Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 90
Filtrer
1.
Mol Med Rep ; 30(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38994773

RÉSUMÉ

Subsequently to the publication of the above article, the authors have realized that, in Fig. 1A, the incorrect image was uploaded to show the ultrastructure of exos isolated from plasma and examined using transmission electron microscopy (essentially, the image in question had already appeared in an article published by the same research group in Journal of Cellular and Molecular Medicine). In addition,  the '+' and '-' signs for the 'Cell lysis' experiments shown underneath the gels in Fig. 1B were incorporated the wrong way around. The revised version of Fig. 1, showing the correct image in Fig. 1A and the correct labels in Fig. 1B, is shown below. Note that the errors made in assembling this figure did not have a major impact on either the results or the conclusions reported in this paper. The authors are grateful to the Editor of Molecular Medicine Reports for allowing them this opportunity to publish a corrigendum, and apologize to the readership of the Journal for any inconvenience caused. [Molecular Medicine Reports 27: 124, 2023; DOI: 10.3892/mmr.2023.13010].

2.
Int Immunopharmacol ; 136: 112338, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38850787

RÉSUMÉ

Cardiac fibrosis is a typical feature of cardiac pathological remodeling, which is associated with adverse clinical outcomes and has no effective therapy. Nicotine is an important risk factor for cardiac fibrosis, yet its underlying molecular mechanism remains poorly understood. This study aimed to identify its potential molecular mechanism in nicotine-induced cardiac fibrosis. Our results showed nicotine exposure led to the proliferation and transformation of cardiac fibroblasts (CFs) into myofibroblasts (MFs) by impairing autophagy flux. Through the use of drug affinity responsive target stability (DARTS) assay, cellular thermal shift assay (CETSA), and surface plasmon resonance (SPR) technology, it was discovered that nicotine directly increased the stability and protein levels of lactate dehydrogenase A (LDHA) by binding to it. Nicotine treatment impaired autophagy flux by regulating the AMPK/mTOR signaling pathway, impeding the nuclear translocation of transcription factor EB (TFEB), and reducing the activity of cathepsin B (CTSB). In vivo, nicotine treatment exacerbated cardiac fibrosis induced in spontaneously hypertensive rats (SHR) and worsened cardiac function. Interestingly, the absence of LDHA reversed these effects both in vitro and in vivo. Our study identified LDHA as a novel nicotine-binding protein that plays a crucial role in mediating cardiac fibrosis by blocking autophagy flux. The findings suggest that LDHA could potentially serve as a promising target for the treatment of cardiac fibrosis.


Sujet(s)
Autophagie , Fibrose , Nicotine , Animaux , Autophagie/effets des médicaments et des substances chimiques , Rats , Mâle , Rats de lignée SHR , Transduction du signal/effets des médicaments et des substances chimiques , Myocarde/anatomopathologie , Myocarde/métabolisme , Lactate dehydrogenase 5/métabolisme , Cellules cultivées , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Rat Sprague-Dawley
3.
Food Funct ; 15(12): 6359-6373, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38787699

RÉSUMÉ

The aim of this study was to evaluate the preventive role and underlying mechanisms of fucoxanthin (Fx) on lipopolysaccharide (LPS)-induced intestinal barrier injury in mice. Our results demonstrated that the oral administration of Fx (50 and 200 mg per kg body weight per day) for consecutive 7 days significantly alleviated the severity of LPS-induced intestinal barrier injury in mice, as evidenced by attenuating body weight loss, improving intestinal permeability, and ameliorating intestinal morphological damage such as reduction in the ratio of the villus length to the crypt depth (V/C), intestinal epithelium distortion, goblet cell depletion, and low mucin 2 (MUC2) expression. Fx also significantly mitigated LPS-induced excessive apoptosis of intestinal epithelial cells (IECs) and curbed the decrease of tight junction proteins including claudin-1, occludin, and zonula occludens-1 in the ileum and colon. Additionally, Fx effectively alleviated LPS-induced extensive infiltration of macrophages and neutrophils into the intestinal mucosa, the overproduction of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 1beta (IL-1ß) and IL-6, and gasdermin D (GSDMD)-mediated pyroptosis of IECs. The underlying mechanisms might be associated with inhibiting the activation of nuclear factor-kappa B (NF-κB), mitogen-activated protein kinases (MAPKs) and nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathways. Moreover, Fx also notably restrained intestinal reactive oxygen species (ROS), malondialdehyde and protein carbonylation levels in LPS-treated mice, and it might be mediated by activating the nuclear factor-erythroid 2 related factor 2 (Nrf2) signaling pathway. Overall, these findings indicated that Fx might be developed as a potential effective dietary supplement to prevent intestinal barrier injury.


