Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
1.
Acta Pharmacol Sin ; 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886550

RÉSUMÉ

Urolithin A (UroA), a dietary phytochemical, is produced by gut bacteria from fruits rich in natural polyphenols ellagitannins (ETs). The efficiency of ETs metabolism to UroA in humans depends on gut microbiota. UroA has shown a variety of pharmacological activities. In this study we investigated the effects of UroA on atherosclerotic lesion development and stability. Apolipoprotein E-deficient (ApoE-/-) mice were fed a high-fat and high-cholesterol diet for 3 months to establish atherosclerosis model. Meanwhile the mice were administered UroA (50 mg·kg-1·d-1, i.g.). We showed that UroA administration significantly decreased diet-induced atherosclerotic lesions in brachiocephalic arteries, macrophage content in plaques, expression of endothelial adhesion molecules, intraplaque hemorrhage and size of necrotic core, while increased the expression of smooth muscle actin and the thickness of fibrous cap, implying features of plaque stabilization. The underlying mechanisms were elucidated using TNF-α-stimulated human endothelial cells. Pretreatment with UroA (10, 25, 50 µM) dose-dependently inhibited TNF-α-induced endothelial cell activation and monocyte adhesion. However, the anti-inflammatory effects of UroA in TNF-α-stimulated human umbilical vein endothelial cells (HUVECs) were independent of NF-κB p65 pathway. We conducted RNA-sequencing profiling analysis to identify the differential expression of genes (DEGs) associated with vascular function, inflammatory responses, cell adhesion and thrombosis in UroA-pretreated HUVECs. Human disease enrichment analysis revealed that the DEGs were significantly correlated with cardiovascular diseases. We demonstrated that UroA pretreatment mitigated endothelial inflammation by promoting NO production and decreasing YAP/TAZ protein expression and TEAD transcriptional activity in TNF-α-stimulated HUVECs. On the other hand, we found that UroA administration modulated the transcription and cleavage of lipogenic transcription factors SREBP1/2 in the liver to ameliorate cholesterol metabolism in ApoE-/- mice. This study provides an experimental basis for new dietary therapeutic option to prevent atherosclerosis.

2.
J Physiol Investig ; 67(1): 47-56, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38780272

RÉSUMÉ

Sepsis-induced kidney injury (SAKI) has been frequently established as a prevailing complication of sepsis which is linked to unfavorable outcomes. Fatty acid-binding protein-4 (FABP4) has been proposed as a possible target for the treatment of SAKI. In the current work, we aimed to explore the role and underlying mechanism of FABP4 in lipopolysaccharide (LPS)-induced human renal tubular epithelial cell damage. In LPS-induced human kidney 2 (HK2) cells, FABP4 expression was tested by the reverse transcription-quantitative polymerase chain reaction and Western blot. Cell counting kit-8 method assayed cell viability. Inflammatory levels were detected using the enzyme-linked immunosorbent assay. Immunofluorescence staining measured the nuclear translocation of nuclear factor kappa B p65. Thiobarbituric acid-reactive substances assay and C11 BODIPY 581/591 probe were used to estimate the level of cellular lipid peroxidation. Fe2+ content was examined by the kit. In addition, the expression of proteins related to inflammation-, ferroptosis- and Janus kinase 2 (JAK2)/signal transducer, and activator of transcription 3 (STAT3) signaling was detected by the Western blot analysis. The results revealed that FABP4 was significantly upregulated in LPS-treated HK2 cells, the knockdown of which elevated the viability, whereas alleviated the inflammation and ferroptosis in HK2 cells challenged with LPS. In addition, down-regulation of FABP4 inactivated JAK2/STAT3 signaling. JAK2/STAT3 stimulator (colivelin) and ferroptosis activator (Erastin) partially restored the effects of FABP4 interference on LPS-triggered inflammation and ferroptosis in HK2 cells. Together, FABP4 knockdown inhibited ferroptosis to alleviate LPS-induced injury of renal tubular epithelial cells through suppressing JAK2/STAT3 signaling.


Sujet(s)
Cellules épithéliales , Protéines de liaison aux acides gras , Ferroptose , Kinase Janus-2 , Tubules rénaux , Facteur de transcription STAT-3 , Transduction du signal , Humains , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/génétique , Atteinte rénale aigüe/anatomopathologie , Atteinte rénale aigüe/induit chimiquement , Lignée cellulaire , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/anatomopathologie , Protéines de liaison aux acides gras/métabolisme , Protéines de liaison aux acides gras/génétique , Ferroptose/effets des médicaments et des substances chimiques , Kinase Janus-2/métabolisme , Tubules rénaux/anatomopathologie , Tubules rénaux/métabolisme , Tubules rénaux/effets des médicaments et des substances chimiques , Lipopolysaccharides/toxicité , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique
3.
Acta Pharmacol Sin ; 45(8): 1604-1617, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38589689

