICs prone to calcification and that COX-2 inhibition may promote aortic valve calcification. Our findings support the notion that celecoxib may facilitate CAVD progression.


Asunto(s)
Estenosis de la Válvula Aórtica/tratamiento farmacológico , Válvula Aórtica/patología , Calcinosis/tratamiento farmacológico , Ciclooxigenasa 2/genética , Inflamación/tratamiento farmacológico , Factor de Crecimiento Transformador beta/genética , Anciano , Anciano de 80 o más Años , Animales , Válvula Aórtica/efectos de los fármacos , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/patología , Apoptosis/efectos de los fármacos , Calcinosis/genética , Calcinosis/patología , Celecoxib/administración & dosificación , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Masculino , Ratones , Persona de Mediana Edad , ARN Mensajero/genética
18.
Oxid Med Cell Longev ; 2020: 2043762, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33149806

RESUMEN

Warfarin, a vitamin K antagonist (VKA), is known to promote arterial calcification (AC). In the present study, we conducted a case-cohort study within the Multi-Ethnic Study of Atherosclerosis (MESA); 6655 participants were included. From MESA data, we found that AC was related to both age and vitamin K; furthermore, the score of AC increased with SASP marker including interlukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) rising. Next, a total of 79 warfarin users in our center developed significantly more calcified coronary plaques as compared to non-VKA users. We investigated the role of warfarin in phosphate-induced AC in different ages by in vitro experimental study. Furthermore, dose-time-response of warfarin was positively correlated with AC score distribution and plasma levels of the SASP maker IL-6 among patients < 65 years, but not among patients ≥ 65 years. In addition, in vitro research suggested that warfarin treatment tended to deteriorate calcification in young VSMC at the early stage of calcification. Our results suggested that aging and warfarin-treatment were independently related to increased AC. Younger patients were more sensitive to warfarin-related AC than older patients, which was possibly due to accumulated warfarin-induced cellular senescence.


Asunto(s)
Válvula Aórtica/patología , Biomarcadores/metabolismo , Senescencia Celular/efectos de los fármacos , Calcificación Vascular/patología , Warfarina/farmacología , Abdomen/patología , Anciano , Animales , Válvula Aórtica/diagnóstico por imagen , Válvula Aórtica/efectos de los fármacos , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/patología , Relación Dosis-Respuesta a Droga , Electrocardiografía , Análisis Factorial , Femenino , Humanos , Interleucina-6/metabolismo , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Fosfatos , Ratas Sprague-Dawley , Factores de Riesgo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Calcificación Vascular/diagnóstico por imagen , Vitamina K/farmacología
19.
J Cell Mol Med ; 24(21): 12476-12490, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32954678

RESUMEN

Vascular calcification is an important risk factor for the mortality and morbidity in chronic kidney disease (CKD). Unfortunately, until now there is no certain medication targeting vascular calcification in CKD. In this study, we explored the inhibitory effect of celastrol on high calcium-induced vascular calcification and the underlying molecular mechanisms. Cell proliferation assay showed that celastrol inhibited aortic valve interstitial cell (VIC) and vascular smooth muscle cell (VSMC) proliferation when its concentration was higher than 0.6 µmol/L. 0.8 µmol/L celastrol inhibited the expression of osteogenic genes and calcium deposition induced by high-calcium medium in both AVICs and VSMCs. In mouse vascular calcification model induced by adenine combined with vitamin D, alizarin red and immunostaining showed that celastrol inhibited pro-calcification gene expression and calcium deposition in aortic wall and aortic valve tissues. At the molecular level, celastrol inhibited the increase of BMP2, phosphorylated Smad1/5 (p-Smad1/5) and non-phosphorylated ß-catenin (n-p-ß-catenin) induced by high-calcium medium both in vitro and in vivo. Also, BMP2 overexpression reversed the anti-calcification effects of celastrol by recovering the decrease of p-Smad1/5 and n-p-ß-catenin. Furthermore, celastrol prevented the up-regulation of BMPRII and down-regulation of Smad6 induced by high calcium, and this protectory effect can be abolished by BMP2 overexpression. In conclusion, our data for the first time demonstrate that celastrol attenuates high calcium-induced arterial and valvular calcification by inhibiting BMP2/Smad1/5 signalling, which may provide a novel therapeutic strategy for arterial and valvular calcification in patients with CKD.


Asunto(s)
Válvula Aórtica/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Triterpenos Pentacíclicos/farmacología , Transducción de Señal/efectos de los fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Calcificación Vascular/metabolismo , Animales , Aorta/metabolismo , Válvula Aórtica/fisiopatología , Calcio/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Porcinos , Vitamina D/metabolismo , beta Catenina/metabolismo
20.
Biol Pharm Bull ; 43(6): 1016-1019, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32475911

RESUMEN

Endurance exercise training has been shown to induce peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) expression in skeletal muscle. We recently reported that skeletal muscle-specific PGC-1α overexpression suppressed atherosclerosis in apolipoprotein E-knockout (ApoE-/-) mice. ß-Aminoisobutyric acid (BAIBA) is a PGC-1α-dependent myokine secreted from myocytes that affects multiple organs. We have also reported that BAIBA suppresses tumor necrosis factor-alpha-induced vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1) gene expression in endothelial cells. In the present study, we hypothesized that BAIBA suppresses atherosclerosis progression, and tested that hypothesis with ApoE-/- mice. The mice were administered water containing BAIBA for 14 weeks, and were then sacrificed at 20 weeks of age. Atherosclerotic plaque area, plasma BAIBA concentration, and plasma lipoprotein profiles were assessed. Immunohistochemical analyses of the plaque were performed to assess VCAM-1 and MCP-1 protein expression levels and macrophage infiltration. The results showed that BAIBA administration decreased atherosclerosis plaque area by 30%, concomitant with the elevation of plasma BAIBA levels. On the other hand, plasma lipoprotein profiles were not changed by the administration. Immunohistochemical analyses indicated reductions in VCAM-1, MCP-1, and Mac-2 protein expression levels in the plaque. These results suggest that BAIBA administration suppresses atherosclerosis progression without changing plasma lipoprotein profiles. We propose that the mechanisms of this suppression are reductions in both VCAM-1 and MCP-1 expression as well as macrophage infiltration into the plaque.


Asunto(s)
Ácidos Aminoisobutíricos/uso terapéutico , Aterosclerosis/tratamiento farmacológico , Ácidos Aminoisobutíricos/sangre , Ácidos Aminoisobutíricos/farmacocinética , Ácidos Aminoisobutíricos/farmacología , Animales , Válvula Aórtica/efectos de los fármacos , Válvula Aórtica/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Quimiocina CCL2/metabolismo , Galectina 3/metabolismo , Lípidos/sangre , Ratones Noqueados para ApoE , Molécula 1 de Adhesión Celular Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...