Sujet(s)
Muqueuse intestinale , Lipopolysaccharides , Xanthophylles , Animaux , Souris , Xanthophylles/pharmacologie , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Lipopolysaccharides/effets indésirables , Mâle , Apoptose/effets des médicaments et des substances chimiques , Facteur de transcription NF-kappa B/métabolisme , Perméabilité , Souris de lignée C57BL , Protéines de la jonction serrée/métabolisme , Cytokines/métabolisme
4.
J Am Heart Assoc ; 13(11): e033669, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38818934

RÉSUMÉ

BACKGROUND: A phase 3 trial was conducted to evaluate the efficacy and safety of ongericimab, a monoclonal antibody that inhibits proprotein convertase subtilisin/kexin type 9, as an add-on treatment to optimized lipid-lowering therapy in Chinese patients with primary hypercholesterolemia and mixed dyslipidemia. METHODS AND RESULTS: A total of 806 patients who were receiving stable and optimized lipid-lowering therapy but did not achieve their low-density lipoprotein cholesterol (LDL-C) targets were enrolled and randomly assigned in a 2:1:2:1 ratio to receive either ongericimab 150 mg or matching placebo every 2 weeks, or ongericimab 300 mg or matching placebo every 4 weeks for 52 weeks. Efficacy and safety were evaluated in 802 patients who received at least 1 dose of ongericimab or placebo. The primary end point was the percentage change in LDL-C from baseline to week 24. Our findings demonstrated that the least-squares mean difference of percentage change in LDL-C from baseline to week 24 was -67.7% (95% CI, -72.5% to -63.0%; P<0.0001) in the ongericimab 150 mg every 2 weeks group compared with the placebo every 2 weeks group, and -61.2% (95% CI, -67.1% to -55.2%; P<0.0001) in the ongericimab 300 mg every 4 weeks group compared with the placebo every 4 weeks group. These reductions were sustained up to week 52. Furthermore, treatment with ongericimab favorably altered other lipid parameters. A similar incidence of adverse events was observed in the ongericimab and placebo groups. CONCLUSIONS: Ongericimab, as an add-on treatment to optimized lipid-lowering therapy, significantly reduced LDL-C and was well-tolerated in Chinese patients with primary hyperlipidemia and mixed dyslipidemia who did not achieve their LDL-C targets. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT04781114.


Sujet(s)
Cholestérol LDL , Dyslipidémies , Hypercholestérolémie , Humains , Mâle , Femelle , Adulte d'âge moyen , Hypercholestérolémie/traitement médicamenteux , Hypercholestérolémie/sang , Hypercholestérolémie/diagnostic , Cholestérol LDL/sang , Chine , Dyslipidémies/traitement médicamenteux , Dyslipidémies/sang , Dyslipidémies/diagnostic , Résultat thérapeutique , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/administration et posologie , Sujet âgé , Méthode en double aveugle , Inhibiteurs de PCSK9 , Adulte , Asiatiques , Proprotéine convertase 9/immunologie , Proprotéine convertase 9/métabolisme , Marqueurs biologiques/sang , Facteurs temps , Association de médicaments , Anticholestérolémiants/usage thérapeutique , Anticholestérolémiants/effets indésirables , Anticholestérolémiants/administration et posologie , Anticorps monoclonaux/usage thérapeutique , Anticorps monoclonaux/effets indésirables , Anticorps monoclonaux/administration et posologie , Peuples d'Asie de l'Est
5.
JAMA Cardiol ; 9(6): 523-531, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38630489

RÉSUMÉ

Importance: Purinergic receptor P2Y12 (P2Y12) inhibitor monotherapy after a certain period of dual antiplatelet therapy (DAPT) may be an attractive option of maintenance antiplatelet treatment for patients undergoing percutaneous coronary intervention (PCI) who are at both high bleeding and ischemic risk (birisk). Objective: To determine if extended P2Y12 inhibitor monotherapy with clopidogrel is superior to ongoing DAPT with aspirin and clopidogrel after 9 to 12 months of DAPT after PCI in birisk patients with acute coronary syndromes (ACS). Design, Setting, and Participants: This was a multicenter, double-blind, placebo-controlled, randomized clinical trial including birisk patients with ACS who had completed 9 to 12 months of DAPT after drug-eluting stent implantation and were free from adverse events for at least 6 months at 101 China centers between February 2018 and December 2020. Study data were analyzed from April 2023 to May 2023. Interventions: Patients were randomized either to clopidogrel plus placebo or clopidogrel plus aspirin for an additional 9 months. Main Outcomes and Measures: The primary end point was Bleeding Academic Research Consortium (BARC) types 2, 3, or 5 bleeding 9 months after randomization. The key secondary end point was major adverse cardiac and cerebral events (MACCE; the composite of all-cause death, myocardial infarction, stroke or clinically driven revascularization). The primary end point was tested for superiority, and the MACCE end point was tested for sequential noninferiority and superiority. Results: A total of 7758 patients (mean [SD] age, 64.8 [9.0] years; 4575 male [59.0%]) were included in this study. The primary end point of BARC types 2, 3, or 5 bleeding occurred in 95 of 3873 patients (2.5%) assigned to clopidogrel plus placebo and 127 of 3885 patients (3.3%) assigned to clopidogrel plus aspirin (hazard ratio [HR], 0.75; 95% CI, 0.57-0.97; difference, -0.8%; 95% CI, -1.6% to -0.1%; P = .03). The incidence of MACCE was 2.6% (101 of 3873 patients) in the clopidogrel plus placebo group and 3.5% (136 of 3885 patients) in the clopidogrel plus aspirin group (HR, 0.74; 95% CI, 0.57-0.96; difference, -0.9%; 95% CI, -1.7% to -0.1%; P < .001 for noninferiority; P = .02 for superiority). Conclusions and Relevance: Among birisk patients with ACS who completed 9 to 12 months of DAPT after drug-eluting stent implantation and were free from adverse events for at least 6 months before randomization, an extended 9-month clopidogrel monotherapy regimen was superior to continuing DAPT with clopidogrel in reducing clinically relevant bleeding without increasing ischemic events. Trial Registration: ClinicalTrials.gov Identifier: NCT03431142.