RÉSUMÉ

Heart failure with preserved ejection fraction (HFpEF) is closely associated with metabolic derangement. Sodium glucose cotransporter-2 inhibitors (SGLT2i) and glucagon-like peptide-1 receptor agonists (GLP-1RA) exert anti-HFpEF effects, but the underlying mechanisms remain unclear. In this study, we explored the anti-HFpEF effects of empagliflozin and liraglutide and the underlying molecular mechanisms in a mouse model of HFpEF. This model was established by high-fat diet (HFD) feeding plus Nω-nitro-L-arginine methyl ester (L-NAME) treatment. The mice were treated with empagliflozin (20 mg·kg-1·d-1, i.g.) or liraglutide (0.3 mg·kg-1·d-1, i.p.) or their combination for 4 weeks. At the end of the experimental protocol, cardiac function was measured using ultrasound, then mice were euthanized and heart, liver, and kidney tissues were collected. Nuclei were isolated from frozen mouse ventricular tissue for single-nucleus RNA-sequencing (snRNA-seq). We showed that administration of empagliflozin or liraglutide alone or in combination significantly improved diastolic function, ameliorated cardiomyocyte hypertrophy and cardiac fibrosis, as well as exercise tolerance but no synergism was observed in the combination group. Furthermore, empagliflozin and/or liraglutide lowered body weight, improved glucose metabolism, lowered blood pressure, and improved liver and kidney function. After the withdrawal of empagliflozin or liraglutide for 1 week, these beneficial effects tended to diminish. The snRNA-seq analysis revealed a subcluster of myocytes, in which Erbb4 expression was down-regulated under HFpEF conditions, and restored by empagliflozin or liraglutide. Pseudo-time trajectory analysis and cell-to-cell communication studies confirmed that the Erbb4 pathway was a prominent pathway essential for both drug actions. In the HFpEF mouse model, both empagliflozin and liraglutide reversed Erbb4 down-regulation. In rat h9c2 cells, we showed that palmitic acid- or high glucose-induced changes in PKCα and/or ERK1/2 phosphorylation at least in part through Erbb4. Collectively, the single-cell atlas reveals the anti-HFpEF mechanism of empagliflozin and liraglutide, suggesting that Erbb4 pathway represents a new therapeutic target for HFpEF. Effects and mechanisms of action of empagliflozin and liraglutide in HFpEF mice. HFpEF was induced with a high-fat diet and L-NAME for 15 weeks, and treatment with empagliflozin and liraglutide improved the HFpEF phenotype. Single nucleus RNA sequencing (snRNA-seq) was used to reveal the underlying mechanism of action of empagliflozin and liraglutide.


Sujet(s)
Composés benzhydryliques , Glucosides , Défaillance cardiaque , Liraglutide , Souris de lignée C57BL , Transduction du signal , Inhibiteurs du cotransporteur sodium-glucose de type 2 , Animaux , Composés benzhydryliques/pharmacologie , Composés benzhydryliques/usage thérapeutique , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Liraglutide/pharmacologie , Liraglutide/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Mâle , Souris , Défaillance cardiaque/traitement médicamenteux , Défaillance cardiaque/métabolisme , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique , Alimentation riche en graisse , Débit systolique/effets des médicaments et des substances chimiques , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Modèles animaux de maladie humaine
4.
Acta Pharmacol Sin ; 45(6): 1316-1320, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38459255

RÉSUMÉ

Within the context of residual cardiovascular risk in post-statin era, emerging evidence from epidemiologic and human genetic studies have demonstrated that triglyceride (TG)-rich lipoproteins and their remnants are causally related to cardiovascular risk. While, carriers of loss-of-function mutations of ApoC3 have low TG levels and are protected from cardiovascular disease (CVD). Of translational significance, siRNAs/antisense oligonucleotide (ASO) targeting ApoC3 is beneficial for patients with atherosclerotic CVD. Therefore, animal models of atherosclerosis with both hypercholesterolemia and hypertriglyceridemia are important for the discovery of novel therapeutic strategies targeting TG-lowering on top of traditional cholesterol-lowering. In this study, we constructed a novel mouse model of familial combined hyperlipidemia through inserting a human ApoC3 transgene (hApoC3-Tg) into C57BL/6 J mice and injecting a gain-of-function variant of adeno-associated virus-proprotein convertase subtilisin/kexin type 9 (AAV-PCSK9)-D377Y concurrently with high cholesterol diet (HCD) feeding for 16 weeks. In the last 10 weeks, hApoC3-Tg mice were orally treated with a combination of atorvastatin (10 mg·kg-1·d-1) and fenofibrate (100 mg·kg-1·d-1). HCD-treated hApoC3-Tg mice demonstrated elevated levels of serum TG, total cholesterol (TC) and low density lipoprotein-cholesterol (LDL-C). Oral administration of atorvastatin and fenofibrate significantly decreased the plaque sizes of en face aorta, aortic sinus and innominate artery accompanied by improved lipid profile and distribution. In summary, this novel mouse model is of considerable clinical relevance for evaluation of anti-atherosclerotic drugs by targeting both hypercholesterolemia and hypertriglyceridemia.