Sujet(s)
Syndrome coronarien aigu , Acide acétylsalicylique , Clopidogrel , Bithérapie antiplaquettaire , Hémorragie , Intervention coronarienne percutanée , Antiagrégants plaquettaires , Humains , Syndrome coronarien aigu/traitement médicamenteux , Clopidogrel/usage thérapeutique , Mâle , Femelle , Adulte d'âge moyen , Méthode en double aveugle , Hémorragie/induit chimiquement , Hémorragie/épidémiologie , Antiagrégants plaquettaires/usage thérapeutique , Sujet âgé , Acide acétylsalicylique/usage thérapeutique , Acide acétylsalicylique/administration et posologie , Intervention coronarienne percutanée/méthodes , Bithérapie antiplaquettaire/méthodes , Endoprothèses à élution de substances , Antagonistes des récepteurs purinergiques P2Y/usage thérapeutique , Antagonistes des récepteurs purinergiques P2Y/administration et posologie
6.
J Cell Mol Med ; 28(7): e18238, 2024 04.
Article de Anglais | MEDLINE | ID: mdl-38509729

RÉSUMÉ

Heart failure with preserved ejection fraction (HFpEF) accounts for approximately 50% of total heart failure patients and is characterized by peripheral circulation, cardiac remodelling and comorbidities (such as advanced age, obesity, hypertension and diabetes) with limited treatment options. Chidamide (HBI-8000) is a domestically produced benzamide-based histone deacetylase isoform-selective inhibitor used for the treatment of relapsed refractory peripheral T-cell lymphomas. Based on our in vivo studies, we propose that HBI-8000 exerts its therapeutic effects by inhibiting myocardial fibrosis and myocardial hypertrophy in HFpEF patients. At the cellular level, we found that HBI-8000 inhibits AngII-induced proliferation and activation of CFs and downregulates the expression of fibrosis-related factors. In addition, we observed that the HFpEF group and AngII stimulation significantly increased the expression of TGF-ß1 as well as phosphorylated p38MAPK, JNK and ERK, whereas the expression of the above factors was significantly reduced after HBI-8000 treatment. Activation of the TGF-ß1/MAPK pathway promotes the development of fibrotic remodelling, and pretreatment with SB203580 (p38MAPK inhibitor) reverses this pathological change. In conclusion, our data suggest that HBI-8000 inhibits fibrosis by modulating the TGF-ß1/MAPK pathway thereby improving HFpEF. Therefore, HBI-8000 may become a new hope for the treatment of HFpEF patients.


Sujet(s)
Défaillance cardiaque , Pyridines , Humains , Défaillance cardiaque/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Débit systolique , Récidive tumorale locale , Benzamides/pharmacologie , Fibrose
7.
Exp Ther Med ; 27(4): 145, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38476905

RÉSUMÉ

Thoracic aortic aneurysms (TAAs) are a major cause of death owing to weaker blood vessel walls and higher rupture rates in affected individuals. Vascular smooth muscle cells (VSMCs) are the predominant cell type within the aortic wall and their dysregulation may contribute to TAA progression. Enhancer of zeste homolog 2 (EZH2), a histone methyltransferase, is involved in several pathological processes; however, the biological functions and mechanisms underlying VSMC phenotype transition and vascular intimal hyperplasia remain unclear. The present study aimed to determine the involvement of EZH2 in mediating VSMC function in the development of TAAs. The expression of EZH2 was revealed to be elevated in patients with thoracic aortic dissection and TAA mouse model through western blotting and reverse transcription-quantitative PCR experiments. Subsequently, a mouse model was established using ß-aminopropionitrile. In vitro, EdU labeling, Transwell assay, wound healing assay and hematoxylin-eosin staining revealed that knocking down the Ezh2 gene could reduce the proliferation, invasion, migration, and calcification of mouse primary aortic smooth muscle cells. Flow cytometry analysis found that EZH2 deficiency increased cell apoptosis. Depletion of Ezh2 in mouse primary aortic VSMCs promoted the transformation of VSMCs from a synthetic to a contractile phenotype. Using RNA-sequencing analysis, it was demonstrated that Ezh2 regulated a group of genes, including integrin ß3 (Itgb3), which are critically involved in the extracellular matrix signaling pathway. qChIP found Ezh2 occupies the Itgb3 promoter, thereby suppressing the expression of Itgb3. Ezh2 promotes the invasion and calcification of VSMCs, and this promoting effect is partially reversed by co-knocking down Itgb3. In conclusion, the present study identified a previously unrecognized EZH2-ITGB3 regulatory axis and thus provides novel mechanistic insights into the pathophysiological function of EZH2. EZH2 may thus serve as a potential target for the management of TAAs.

8.
Biomed Pharmacother ; 173: 116396, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38460370

RÉSUMÉ

Aortic aneurysm/dissection (AAD) is a serious cardiovascular condition characterized by rapid onset and high mortality rates. Currently, no effective drug treatment options are known for AAD. AAD pathogenesis is associated with the phenotypic transformation and abnormal proliferation of vascular smooth muscle cells (VSMCs). However, endogenous factors that contribute to AAD progression remain unclear. We aimed to investigate the role of histone deacetylase 9 (HDAC9) in AAD pathogenesis. HDAC9 expression was considerably increased in human thoracic aortic dissection specimens. Using RNA-sequencing (RNA-seq) and chromatin immunoprecipitation, we demonstrated that HDAC9 transcriptionally inhibited the expression of superoxide dismutase 2 and insulin-like growth factor-binding protein-3, which are critically involved in various signaling pathways. Furthermore, HDAC9 triggered the transformation of VSMCs from a systolic to synthetic phenotype, increasing their proliferation and migration abilities and suppressing their apoptosis. Consistent with these results, in vivo experiments revealed that TMP195, a pharmacological inhibitor of HDAC9, suppressed the formation of the ß-aminopropionitrile-induced AAD phenotype in mice. Our findings indicate that HDAC9 may be a novel endogenous risk factor that promotes the onset of AAD by mediating the phenotypic transformation of VSMCs. Therefore, HDAC9 may serve as a potential therapeutic target for drug-based AAD treatment. Furthermore, TMP195 holds potential as a therapeutic agent for AAD treatment.