Sujet(s)
Athérosclérose , Modèles animaux de maladie humaine , Hyperlipidémie familiale mixte , Souris de lignée C57BL , Souris transgéniques , Animaux , Athérosclérose/traitement médicamenteux , Humains , Souris , Hyperlipidémie familiale mixte/traitement médicamenteux , Hyperlipidémie familiale mixte/génétique , Apolipoprotéine C-III/génétique , Mâle , Proprotéine convertase 9/génétique , Proprotéine convertase 9/métabolisme , Hypolipémiants/usage thérapeutique , Hypolipémiants/pharmacologie , Triglycéride/sang , Alimentation riche en graisse , Atorvastatine/usage thérapeutique , Atorvastatine/pharmacologie
5.
Heliyon ; 10(2): e22772, 2024 Jan 30.
Article de Anglais | MEDLINE | ID: mdl-38298668

RÉSUMÉ

Purpose: Sepsis-induced acute lung injury is related to high mortality. MiR-2113 possesses important functions in human diseases. This research aimed to clarify the role and mechanism of miR-2113 in sepsis-induced acute lung injury. Methods: The expression of miR-2113 in lipopolysaccharide (LPS)-induced MLE-12 cells, serum of sepsis patients, and cecal ligation and puncture mouse models was examined using quantitative real-time PCR. The functions of miR-2113 in LPS-treated MLE-12 cells were estimated by Cell Counting Kit-8 assay, flow cytometry, enzyme-linked immunosorbent assay, Western blot, and immunofluorescence. The influences of miR-2113 in cecal ligation and puncture-induced acute lung injury in mice were assessed by hematoxylin-eosin staining, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay, acute pulmonary dysfunction analysis, lactate dehydrogenase levels and total protein concentrations in bronchoalveolar lavage fluid, and Masson staining. Also, the mechanism of miR-2113 was examined using a dual-luciferase reporter assay. Results: MiR-2113 expression was decreased in LPS-induced MLE-12 cells, serum of sepsis patients, and cecal ligation and puncture mouse models. miR-2113 overexpression restored LPS-reduced MLE-12 cell proliferation, but alleviated LPS-induced apoptosis and markers of inflammation and fibrosis in MLE-12 cells. Moreover, we found that miR-2113 mimic reduced LPS-induced MLE-12 cell injury by negatively regulating high-mobility group box 1. In vivo data further confirmed that miR-2113 overexpression alleviated acute pulmonary dysfunction, inflammation and fibrosis in cecal ligation and puncture-induced sepsis mice. Conclusion: MiR-2113 relieved sepsis-induced acute pulmonary dysfunction, inflammation and fibrosis through decreasing high-mobility group box 1.

6.
Acta Pharmacol Sin ; 45(1): 23-35, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37644131

RÉSUMÉ

Heart failure (HF) with preserved ejection fraction (HFpEF) is currently a preeminent challenge for cardiovascular medicine. It has a poor prognosis, increasing mortality, and is escalating in prevalence worldwide. Despite accounting for over 50% of all HF patients, the mechanistic underpinnings driving HFpEF are poorly understood, thus impeding the discovery and development of mechanism-based therapies. HFpEF is a disease syndrome driven by diverse comorbidities, including hypertension, diabetes and obesity, pulmonary hypertension, aging, and atrial fibrillation. There is a lack of high-fidelity animal models that faithfully recapitulate the HFpEF phenotype, owing primarily to the disease heterogeneity, which has hampered our understanding of the complex pathophysiology of HFpEF. This review provides an updated overview of the currently available animal models of HFpEF and discusses their characteristics from the perspective of energy metabolism. Interventional strategies for efficiently utilizing energy substrates in preclinical HFpEF models are also discussed.


Sujet(s)
Défaillance cardiaque , Hypertension artérielle , Animaux , Humains , Débit systolique/physiologie , Comorbidité , Découverte de médicament
7.
Acta Pharmacol Sin ; 44(12): 2358-2375, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37550526

RÉSUMÉ

Atherosclerosis, one of the life-threatening cardiovascular diseases (CVDs), has been demonstrated to be a chronic inflammatory disease, and inflammatory and immune processes are involved in the origin and development of the disease. Toll-like receptors (TLRs), a class of pattern recognition receptors that trigger innate immune responses by identifying pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs), regulate numerous acute and chronic inflammatory diseases. Recent studies reveal that TLRs have a vital role in the occurrence and development of atherosclerosis, including the initiation of endothelial dysfunction, interaction of various immune cells, and activation of a number of other inflammatory pathways. We herein summarize some other inflammatory signaling pathways, protein molecules, and cellular responses associated with TLRs, such as NLRP3, Nrf2, PCSK9, autophagy, pyroptosis and necroptosis, which are also involved in the development of AS. Targeting TLRs and their regulated inflammatory events could be a promising new strategy for the treatment of atherosclerotic CVDs. Novel drugs that exert therapeutic effects on AS through TLRs and their related pathways are increasingly being developed. In this article, we comprehensively review the current knowledge of TLR signaling pathways in atherosclerosis and actively seek potential therapeutic strategies using TLRs as a breakthrough point in the prevention and therapy of atherosclerosis.


Sujet(s)
Athérosclérose , Proprotéine convertase 9 , Humains , Proprotéine convertase 9/métabolisme , Récepteurs de type Toll/métabolisme , Transduction du signal/physiologie , Athérosclérose/métabolisme
8.
Acta Pharmacol Sin ; 44(8): 1625-1636, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-36997664

RÉSUMÉ

Vascular calcification is caused by the deposition of calcium salts in the intimal or tunica media layer of the aorta, which increases the risk of cardiovascular events and all-cause mortality. However, the mechanisms underlying vascular calcification are not fully clarified. Recently it has been shown that transcription factor 21 (TCF21) is highly expressed in human and mouse atherosclerotic plaques. In this study we investigated the role of TCF21 in vascular calcification and the underlying mechanisms. In carotid artery atherosclerotic plaques collected from 6 patients, we found that TCF21 expression was upregulated in calcific areas. We further demonstrated TCF21 expression was increased in an in vitro vascular smooth muscle cell (VSMC) osteogenesis model. TCF21 overexpression promoted osteogenic differentiation of VSMC, whereas TCF21 knockdown in VSMC attenuated the calcification. Similar results were observed in ex vivo mouse thoracic aorta rings. Previous reports showed that TCF21 bound to myocardin (MYOCD) to inhibit the transcriptional activity of serum response factor (SRF)-MYOCD complex. We found that SRF overexpression significantly attenuated TCF21-induced VSMC and aortic ring calcification. Overexpression of SRF, but not MYOCD, reversed TCF21-inhibited expression of contractile genes SMA and SM22. More importantly, under high inorganic phosphate (3 mM) condition, SRF overexpression reduced TCF21-induced expression of calcification-related genes (BMP2 and RUNX2) as well as vascular calcification. Moreover, TCF21 overexpression enhanced IL-6 expression and downstream STAT3 activation to facilitate vascular calcification. Both LPS and STAT3 could induce TCF21 expression, suggesting that the inflammation and TCF21 might form a positive feedback loop to amplify the activation of IL-6/STAT3 signaling pathway. On the other hand, TCF21 induced production of inflammatory cytokines IL-1ß and IL-6 in endothelial cells (ECs) to promote VSMC osteogenesis. In EC-specific TCF21 knockout (TCF21ECKO) mice, VD3 and nicotine-induced vascular calcification was significantly reduced. Our results suggest that TCF21 aggravates vascular calcification by activating IL-6/STAT3 signaling and interplay between VSMC and EC, which provides new insights into the pathogenesis of vascular calcification. TCF21 enhances vascular calcification by activating the IL-6-STAT3 signaling pathway. TCF21 inhibition may be a new potential therapeutic strategy for the prevention and treatment of vascular calcification.


Sujet(s)
Plaque d'athérosclérose , Calcification vasculaire , Animaux , Humains , Souris , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Cellules cultivées , Cellules endothéliales/métabolisme , Interleukine-6/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Ostéogenèse , Plaque d'athérosclérose/métabolisme , Transduction du signal , Facteur de transcription STAT-3/métabolisme , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie
9.
Acta Pharmacol Sin ; 44(4): 695-709, 2023 Apr.
Article de Anglais | MEDLINE | ID: mdl-36253560

RÉSUMÉ

The fight against coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection is still raging. However, the pathophysiology of acute and post-acute manifestations of COVID-19 (long COVID-19) is understudied. Endothelial cells are sentinels lining the innermost layer of blood vessel that gatekeep micro- and macro-vascular health by sensing pathogen/danger signals and secreting vasoactive molecules. SARS-CoV-2 infection primarily affects the pulmonary system, but accumulating evidence suggests that it also affects the pan-vasculature in the extrapulmonary systems by directly (via virus infection) or indirectly (via cytokine storm), causing endothelial dysfunction (endotheliitis, endothelialitis and endotheliopathy) and multi-organ injury. Mounting evidence suggests that SARS-CoV-2 infection leads to multiple instances of endothelial dysfunction, including reduced nitric oxide (NO) bioavailability, oxidative stress, endothelial injury, glycocalyx/barrier disruption, hyperpermeability, inflammation/leukocyte adhesion, senescence, endothelial-to-mesenchymal transition (EndoMT), hypercoagulability, thrombosis and many others. Thus, COVID-19 is deemed as a (micro)vascular and endothelial disease. Of translational relevance, several candidate drugs which are endothelial protective have been shown to improve clinical manifestations of COVID-19 patients. The purpose of this review is to provide a latest summary of biomarkers associated with endothelial cell activation in COVID-19 and offer mechanistic insights into the molecular basis of endothelial activation/dysfunction in macro- and micro-vasculature of COVID-19 patients. We envisage further development of cellular models and suitable animal models mimicking endothelial dysfunction aspect of COVID-19 being able to accelerate the discovery of new drugs targeting endothelial dysfunction in pan-vasculature from COVID-19 patients.