Sujet(s)
Anévrysme de l'aorte , , Benzamides , Oxadiazoles , Humains , Souris , Animaux , Muscles lisses vasculaires/anatomopathologie , /traitement médicamenteux , /génétique , Histone deacetylases/génétique , Anévrysme de l'aorte/traitement médicamenteux , Anévrysme de l'aorte/génétique , Anévrysme de l'aorte/anatomopathologie , Phénotype , Myocytes du muscle lisse/anatomopathologie , Cellules cultivées
9.
Cell Signal ; 117: 111080, 2024 05.
Article de Anglais | MEDLINE | ID: mdl-38320624

RÉSUMÉ

Rap1 GTPase-activating protein (Rap1GAP) is an important tumor suppressor. The purpose of this study was to investigate the role of Rap1GAP in myocardial infarction (MI) and its potential mechanism. Left anterior descending coronary artery ligation was performed on cardiac-specific Rap1GAP conditional knockout (Rap1GAP-CKO) mice and control mice with MI. Seven days after MI, Rap1GAP expression in the hearts of control mice peaked, the expression of proapoptotic markers (Bax and cleaved caspase-3) increased, the expression of antiapoptotic factors (Bcl-2) decreased, and the expression of the inflammatory factors IL-6 and TNF-α increased; thus, apoptosis occurred, inflammation, infarct size, and left ventricular dysfunction increased, while the heart changes caused by MI were alleviated in Rap1GAP-CKO mice. Mouse heart tissue was obtained for transcriptome sequencing, and gene set enrichment analysis (GSEA) was used to analyze Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. We found that Rap1GAP was associated with the AMPK and NF-κB signaling pathways and that Rap1GAP inhibited AMPK/SIRT1 and activated the NF-κB signaling pathway in model animals. Similar results were observed in primary rat myocardial cells subjected to oxygen-glucose deprivation (OGD) to induce ischemia and hypoxia. Activating AMPK with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) reversed the damage caused by Rap1GAP overexpression in cardiomyocytes. In addition, the coimmunoprecipitation results showed that exogenous Rap1GAP interacted with AMPK. Rap1GAP was verified to regulate the AMPK SIRT1/NF-κB signaling pathway and exacerbate the damage to myocardial cells caused by ischemia and hypoxia. In conclusion, our results suggest that Rap1GAP promotes MI by modulating the AMPK/SIRT1/NF-κB signaling pathway and that Rap1GAP may be a therapeutic target for MI treatment in the future.


Sujet(s)
Infarctus du myocarde , Facteur de transcription NF-kappa B , Rats , Souris , Animaux , Facteur de transcription NF-kappa B/métabolisme , AMP-Activated Protein Kinases/métabolisme , Sirtuine-1/métabolisme , Transduction du signal , Infarctus du myocarde/métabolisme , Myocytes cardiaques/métabolisme , Apoptose , Hypoxie/métabolisme
10.
ESC Heart Fail ; 11(2): 846-858, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38193606

RÉSUMÉ

AIMS: A therapeutic strategy for chronic heart failure (HF) is to lower resting heart rate (HR). Ivabradine is a well-known HR-lowering agent, but limited prospective data exist regarding its use in Chinese patients. This study aimed to evaluate the effectiveness and safety of ivabradine in Chinese patients with chronic HF. METHODS AND RESULTS: This multicentre, single-arm, prospective, observational study enrolled Chinese patients with chronic HF. The primary outcome was change from baseline in HR at 1 and 6 months, measured by pulse counting. Effectiveness was also evaluated using laboratory tests, the Kansas City Cardiomyopathy Questionnaire (KCCQ) clinical summary score (CSS) and overall summary score (OSS), and New York Heart Association (NYHA) class. Treatment-emergent adverse events (TEAEs) were assessed. A post hoc analysis examined the effectiveness and safety of ivabradine combined with an angiotensin receptor-neprilysin inhibitor (ARNI) or beta-blocker. A total of 1003 patients were enrolled [mean age 54.4 ± 15.0 years, 773 male (77.1%), mean baseline HR 88.5 ± 11.3 b.p.m., mean blood pressure 115.7/74.4 ± 17.2/12.3 mmHg, mean left ventricular ejection fraction 30.9 ± 7.6%, NYHA Classes III and IV in 48.8% and 22.0% of patients, respectively]. HR decreased by a mean of 12.9 and 16.1 b.p.m. after 1 and 6 months, respectively (both P < 0.001). At Month 6, improvements in the KCCQ CSS and OSS of ≥5 points were observed in 72.1% and 74.1% of patients, respectively (both P < 0.001). Left ventricular ejection fraction increased by 12.1 ± 11.6 (P < 0.001), and 66.7% of patients showed improvement in NYHA class (P < 0.001). At Month 6, the overall proportion of patients in NYHA Classes III and IV was reduced to 13.5% and 2.1%, respectively. Serum brain natriuretic peptide (BNP) and N-terminal pro-BNP changed by -331.9 ng/L (-1238.6, -134.0) and -1113.8 ng/L (-2202.0, -297.2), respectively (P < 0.001). HR reductions and improvements in NYHA and KCCQ scores with ivabradine were similar with and without use of ARNIs or beta-blockers. Of 498 TEAEs in 296 patients (29.5%), 73 TEAEs in 55 patients (5.5%) were considered related to ivabradine [most frequent sinus bradycardia (n = 7) and photopsia (n = 7)]. TEAEs were reported in a similar number of patients in ARNI and beta-blocker subgroups (21.9-35.6%). CONCLUSIONS: Ivabradine treatment reduced HR and improved cardiac function and health-related quality of life in Chinese patients with chronic HF. Benefits were seen irrespective of whether or not patients were also taking ARNIs or beta-blockers. Treatment was well tolerated with a similar profile to previous ivabradine studies.