Sujet(s)
COVID-19 , Cellules endothéliales , Animaux , Humains , Marqueurs biologiques , COVID-19/anatomopathologie , Cellules endothéliales/anatomopathologie , Syndrome de post-COVID-19 , SARS-CoV-2
10.
Acta Pharmacol Sin ; 43(9): 2173-2190, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35046517

RÉSUMÉ

Colchicine is an ancient herbal drug derived from Colchicum autumnale. It was first used to treat familial Mediterranean fever and gout. Based on its unique efficacy as an anti-inflammatory agent, colchicine has been used in the therapy of cardiovascular diseases including coronary artery disease, atherosclerosis, recurrent pericarditis, vascular restenosis, heart failure, and myocardial infarction. More recently, colchicine has also shown therapeutic efficacy in alleviating cardiovascular complications of COVID-19. COLCOT and LoDoCo2 are two milestone clinical trials that confirm the curative effect of long-term administration of colchicine in reducing the incidence of cardiovascular events in patients with coronary artery disease. There is growing interest in studying the anti-inflammatory mechanisms of colchicine. The anti-inflammatory action of colchicine is mediated mainly through inhibiting the assembly of microtubules. At the cellular level, colchicine inhibits the following: (1) endothelial cell dysfunction and inflammation; (2) smooth muscle cell proliferation and migration; (3) macrophage chemotaxis, migration, and adhesion; (4) platelet activation. At the molecular level, colchicine reduces proinflammatory cytokine release and inhibits NF-κB signaling and NLRP3 inflammasome activation. In this review, we summarize the current clinical trials with proven curative effect of colchicine in treating cardiovascular diseases. We also systematically discuss the mechanisms of colchicine action in cardiovascular therapeutics. Altogether, colchicine, a bioactive constituent from an ancient medicinal herb, exerts unique anti-inflammatory effects and prominent cardiovascular actions, and will charter a new page in cardiovascular medicine.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Agents cardiovasculaires , Maladie des artères coronaires , Infarctus du myocarde , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Agents cardiovasculaires/pharmacologie , Agents cardiovasculaires/usage thérapeutique , Colchicine/pharmacologie , Colchicine/usage thérapeutique , Maladie des artères coronaires/traitement médicamenteux , Humains , Infarctus du myocarde/traitement médicamenteux
11.
Dis Markers ; 2020: 8895900, 2020.
Article de Anglais | MEDLINE | ID: mdl-32831973

RÉSUMÉ

OBJECTIVE: Family with sequence similarity 19 member A5 (FAM19A5), a novel chemokine-like peptide, is a secreted protein mainly expressed in the brain. FAM19A5 was recently found to be involved in a variety of neurological diseases; however, its correlation with vascular dementia (VaD) remains unclear. The aim of the study is to explore the association between serum FAM19A5 and cognitive impairment in subjects with VaD. METHOD: 136 VaD subjects and 81 normal controls were recruited in the study. Their demographic and clinical baseline data were collected on admission. All subjects received Mini-Mental State Examination (MMSE) evaluation, which was used to test their cognitive functions. A sandwich enzyme-linked immunosorbent assay (ELISA) was applied to detect the serum levels of FAM19A5. RESULTS: No significant differences were found between the two groups regarding the demographic and clinical baseline data (p > 0.05). The serum FAM19A5 levels were significantly higher compared to normal controls (p < 0.001). The Spearman correlation analysis indicated that serum FAM19A5 levels and MMSE scores have a significant negative correlation in VaD patients (r = -0.414, <0.001). Further multiple regression analysis indicated that serum FAM19A5 levels were independent risk predictors for cognitive functions in VaD (ß = 0.419, p = 0.031). CONCLUSION: The serum FAM19A5 level of VaD patients is significantly increased, which may serve as a biomarker to predict cognitive function of VaD.


Sujet(s)
Dysfonctionnement cognitif/diagnostic , Cytokines/sang , Démence vasculaire/psychologie , Régulation positive , Sujet âgé , Marqueurs biologiques/sang , Études cas-témoins , Dysfonctionnement cognitif/sang , Démence vasculaire/sang , Femelle , Humains , Mâle , Tests de l'état mental et de la démence , Adulte d'âge moyen , Analyse de régression
12.
Inflammation ; 42(3): 987-993, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-30707387

RÉSUMÉ

Umbilical cord blood mesenchymal stem cells (UCB-MSCs) have been shown to be a source of stem cells for use in cellular therapies and have immunomodulatory effects on several immune cells in an inflammatory environment. However, whether UCB-MSCs have immunomodulatory effects against lipopolysaccharide (LPS)-induced inflammatory cytokine secretion in macrophages and whether it is involved in phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway remain unclear. After co-culture of UCB-MSCs and phorbol 12-myristate 13-acetate (PMA)-activated human THP-1 cells using a transwell system, it showed that LPS significantly induced increases in the expression levels of interleukin 10 (IL-10), interleukin 37 (IL-37), phospho-PI3K (p-PI3K), and phospho-Akt (p-Akt) in macrophages. UCB-MSCs upregulated the expression of IL-10, IL-37, p-PI3K, and p-Akt, while it had no obvious effect on PI3K and Akt levels. Inhibitors of PI3K (LY294002) significantly suppressed the expression of IL-10, IL-37, p-PI3K, and p-Akt; however, it had no effect on the expression levels of PI3K and Akt. The present study demonstrated that UCB-MSCs increased the LPS-stimulated expression of IL-10 and IL-37 in macrophages through the PI3K/Akt signaling pathway.


Sujet(s)
Interleukine-10/biosynthèse , Interleukine-1/biosynthèse , Macrophages/métabolisme , Cellules souches mésenchymateuses/physiologie , Techniques de coculture , Sang foetal/cytologie , Humains , Lipopolysaccharides/pharmacologie , Macrophages/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/cytologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Cellules THP-1/cytologie , Cellules THP-1/métabolisme
13.
Pharmacol Res ; 141: 541-550, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30616017

RÉSUMÉ

Rutaecarpine is a bioactive alkaloid isolated from Evodia rutaecarpa (Wu Zhu Yu, Family: Rutaceae), a versatile medicinal herb which is clinically used to treat headache, abdominal pain, postpartum hemorrhage, dysentery, and amenorrhea in China. As one of the most representative indolopyridoquinazoline alkaloids of Evodia rutaecarpa, rutaecarpine has broad pharmacological actions in treating various cardiovascular, cerebrovascular, and metabolic diseases. The cardiovascular actions of rutaecarpine have aroused intense research interest due to its purported inotropic and chronotropic, vasodilatory, anti-platelet activation, anti-oxidant, anti-inflammatory, and lipid-lowering effects. Biochemical and pharmacological studies have illustrated the molecular targets of rutaecarpine, such as TRPV1, CGRP, AMPK, ABCA1, and ß1-AR. Furthermore, several rutaecarpine derivatives (such as bromorutaecarpine and fluororutaecarpine) have been shown to possess cardioprotective and vasculoprotective effects with improved safety profile. Hereby, we provide a systematic overview of pharmacological actions, toxicological effects, and molecular targets of rutaecarpine in cardiovascular disease prevention/treatment, aiming to exploit the therapeutic potential of rutaecarpine and its derivatives in treating cardiovascular diseases.


Sujet(s)
Cardiotoniques/pharmacologie , Maladies cardiovasculaires/traitement médicamenteux , Evodia/composition chimique , Alcaloïdes indoliques/pharmacologie , Quinazolines/pharmacologie , Alcaloïdes/composition chimique , Alcaloïdes/pharmacologie , Alcaloïdes/usage thérapeutique , Animaux , Anti-inflammatoires/composition chimique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Antioxydants/composition chimique , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique , Cardiotoniques/composition chimique , Cardiotoniques/usage thérapeutique , Humains , Hypolipémiants/composition chimique , Hypolipémiants/pharmacologie , Hypolipémiants/usage thérapeutique , Alcaloïdes indoliques/composition chimique , Alcaloïdes indoliques/usage thérapeutique , Antiagrégants plaquettaires/composition chimique , Antiagrégants plaquettaires/pharmacologie , Antiagrégants plaquettaires/usage thérapeutique , Quinazolines/composition chimique , Quinazolines/usage thérapeutique , Vasodilatateurs/composition chimique , Vasodilatateurs/pharmacologie , Vasodilatateurs/usage thérapeutique
14.
Pharmacol Res ; 139: 62-75, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30408571

RÉSUMÉ

Cardiovascular and cerebrovascular diseases are the main cause of mortality worldwide, currently with less than optimum therapeutic options. Danhong injection (DHI) is a medicinal preparation based on two eminent Chinese herbal medicines, Salviae Miltiorrhizae (Dan Shen; family: Lamiaceae) and Flos Carthami (Hong Hua; family: Compositae/Asteraceae). DHI has been mainly used in the clinical therapy of cardiovascular (such as acute coronary syndrome and angina pectoris) and cerebrovascular diseases (such as stroke) in China for many years. The pharmacological properties of DHI include anti-inflammatory, anti-oxidant, anti-coagulatory, hypolipidemic, anti-apoptotic, vasodilatory, and angiogenesis-promoting actions. DHI offers a safe and effective therapeutic agent against cardiovascular and cerebrovascular diseases by modulating multiple disease-relevant signaling pathways and molecular targets. Herein, we provide a comprehensive review of the phytochemistry, therapeutic effects, molecular mechanisms, and adverse reactions of DHI in cardiovascular and cerebrovascular diseases. We also highlight the latest pharmacological advances and therapeutic potential of this promising herb-derived cardiovascular drug preparation.


Sujet(s)
Maladies cardiovasculaires/traitement médicamenteux , Angiopathies intracrâniennes/traitement médicamenteux , Médicaments issus de plantes chinoises/usage thérapeutique , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Humains
15.
Ann N Y Acad Sci ; 1443(1): 34-53, 2019 05.
Article de Anglais | MEDLINE | ID: mdl-30381837

RÉSUMÉ

LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1; also known as OLR1) is the dominant receptor that recognizes and internalizes oxidized low-density lipoproteins (ox-LDLs) in endothelial cells. Several genetic variants of LOX-1 are associated with the risk and severity of coronary artery disease. The LOX-1-ox-LDL interaction induces endothelial dysfunction, leukocyte adhesion, macrophage-derived foam cell formation, smooth muscle cell proliferation and migration, and platelet activation. LOX-1 activation eventually leads to the rupture of atherosclerotic plaques and acute cardiovascular events. In addition, LOX-1 can be cleaved to generate soluble LOX-1 (sLOX-1), which is a useful diagnostic and prognostic marker for atherosclerosis-related diseases in human patients. Of therapeutic relevance, several natural products and clinically used drugs have emerged as LOX-1 inhibitors that have antiatherosclerotic actions. We hereby provide an updated overview of role of LOX-1 in atherosclerosis and associated vascular diseases, with an aim to highlighting the potential of LOX-1 as a novel theranostic tool for cardiovascular disease prevention and treatment.