Sujet(s)
Agents cardiovasculaires , Défaillance cardiaque , Troubles de la vision , Adulte , Sujet âgé , Humains , Mâle , Adulte d'âge moyen , Antagonistes bêta-adrénergiques/usage thérapeutique , Benzazépines , Agents cardiovasculaires/usage thérapeutique , Chine , Ivabradine/usage thérapeutique , Études prospectives , Qualité de vie , Débit systolique , Résultat thérapeutique , Fonction ventriculaire gauche , Femelle
11.
Int Immunopharmacol ; 125(Pt B): 111151, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37948859

RÉSUMÉ

Breast cancer is the predominant cancer among women worldwide, and chemotherapeutic agents, such as doxorubicin (DOX), have the potential to significantly prolong survival, albeit at the cost of inducing severe cardiovascular toxicity. Inflammation has emerged as a crucial biological process contributing to the remodeling of cardiovascular toxicity. The role of serum glucocorticoid kinase 1 (SGK1) in various inflammatory diseases has been extensively investigated. Here, we studied the molecular mechanisms underlying the function of SGK1 in DOX-induced cardiotoxicity in HL-1 cardiomyocyte cell lines and in a tumor-bearing mouse model. SGK1 was upregulated in the DOX-induced cardiotoxicity model, accompanied by increased levels of inflammatory factors. Furthermore, inhibition of SGK1 suppresses the phosphorylation of nuclear factor-kappa B (NFκB) in cardiomyocytes, which inhibits the production of inflammatory factors and apoptosis of cardiomyocytes, and has cardioprotective effects. Simultaneously, small interfering RNA targeting SGK1 inhibited the proliferation of breast cancer cells. Conversely, overexpression of SGK1 increases the phosphorylation of NFκB and aggravates myocardial injury. In conclusion, our study demonstrates that SGK1 promotes DOX-induced cardiac inflammation and apoptosis by promoting NFκB activity. Our results indicate that inhibiting SGK1 might be an effective treatment strategy that can provide both tumor-killing and cardioprotective functions. Further in vivo research is needed to fully elucidate the effects and mechanisms of combination therapy with SGK1 inhibitors and DOX in breast cancer treatment.


Sujet(s)
Tumeurs du sein , Cardiotoxicité , Femelle , Humains , Animaux , Souris , Glucocorticoïdes , Doxorubicine , Tumeurs du sein/traitement médicamenteux , Inflammation , Facteur de transcription NF-kappa B
12.
Int Immunopharmacol ; 125(Pt A): 110978, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37925944

RÉSUMÉ

Tamoxifen (TAM) is an effective anticancer drug for breast and ovarian cancer. However, increased risk of cardiotoxicity is a long-term clinical problem associated with TAM, while the underlying mechanisms remain unclear. Here, we performed experiments in cardiomyocytes and tumor-bearing or nontumor-bearing mice, and demonstrated that TAM induced cardiac injury via the IL-6/p-STAT3/PGC-1α/IL-6 feedback loop, which is responsible for reactive oxygen species (ROS) accumulation. Compared with non-tumor bearing mice, tumor-bearing mice showed stronger cardiac toxicity after TAM injection, although there was no significant difference. In vitro experiments demonstrated STAT3 phosphorylation inhibitor can increase PGC-1α expression and protect cardiomyocyte via decreasing ROS. Since tumor has higher STAT3 phosphorylation and IL-6 expression level, our research results indicated combining TAM and STAT3 inhibitor might be an effective treatment strategy which can provide both tumor killing and cardioprotective function. Further in vivo research is needed to fully elucidate the effect and mechanisms of the combination therapy of TAM and STAT3 inhibitor.


Sujet(s)
Interleukine-6 , Tumeurs , Souris , Animaux , Espèces réactives de l'oxygène/métabolisme , Interleukine-6/métabolisme , Tamoxifène , Myocytes cardiaques/métabolisme , Cardiotoxicité/métabolisme , Tumeurs/métabolisme
13.
Cardiorenal Med ; 13(1): 344-353, 2023.
Article de Anglais | MEDLINE | ID: mdl-37839394

RÉSUMÉ

INTRODUCTION: Due to the cardiotoxicity of cancer treatment and traditional risk factors for cardiovascular disease (CVD) such as obesity, diabetes, dyslipidemia, and hypertension, cancer patients are at higher risk of developing CVD. However, limited research exists on the correlation between chronic kidney disease (CKD) and CVD risk in cancer patients. METHODS: This cross-sectional study selected cancer patients aged ≥20 years from the National Health and Nutrition Examination Survey (NHANES) conducted from 2015 to 2020. Multivariable logistic regression was used to assess the association between CKD and CVD in cancer patients. Additionally, subgroup analyses were conducted to investigate the association among different groups of cancer patients. RESULTS: We included 1,700 adult cancer patients (52.53% were females). After multivariable adjustment for covariates including traditional CVD factors, CKD was significantly associated with CVD, with an odds ratio (95% confidence interval) and p value of 1.61 (1.18, 2.19) and 0.004. Subgroup analyses after multivariable adjustment showed a significant correlation between CKD and increased CVD risk in the following cancer patients: age ≥60 years, males, white ethnicity, and individuals with or without traditional CVD factors (obesity, diabetes, dyslipidemia, and hypertension). CONCLUSIONS: CKD remains a significant factor in the higher risk of CVD among adult cancer patients in the United States, even after adjustment for traditional CVD risk factors. Therefore, to reduce the risk of CVD in cancer patients, it is important to treat CKD as a non-traditional risk factor for CVD and actively manage it.


Sujet(s)
Maladies cardiovasculaires , Diabète , Dyslipidémies , Hypertension artérielle , Tumeurs , Insuffisance rénale chronique , Adulte , Mâle , Femelle , Humains , États-Unis/épidémiologie , Maladies cardiovasculaires/complications , Maladies cardiovasculaires/épidémiologie , Enquêtes nutritionnelles , Études transversales , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/épidémiologie , Hypertension artérielle/complications , Hypertension artérielle/épidémiologie , Obésité/complications , Obésité/épidémiologie , Dyslipidémies/complications , Dyslipidémies/épidémiologie , Tumeurs/complications , Tumeurs/épidémiologie
14.
Sci Rep ; 13(1): 8324, 2023 05 23.
Article de Anglais | MEDLINE | ID: mdl-37221286

RÉSUMÉ

Radiation pneumonitis (RP) affects both patients and physicians during radiation therapy for lung cancer. To date, there are no effective drugs for improving the clinical outcomes of RP. The activation of angiotensin-converting enzyme 2 (ACE2) improves experimental acute lung injury caused by severe acute respiratory syndrome coronavirus, acid inhalation, and sepsis. However, the effects and underlying mechanisms of ACE2 in RP remain unclear. Therefore, this study aimed to investigate the effects of angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers on RP and ACE2/angiotensin-(1-7)/Mas receptor pathway activation. We found that radiotherapy decreased the expression of ACE2 and that overexpression of ACE2 alleviated lung injury in an RP mouse model. Moreover, captopril and valsartan restored ACE2 activation; attenuated P38, ERK, and p65 phosphorylation; and effectively mitigated RP in the mouse model. Further systematic retrospective analysis illustrated that the incidence of RP in patients using renin-angiotensin system inhibitors (RASis) was lower than that in patients not using RASis (18.2% vs. 35.8% at 3 months, p = 0.0497). In conclusion, the current findings demonstrate that ACE2 plays a critical role in RP and suggest that RASis may be useful potential therapeutic drugs for RP.


Sujet(s)
Lésion pulmonaire aigüe , Poumon radique , Animaux , Souris , Facteur de transcription NF-kappa B , Peptidyl-Dipeptidase A , Angiotensin-converting enzyme 2 , Système rénine-angiotensine , Études rétrospectives , Antihypertenseurs , Antienzymes
15.
Mol Med Rep ; 27(6)2023 06.
Article de Anglais | MEDLINE | ID: mdl-37203392

RÉSUMÉ

Acute myocardial infarction (AMI) is a serious disease which threatens public health. Exosomes (exos) contain certain genetic information and are important communication vehicles between cells. In the present study, different exosomal microRNAs (miRs), which exhibit a notable association between expression levels in plasma and AMI were assessed to support the development of new diagnostic and clinical assessment markers of patients with AMI. In total, 93 individuals, including 31 healthy controls and 62 patients with AMI, were recruited for the present study. Data on age, blood pressure, glucose levels, lipid levels and coronary angiography images were collected from the enrolled individuals, and plasma samples were collected. Plasma exos were extracted and verified using ultracentrifugation, transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blotting (WB). Exo­miR­4516 and exo­miR­203 in plasma exos were identified by exosomal miRNA sequencing analysis, reverse transcription­quantitative PCR was performed to detect the levels of exo­miR­4516 and exo­miR­203 in plasma exos, and ELISA was performed to detect the levels of secretory frizzled­related protein 1 (SFRP1) in samples. The correlation analysis between exo­miR­4516, exo­miR­203 and SFRP1 in plasma exos and AMI was presented as receiver operating characteristic curves (ROCs) of the SYNTAX score, cardiac troponin I (cTnI), low­density lipoprotein (LDL) and each indicator separately. Kyoto Encyclopedia of Genes and Genomes enrichment analysis was performed to predict relevant enrichment pathways. Exos were successfully isolated from plasma by ultracentrifugation, which was confirmed by TEM, NTA and WB. Exo­miR­4516, exo­miR­203 and SFRP1 levels in plasma were significantly higher in the AMI group compared with the healthy control group. ROCs demonstrated that exo­miR­4516, exo­miR­203 and SFRP1 levels had a high diagnostic efficiency in predicting AMI. Exo­miR­4516 was positively correlated with SYNTAX score, and plasma SFRP1 was positively correlated with plasma cTnI and LDL. In conclusion, the data demonstrated that exo­miR­4516, exo­miR­203 and SFRP1 levels could be used in combination to diagnose and assess the severity of AMI. The present study was retrospectively registered (TRN, NCT02123004).


Sujet(s)
microARN , Infarctus du myocarde , Humains , microARN/génétique , Infarctus du myocarde/diagnostic , Infarctus du myocarde/génétique , Marqueurs biologiques , Protéines et peptides de signalisation intracellulaire , Troponine I/génétique , Troponine I/métabolisme , Protéines membranaires , Protéines et peptides de signalisation intercellulaire
16.
J Cardiovasc Pharmacol ; 82(2): 128-137, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37155368

RÉSUMÉ

ABSTRACT: Six-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 2 (PFKFB2) is a key regulator of glycolytic enzyme. This study identified whether PFKFB2 can regulate myocardial ferroptosis in ischemia/reperfusion (I/R) injury. Mice myocardial (I/R) injury and H9c2 cells oxygen-glucose deprivation/reperfusion (OGD/R) models were established. PFKFB2 expression was enhanced in I/R mice and OGD/R H9c2 cells. Overexpression of PFKFB2 improves heart function in I/R mice. Overexpression of PFKFB2 inhibits I/R and OGD/R-induced ferroptosis in mice and H9c2 cells. Mechanistically, overexpression of PFKFB2 activates the adenosine monophosphate-activated protein kinase (AMPK). AMPK inhibitor compound C reverses effect of PFKFB2 overexpression in reducing ferroptosis under OGD/R treatment. In conclusion, PFKFB2 protects hearts against I/R-induced ferroptosis through activation of the AMPK signaling pathway.


Sujet(s)
Ferroptose , Lésion de reperfusion myocardique , Lésion d'ischémie-reperfusion , Souris , Animaux , Lésion de reperfusion myocardique/métabolisme , AMP/pharmacologie , AMP-Activated Protein Kinases/métabolisme , Transduction du signal , Lésion d'ischémie-reperfusion/métabolisme , Apoptose , Glucose/métabolisme
17.
Arterioscler Thromb Vasc Biol ; 43(5): 755-773, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36951060

RÉSUMÉ

BACKGROUND: Atherosclerosis is a chronic inflammatory disease, in which macrophages determine the progression of atherosclerotic plaques. However, no studies have investigated how METTL3 (methyltransferase like 3) in macrophages affects atherosclerotic plaque formation in vivo. Additionally, whether Braf mRNA is modified by METTL3-dependent N6-methyladenosine (m6A) methylation remains unknown. METHODS: We analyzed single-cell sequencing data of atherosclerotic plaques in mice fed with a high fat diet for different periods. Mettl3fl/fl Lyz2cre Apoe-/- mice and littermate control Mettl3fl/fl Apoe-/- mice were generated and fed high fat diet for 14 weeks. In vitro, we stimulated peritoneal macrophages with ox-LDL (oxidized low-density lipoprotein) and tested the mRNA and protein expression levels of inflammatory factors and molecules regulating ERK (extracellular signal-regulated kinase) phosphorylation. To find METTL3 targets in macrophages, we performed m6A-methylated RNA immunoprecipitation sequencing and m6A-methylated RNA immunoprecipitation-qPCR. Further, point mutation experiments were used to explore m6A-methylated adenine. Using RNA immunoprecipitation assay, we explored m6A methylation-writing protein bound to Braf mRNA. RESULTS: In vivo, METTL3 expression in macrophages increased with the progression of atherosclerosis. Myeloid cell-specific METTL3 deletion negatively regulated atherosclerosis progression and the inflammatory response. In vitro, METTL3 knockdown or knockout in macrophages attenuated ox-LDL-mediated ERK phosphorylation rather than JNK (c-Jun N-terminal kinase) and p38 phosphorylation and reduced the level of inflammatory factors by affecting BRAF protein expression. The negative regulation of inflammation response caused by METTL3 knockout was rescued by overexpression of BRAF. In mechanism, METTL3 targeted adenine (39725126 in chromosome 6) on the Braf mRNA. Then, YTHDF1 could bind to m6A-methylated Braf mRNA and promoted its translation. CONCLUSIONS: Myeloid cell-specific Mettl3 deficiency suppressed hyperlipidemia-induced atherosclerotic plaque formation and attenuated atherosclerotic inflammation. We identified Braf mRNA as a novel target of METTL3 in the activation of the ox-LDL-induced ERK pathway and inflammatory response in macrophages. METTL3 may represent a potential target for the treatment of atherosclerosis.


Sujet(s)
Athérosclérose , Plaque d'athérosclérose , Souris , Animaux , Plaque d'athérosclérose/métabolisme , Protéines proto-oncogènes B-raf/génétique , ARN messager/génétique , ARN messager/métabolisme , Methyltransferases/génétique , Methyltransferases/métabolisme , Macrophages/métabolisme , Inflammation/génétique , Inflammation/prévention et contrôle , Inflammation/métabolisme , Athérosclérose/génétique , Athérosclérose/prévention et contrôle , Athérosclérose/métabolisme , Apolipoprotéines E/métabolisme
18.
Exp Ther Med ; 25(1): 56, 2023 Jan.
Article de Anglais | MEDLINE | ID: mdl-36588807

RÉSUMÉ

Atrial fibrillation (AF) is the most common type of supraventricular tachyarrhythmia. Nifekalant is a new class III antiarrhythmic drug approved for the treatment of ventricular tachyarrhythmias, but its effectiveness in converting AF to sinus rhythm remains unclear. The present analysis aimed to investigate the effect of nifekalant in the conversion of AF. PubMed, Cochrane Library and China National Knowledge Infrastructure databases were systematically used to search relevant studies published between 1999 (data at which the drug was first approved for marketing in Japan) and 2022. Randomized clinical trials, prospective studies and retrospective studies on the use of nifekalant for AF were screened. The study metrics included the success rate of the conversion of AF, the mean time to conversion, the success rate of 12 months after a single AF catheter ablation procedure and the incidence of adverse events. The eligible studies screened included six randomized clinical trials, three prospective studies and three retrospective studies, totalling 12 studies with 1,162 patients. The risk ratio (RR) for successful conversion in the nifekalant and control groups was 1.95 [95% confidence interval (CI), 1.23-3.08; P=0.005] and the mean difference for the mean time to conversion was -1.73 [95% CI, -2.69-(-0.77); P=0.0004]. Statistically significant differences were observed between nifekalant and control groups. Subgroup analysis revealed a statistically significant difference in the success rate of conversion following catheter ablation in the nifekalant group compared with the amiodarone group and the RR value was 1.95 (95% CI, 1.37-2.77; P=0.0002). Statistically significant difference was observed compared with the electrical cardioversion group and the RR value was 0.90 (95% CI, 0.84-0.98; P=0.01). However, the combined RR values for the two groups were 1.18 (95% CI, 0.85-1.65; P<0.0002). The RR value for adverse events was 0.85 (95% CI, 0.51-1.43; P=0.55), with no statistically significant differences between nifekalant and control groups. In conclusion, the results demonstrated that the success rate and time to conversion in the nifekalant group were improved compared with those in the control group, particularly after catheter ablation, and the conversion effect with nifekalant was significantly improved compared with that in the control group.

19.
Environ Sci Technol ; 57(4): 1701-1711, 2023 01 31.
Article de Anglais | MEDLINE | ID: mdl-36668989

RÉSUMÉ

Associations between ultrafine particles (UFPs) and hourly onset of acute myocardial infarction (AMI) have rarely been investigated. We aimed to evaluate the impacts of UFPs on AMI onset and the lag patterns. A time-stratified case-crossover study was performed among 20,867 AMI patients from 46 hospitals in Shanghai, China, between January 2015 and December 2020. Hourly data of AMI onset and number concentrations of nanoparticles of multiple size ranges below 0.10 µm (0.01-0.10, UFP/PNC0.01-0.10; 0.01-0.03, PNC0.01-0.03; 0.03-0.05, PNC0.03-0.05; and 0.05-0.10 µm, PNC0.05-0.10) were collected. Conditional logistic regressions were applied. Transient exposures to these nanoparticles were significantly associated with AMI onset, with almost linear exposure-response curves. These associations occurred immediately after exposure, lasted for approximately 6 h, and attenuated to be null thereafter. Each interquartile range increase in concentrations of total UFPs, PNC0.01-0.03, PNC0.03-0.05, and PNC0.05-0.10 during the preceding 0-6 h was associated with increments of 3.29, 2.08, 2.47, and 2.93% in AMI onset risk, respectively. The associations were stronger during warm season and at high temperatures and were robust after adjusting for criteria air pollutants. Our findings provide novel evidence that hourly UFP exposure is associated with immediate increase in AMI onset risk.


Sujet(s)
Polluants atmosphériques , Pollution de l'air , Infarctus du myocarde , Humains , Matière particulaire/analyse , Études croisées , Exposition environnementale/analyse , Chine/épidémiologie , Polluants atmosphériques/analyse , Infarctus du myocarde/épidémiologie , Pollution de l'air/analyse , Taille de particule
20.
Cell Death Differ ; 30(4): 966-978, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36681779

RÉSUMÉ

Innate immunity is the first line to defend against pathogenic microorganisms, and Toll-like receptor (TLR)-mediated inflammatory responses are an essential component of innate immunity. However, the regulatory mechanisms of TLRs in innate immunity remain unperfected. We found that the expression of E3 ligase Ring finger protein 99 (RNF99) decreased significantly in peripheral blood monocytes from patients infected with Gram negative bacteria (G-) and macrophages stimulated by TLRs ligands, indicating the role of RNF99. We also demonstrated for the first time, the protective role of RNF99 against LPS-induced septic shock and dextran sodium sulfate (DSS)-induced colitis using RNF99 knockout mice (RNF99-/-) and bone marrow-transplanted mice. In vitro experiments revealed that RNF99 deficiency significantly promoted TLR-mediated inflammatory cytokine expression and activated the NF-κB and MAPK pathways in macrophages. Mechanistically, in both macrophages and HEK293 cell line with TLR4 stably transfection, RNF99 interacted with and degraded TAK1-binding protein (TAB) 2, a regulatory protein of the kinase TAK1, via the lysine (K)48-linked ubiquitin-proteasomal pathway on lysine 611 of TAB2, which further regulated the TLR-mediated inflammatory response. Overall, these findings indicated the physiological significance of RNF99 in macrophages in regulating TLR-mediated inflammatory reactions. It provided new insight into TLRs signal transduction, and offered a novel approach for preventing bacterial infections, endotoxin shock, and other inflammatory ills.


Sujet(s)
Lysine , Ubiquitin-protein ligases , Humains , Animaux , Souris , Ubiquitin-protein ligases/métabolisme , Lysine/métabolisme , Cellules HEK293 , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Récepteurs de type Toll/métabolisme , Ubiquitination , Facteur de transcription NF-kappa B/métabolisme , Immunité innée , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...