Sujet(s)
Athérosclérose/traitement médicamenteux , Maladie des artères coronaires/traitement médicamenteux , Récepteurs éboueurs de classe E/effets des médicaments et des substances chimiques , Marqueurs biologiques/métabolisme , Humains , Facteurs de risque , Récepteurs éboueurs de classe E/génétique , Récepteurs éboueurs de classe E/métabolisme
16.
J Agric Food Chem ; 66(48): 12748-12755, 2018 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-30441891

RÉSUMÉ

Microalgae starch is receiving increasing attention as a renewable feedstock for biofuel production. Raw microalgae starch from Tetraselmis subcordiformis was proven to be very efficiently hydrolyzed by an α-amylase (AmyP) of glycoside hydrolase subfamily GH13_37 below the temperature of gelatinization (40 °C). The hydrolysis degree reached 74.4 ± 2.2% for 4% raw microalgae starch and 53.2 ± 1.7% for 8% raw microalgae starch after only 2 h. The hydrolysis efficiency was significantly stimulated by calcium ions. The enzyme catalysis of AmyP and its mutants (Q306A and E347A) suggested that calcium ions contributed to the hydrolysis of cyclic structures in raw microalgae starch by a distinctive calcium-binding site Ca2 of AmyP. The study explored raw microalgae starch as a new resource for cold enzymatic hydrolysis and extended our knowledge on the function of calcium in amylolytic enzyme.


Sujet(s)
Protéines bactériennes/composition chimique , Chlorophyta/composition chimique , Microalgues/composition chimique , Extraits de plantes/composition chimique , Amidon/composition chimique , alpha-Amylases/composition chimique , Séquence d'acides aminés , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Biocatalyse , Glycosidases/composition chimique , Glycosidases/génétique , Glycosidases/métabolisme , Hydrolyse , Cinétique , Données de séquences moléculaires , Famille multigénique , Extraits de plantes/métabolisme , Alignement de séquences , Amidon/métabolisme , Spécificité du substrat , Température , alpha-Amylases/génétique , alpha-Amylases/métabolisme
17.
Acta Pharmacol Sin ; 39(5): 802-824, 2018 May.
Article de Anglais | MEDLINE | ID: mdl-29698387

RÉSUMÉ

Salvia miltiorrhiza Burge (Danshen) is an eminent medicinal herb that possesses broad cardiovascular and cerebrovascular protective actions and has been used in Asian countries for many centuries. Accumulating evidence suggests that Danshen and its components prevent vascular diseases, in particular, atherosclerosis and cardiac diseases, including myocardial infarction, myocardial ischemia/reperfusion injury, arrhythmia, cardiac hypertrophy and cardiac fibrosis. The published literature indicates that lipophilic constituents (tanshinone I, tanshinone IIa, tanshinone IIb, cryptotanshinone, dihydrotanshinone, etc) as well as hydrophilic constituents (danshensu, salvianolic acid A and B, protocatechuic aldehyde, etc) contribute to the cardiovascular protective actions of Danshen, suggesting a potential synergism among these constituents. Herein, we provide a systematic up-to-date review on the cardiovascular actions and therapeutic potential of major pharmacologically active constituents of Danshen. These bioactive compounds will serve as excellent drug candidates in small-molecule cardiovascular drug discovery. This article also provides a scientific rationale for understanding the traditional use of Danshen in cardiovascular therapeutics.


Sujet(s)
Cardiotoniques/usage thérapeutique , Maladies cardiovasculaires/traitement médicamenteux , Médicaments issus de plantes chinoises/usage thérapeutique , Animaux , Maladies cardiovasculaires/physiopathologie , Synergie des médicaments , Cellules endothéliales/effets des médicaments et des substances chimiques , Fibrinolytiques/usage thérapeutique , Humains , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Salvia miltiorrhiza
18.
Cancer Cell Int ; 17: 64, 2017.
Article de Anglais | MEDLINE | ID: mdl-28615992

RÉSUMÉ

BACKGROUND: Recent studies have verified that long noncoding RNAs (lncRNAs) involved in many biological functions and play crucial roles in human cancers progression, the study aimed to detect the association between long non-coding RNA HOXA11-AS and epithelial-mesenchymal transition (EMT) process in non-small cell lung cancer (NSCLC). METHODS: The lncRNA HOXA11-AS expression levels were determined by quantitative real-time polymerase chain reaction (qRT-PCR) assays in 78 paired of tumor tissue and adjacent normal tissue samples in NSCLC patients. Kaplan-Meier survival curves and log-rank test was used to examine the association between lncRNA HOXA11-AS expression and the over survival time in NSCLC patients. Transwell invasion assay was performed to detect the cell invasion ability. QRT-PCR and western-blot analysis detected the mRNA and protein expression of EMT related transcription factors ZEB1/ZEB2, Snail1/2 and EMT marker E-cadherin and N-cadherin in NSCLC cells. RIP and Chromatin immunoprecipitation assays were performed to analyze the association between lncRNA HOXA11-AS and miR-200b expression in NSCLC cells. RESULTS: The lncRNA HOXA11-AS expression levels were significantly higher in NSCLC tissues compared with adjacent normal tissues and higher HOXA11-AS expression levels had a poor prognosis in NSCLC patients. Furthermore, knockdown of lncRNA HOXA11-AS in A549 and H1299 cells dramatically inhibited cell invasive abilities. Besides, the transcription levels and protein levels of EMT related transcription factors ZEB1/ZEB2, Snail1/2, and EMT maker N-cadherin were down-regulated after lncRNA HOXA11-AS was knocked down, but the mRNA and protein expression levels of EMT maker E-cadherin was increasing in A549 and H1299 cells. The mechanistic findings showed demonstrated that HOXA11-AS interacted with EZH2 and DNMT1 and recruited them to the miR-200b promoter regions to repress miR-200b expression in NSCLC cells, which promoted cell EMT in NSCLC. CONCLUSIONS: Our results showed that up-regulation of lncRNA HOXA11-AS predicted a poor prognosis and lncRNA HOXA11-AS promoted cell epithelial-mesenchymal transition (EMT) by inhibiting miR-200b expression in NSCLC.

19.
Biochem Biophys Res Commun ; 447(2): 271-7, 2014 May 02.
Article de Anglais | MEDLINE | ID: mdl-24704450

RÉSUMÉ

The molecular mechanisms of multiple myeloma are not well defined. EEN is an endocytosis-regulating molecule. Here we report that EEN regulates the proliferation and survival of multiple myeloma cells, by regulating IGF-1 secretion. In the present study, we observed that EEN expression paralleled with cell proliferation, EEN accelerated cell proliferation, facilitated cell cycle transition from G1 to S phase by regulating cyclin-dependent kinases (CDKs) pathway, and delayed cell apoptosis via Bcl2/Bax-mitochondrial pathway. Mechanistically, we found that EEN was indispensable for insulin-like growth factor-1 (IGF-1) secretion and the activation of protein kinase B-mammalian target of rapamycin (Akt-mTOR) pathway. Exogenous IGF-1 overcame the phenotype of EEN depletion, while IGF-1 neutralization overcame that of EEN over-expression. Collectively, these data suggest that EEN may play a pivotal role in excessive cell proliferation and insufficient cell apoptosis of bone marrow plasma cells in multiple myeloma. Therefore, EEN may represent a potential diagnostic marker or therapeutic target for multiple myeloma.


Sujet(s)
Marqueurs biologiques tumoraux/physiologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Myélome multiple/anatomopathologie , Récepteur IGF de type 1/métabolisme , Apoptose , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Survie cellulaire , Points de contrôle de la phase G1 du cycle cellulaire , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Myélome multiple/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteur IGF de type 1/pharmacologie , Sérine-thréonine kinases TOR/métabolisme
20.
Arterioscler Thromb Vasc Biol ; 33(2): e31-8, 2013 Feb.
Article de Anglais | MEDLINE | ID: mdl-23220274

RÉSUMÉ

OBJECTIVE: Cell-surface localization and intracellular trafficking are essential for the function of ATP-binding cassette transporter A-1 (ABCA1). However, regulation of these activities is still largely unknown. Brefeldin A, an uncompetitive inhibitor of brefeldin A-inhibited guanine nucleotide-exchange proteins (BIGs), disturbs the intracellular distribution of ABCA1, and thus inhibits cholesterol efflux. This study aimed to define the possible roles of BIGs in regulating ABCA1 trafficking and cholesterol efflux, and further to explore the potential mechanism. METHODS AND RESULTS: By vesicle immunoprecipitation, we found that BIG1 was associated with ABCA1 in vesicles preparation from rat liver. BIG1 depletion reduced surface ABCA1 on HepG2 cells, and inhibited by 60% cholesterol release. In contrast, BIG1 overexpression increased surface ABCA1 and cholesterol secretion. With partial restoration of BIG1 through overexpression in BIG1-depleted cells, surface ABCA1 was also restored. Biotinylation and glutathione cleavage revealed that BIG1 small interfering RNA dramatically decreased the internalization and recycling of ABCA1. This novel function of BIG1 was dependent on the guanine nucleotide-exchange activity and achieved through activation of ADP-ribosylation factor 1. CONCLUSIONS: BIG1, through its ability to activate ADP-ribosylation factor 1, regulates cell-surface levels and function of ABCA1, indicating a transcription-independent mechanism for controlling ABCA1 action.


Sujet(s)
Transporteurs ABC/métabolisme , Membrane cellulaire/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Facteur-1 d'ADP-ribosylation/métabolisme , Membre-1 de la sous-famille A des transporteurs à cassette liant l'ATP , Animaux , Apolipoprotéine A-I/métabolisme , Biotinylation , Cholestérol/métabolisme , Glutathion/métabolisme , Facteurs d'échange de nucléotides guanyliques/génétique , Cellules HeLa , Cellules HepG2 , Humains , Immunoprécipitation , Lipoprotéines LDL/métabolisme , Foie/métabolisme , Mâle , Transport des protéines , Interférence par ARN , Rats , Rat Sprague-Dawley , Facteurs temps , Transfection
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE