Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 83
Filtrer
1.
Med Sci Monit ; 27: e932404, 2021 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-34493698

RÉSUMÉ

BACKGROUND Studies in ApoE knockout mice have shown that pseudolaric acid B (PB) can act as an immunomodulatory drug and attenuate atherosclerosis progression by modulating monocyte/macrophage phenotypes. Our previous study demonstrated that high salt intake could shift the phenotype of monocytes/macrophages to an inflammatory phenotype, and that this shift was related to hypertension and hypertensive left ventricular (LV) remodeling. However, no comprehensive assessment of the effects of PB on hypertensive LV remodeling has been conducted. MATERIAL AND METHODS In this study, RAW264.7 macrophages cultured with different concentrations of NaCl were used to investigate the modulating effects of PB on macrophage phenotype. Furthermore, N-nitro-L-arginine methyl ester hypertensive mice were used to investigate the modulating effects of PB on monocyte phenotype. LV remodeling was investigated by echocardiography. LV morphologic staining (for cardiomyocyte hypertrophy and collagen deposition) was performed at the time of sacrifice. RESULTS The results showed that PB significantly improved the viability of RAW264.7 cells, suppressed their phagocytic and migration abilities, and inhibited their phenotypic shift to M1 macrophages. In addition, the blood pressure of PB-treated mice was significantly decreased relative to that of control mice. Furthermore, after PB treatment, the percentage of Ly6Chi monocytes was significantly decreased while that of Ly6Clo monocytes was apparently increased. Moreover, PB preserved LV function and alleviated myocardial fibrosis and cardiomyocyte hypertrophy as measured at the end of the experimental period. The transfer of monocytes from PB-treated mice to hypertensive mice achieved the same effects. CONCLUSIONS Together, these findings indicate that PB exerts its protective effects on hypertensive LV remodeling by modulating monocyte/macrophage phenotypes and warrants further investigation.


Sujet(s)
Diterpènes/usage thérapeutique , Ventricules cardiaques/effets des médicaments et des substances chimiques , Hypertension artérielle/traitement médicamenteux , Macrophages/effets des médicaments et des substances chimiques , Monocytes/effets des médicaments et des substances chimiques , Chlorure de sodium/effets indésirables , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Animaux , Marqueurs biologiques/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/usage thérapeutique , Échocardiographie , Hypertension artérielle/induit chimiquement , Hypertension artérielle/immunologie , Hypertension artérielle/physiopathologie , Souris , Souris de lignée C57BL , Phagocytose/effets des médicaments et des substances chimiques , Phénotype , Cellules RAW 264.7 , Remodelage ventriculaire/immunologie
2.
Eur J Pharmacol ; 910: 174470, 2021 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-34478691

RÉSUMÉ

Myocardial fibrosis in post-myocardial infarction is a self-healing process of the myocardium, making ventricular remodelling difficult to reverse and develop continuously. Fibroblast growth factor 21 (FGF21) plays an essential role in cardiovascular and metabolic diseases. However, the effect and mechanism of FGF21 action on cardiac inflammation and fibrosis caused by myocardial injury have rarely been reported. Adult male Sprague-Dawley rats administered with or without recombinant human basic FGF21 (rhbFGF21) were assessed using echocardiography and haematoxylin-eosin and Masson's trichrome staining to determine the cardiac function and cardiac inflammation and fibrosis levels. FGF21 might improve cardiac remodelling, as characterised by a decrease in the expression of a series of inflammatory and fibrosis-related factors. Moreover, when FGF receptors (FGFRs) were blocked, the effects of FGF21 disappeared. Mechanistically, we found that oxidative stress induced the downregulation of early growth response protein 1 (EGR1), which contributed to inflammatory factors and fibrosis reduction in cardiomyocytes treated with H2O2. Collectively, FGF21 effectively suppressed the inflammation and fibrosis in post-infarcted hearts by regulating FGFR-EGR1.


Sujet(s)
Facteur de transcription EGR-1/antagonistes et inhibiteurs , Facteurs de croissance fibroblastique/pharmacologie , Infarctus du myocarde/traitement médicamenteux , Myocarde/anatomopathologie , Animaux , Cellules cultivées , Modèles animaux de maladie humaine , Facteur de transcription EGR-1/métabolisme , Facteurs de croissance fibroblastique/usage thérapeutique , Fibrose , Coeur/effets des médicaments et des substances chimiques , Humains , Inflammation/traitement médicamenteux , Inflammation/immunologie , Inflammation/anatomopathologie , Mâle , Infarctus du myocarde/complications , Infarctus du myocarde/immunologie , Infarctus du myocarde/anatomopathologie , Myocarde/immunologie , Myocytes cardiaques , Culture de cellules primaires , Rats , Protéines recombinantes/pharmacologie , Protéines recombinantes/usage thérapeutique , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Remodelage ventriculaire/immunologie
3.
JCI Insight ; 6(13)2021 07 08.
Article de Anglais | MEDLINE | ID: mdl-34236048

RÉSUMÉ

BackgroundImmunomodulatory therapy may help prevent heart failure (HF). Data on immune cells and myocardial remodeling in older adults with cardiovascular risk factors are limited.MethodsIn the Multi-Ethnic Study of Atherosclerosis cohort, 869 adults had 19 peripheral immune cell subsets measured and underwent cardiac MRI during the baseline exam, of which 321 had assessment of left ventricular global circumferential strain (LV-GCS). We used linear regression with adjustment for demographics, cardiovascular risk factors, and cytomegalovirus serostatus to evaluate the cross-sectional association of immune cell subsets with left ventricular mass index (LVMI) and LV-GCS.ResultsThe average age of the cohort was 61.6 ± 10.0 years and 53% were women. Higher proportions of γ/δ T cells were associated with lower absolute (worse) LV-GCS (-0.105% [95% CI -0.164%, -0.046%] per 1 SD higher proportion of γ/δ T cells, P = 0.0006). This association remained significant after Bonferroni's correction. Higher proportions of classical monocytes were associated with worse absolute LV-GCS (-0.04% [95% CI -0.07%, 0.00%] per 1 SD higher proportion of classical monocytes, P = 0.04). This did not meet significance after Bonferroni's correction. There were no other significant associations with LV-GCS or LVMI.ConclusionPathways associated with γ/δ T cells may be potential targets for immunomodulatory therapy targeted at HF prevention in populations at risk.FundingContracts 75N92020D00001, HHSN268201500003I, N01-HC-95159, 75N92020D00005, N01-HC-95160, 75N92020D00002, N01-HC-95161, 75N92020D00003, N01-HC-95162, 75N92020D00006, N01-HC-95163, 75N92020D00004, N01-HC-95164, 75N92020D00007, N01-HC-95165, N01-HC-95166, N01-HC-95167, N01-HC-95168, and N01-HC-95169 and grant R01 HL98077 from the National Heart, Lung, and Blood Institute/NIH and grants KL2TR001424, UL1-TR-000040, UL1-TR-001079, and UL1-TR-001420 from the National Center for Advancing Translational Sciences/NIH.


Sujet(s)
Athérosclérose , Défaillance cardiaque , Ventricules cardiaques , Monocytes/immunologie , Sous-populations de lymphocytes T/immunologie , Dysfonction ventriculaire gauche , Athérosclérose/sang , Athérosclérose/physiopathologie , Études de cohortes , Femelle , Facteurs de risque de maladie cardiaque , Défaillance cardiaque/immunologie , Défaillance cardiaque/prévention et contrôle , Ventricules cardiaques/imagerie diagnostique , Ventricules cardiaques/anatomopathologie , Humains , Immunomodulation , IRM dynamique/méthodes , IRM dynamique/statistiques et données numériques , Mâle , Adulte d'âge moyen , Taille d'organe , Facteurs de risque , Dysfonction ventriculaire gauche/diagnostic , Dysfonction ventriculaire gauche/immunologie , Remodelage ventriculaire/immunologie
5.
Front Immunol ; 12: 605857, 2021.
Article de Anglais | MEDLINE | ID: mdl-34046028

RÉSUMÉ

Aims: Latent cytomegalovirus (CMV) infection is associated with adverse cardiovascular outcomes. Virus-specific CX3CR1+ effector memory T-cells may be instrumental in this process due to their pro-inflammatory properties. We investigated the role of CX3CR1 (fractalkine receptor) in CMV-related lymphocyte kinetics and cardiac remodeling in patients with ST-elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (pPCI). Methods and Results: We retrospectively analysed lymphocyte count, troponin, and survival in 4874 STEMI/pPCI patients, evaluated lymphocyte kinetics during reperfusion in a prospective cohort, and obtained sequential cardiac MRI (cMRI) to assess remodeling. Pre-reperfusion lymphopenia independently predicted mortality at 7.5 years. Prior to reperfusion, CCR7+ T-lymphocytes appeared to be depleted. After reperfusion, T-lymphocytes expressing CX3CR1 were depleted predominantly in CMV-seropositive patients. During ischaemia/reperfusion, a drop in CX3CR1+ T-lymphocytes was significantly linked with microvascular obstruction in CMV+ patients, suggesting increased fractalkine-receptor interaction. At 12 weeks, CMV+ patients displayed adverse LV remodeling. Conclusion: We show that lymphopenia occurs before and after reperfusion in STEMI by different mechanisms and predicts long-term outcome. In CMV+ patients, increased fractalkine induction and sequestration of CX3CR1+ T-cells may contribute to adverse remodeling, suggesting a pro-inflammatory pathomechanism which presents a novel therapeutic target.


Sujet(s)
Récepteur-1 de la chimiokine CX3C/génétique , Infections à cytomégalovirus/complications , Lymphocytes/métabolisme , Infarctus du myocarde/complications , Infarctus du myocarde/métabolisme , Remodelage ventriculaire , Sujet âgé , Marqueurs biologiques , Récepteur-1 de la chimiokine CX3C/métabolisme , Cytomegalovirus , Infections à cytomégalovirus/virologie , Femelle , Tests de la fonction cardiaque , Humains , Immunophénotypage , Lymphocytes/immunologie , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Infarctus du myocarde/diagnostic , Infarctus du myocarde/mortalité , Récepteurs CCR7/métabolisme , Remodelage ventriculaire/génétique , Remodelage ventriculaire/immunologie
6.
Front Immunol ; 12: 664457, 2021.
Article de Anglais | MEDLINE | ID: mdl-33868315

RÉSUMÉ

The burden of heart failure (HF), developing after myocardial infarction MI, still represents a major issue in clinical practice. Failure of appropriate resolution of inflammation during post-myocardial injury is associated with unsuccessful left ventricular remodeling and underlies HF pathogenesis. Cells of the immune system have been shown to mediate both protective and damaging effects in heart remodeling. This ambiguity of the role of the immune system and inconsistent results of the recent clinical trials question the benefits of anti-inflammatory therapies during acute MI. The present review will summarize knowledge of the roles that different cells of the immune system play in the process of post-infarct cardiac healing. Data on the phenotype, active molecules and functions of the immune cells, based on the results of both experimental and clinical studies, will be provided. For some cellular subsets, such as macrophages, neutrophils, dendritic cells and lymphocytes, an anti-inflammatory activity has been attributed to the specific subpopulations. Activity of other cells, such as eosinophils, mast cells, natural killer (NK) cells and NKT cells has been shown to be highly dependent of the signals created by micro-environment. Also, new approaches for classification of cellular phenotypes based on the single-cell RNA sequencing allow better understanding of the phenotype of the cells involved in resolution of inflammation. Possible perspectives of immune-mediated therapy for AMI patients are discussed in the conclusion. We also outline unresolved questions that need to be solved in order to implement the current knowledge on the role of the immune cells in post-MI tissue repair into practice.


Sujet(s)
Prédisposition aux maladies , Défaillance cardiaque/étiologie , Défaillance cardiaque/anatomopathologie , Système immunitaire/immunologie , Myocarde/immunologie , Myocarde/anatomopathologie , Remodelage ventriculaire/immunologie , Animaux , Marqueurs biologiques , Cellules dendritiques , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/métabolisme , Humains , Système immunitaire/métabolisme , Inflammation/étiologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Médiateurs de l'inflammation/métabolisme , Métabolisme lipidique , Lymphocytes/immunologie , Lymphocytes/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Mastocytes , Infarctus du myocarde/étiologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/anatomopathologie , Myocarde/métabolisme , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Cicatrisation de plaie
7.
Front Immunol ; 12: 790511, 2021.
Article de Anglais | MEDLINE | ID: mdl-34992607

RÉSUMÉ

Cardiac fibrosis, a pathological condition due to excessive extracellular matrix (ECM) deposition in the myocardium, is associated with nearly all forms of heart disease. The processes and mechanisms that regulate cardiac fibrosis are not fully understood. In response to cardiac injury, macrophages undergo marked phenotypic and functional changes and act as crucial regulators of myocardial fibrotic remodeling. Here we show that the mitogen-activated protein kinase (MAPK) phosphatase-5 (MKP-5) in macrophages is involved in pressure overload-induced cardiac fibrosis. Cardiac pressure overload resulting from transverse aortic constriction (TAC) leads to the upregulation of Mkp-5 gene expression in the heart. In mice lacking MKP-5, p38 MAPK and JNK were hyperactivated in the heart, and TAC-induced cardiac hypertrophy and myocardial fibrosis were attenuated. MKP-5 deficiency upregulated the expression of the ECM-degrading matrix metalloproteinase-9 (Mmp-9) in the Ly6Clow (M2-type) cardiac macrophage subset. Consistent with in vivo findings, MKP-5 deficiency promoted MMP-9 expression and activity of pro-fibrotic macrophages in response to IL-4 stimulation. Furthermore, using pharmacological inhibitors against p38 MAPK, JNK, and ERK, we demonstrated that MKP-5 suppresses MMP-9 expression through a combined effect of p38 MAPK/JNK/ERK, which subsequently contributes to the inhibition of ECM-degrading activity. Taken together, our study indicates that pressure overload induces MKP-5 expression and facilitates cardiac hypertrophy and fibrosis. MKP-5 deficiency attenuates cardiac fibrosis through MAPK-mediated regulation of MMP-9 expression in Ly6Clow cardiac macrophages.


Sujet(s)
Cardiomégalie/immunologie , Dual-specificity phosphatases/déficit , Défaillance cardiaque/immunologie , Système de signalisation des MAP kinases/immunologie , Myocarde/anatomopathologie , Animaux , Pression sanguine , Cardiomégalie/diagnostic , Cardiomégalie/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Dual-specificity phosphatases/génétique , Échocardiographie , Fibrose , Coeur/imagerie diagnostique , Défaillance cardiaque/anatomopathologie , Humains , Interleukine-4/immunologie , Macrophages/immunologie , Macrophages/métabolisme , Mâle , Matrix metalloproteinase 9/métabolisme , Souris , Souris knockout , Phosphorylation/immunologie , Culture de cellules primaires , Remodelage ventriculaire/immunologie
8.
J Mol Med (Berl) ; 99(1): 33-46, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33130927

RÉSUMÉ

Macrophages are integral components of the mammalian heart that show extensive expansion in response to various internal or external stimuli. After the onset of sustained pressure overload (PO), the accumulation of cardiac macrophages through local macrophage proliferation and monocyte migration has profound effects on the transition to cardiac hypertrophy and remodeling. In this review, we describe the heterogeneity and diversity of cardiac macrophages and summarize the current understanding of the important roles of macrophages in PO-induced cardiac remodeling. In addition, the possible mechanisms involved in macrophage modulation are also described. Finally, considering the significant effects of cardiac macrophages, we highlight their emerging role as therapeutic targets for alleviating pathological cardiac remodeling after PO.


Sujet(s)
Macrophages/immunologie , Myocarde/immunologie , Remodelage ventriculaire/immunologie , Animaux , Humains , Pression ventriculaire
9.
Oxid Med Cell Longev ; 2020: 5041791, 2020.
Article de Anglais | MEDLINE | ID: mdl-32765807

RÉSUMÉ

The objective of this study was to analyze the impact of different modalities and intensities of exercise training on cardiac remodeling started early after experimental myocardial infarction (MI). Male Wistar rats, weighing 200-250 g, were subjected to experimental MI. After 5 days, the animals were allocated into three experimental groups and observed for three months: S (sedentary control animals), C (animals subjected to continuous low-intensity training), and HIT (animals subjected to high-intensity interval training). Low-intensity exercise training was performed at a treadmill speed corresponding to 40% VO2 max, which was kept unchanged throughout the entire session (i.e., continuous low-intensity training). High-intensity interval training was performed in such a way that rats run during 3 min at 60% VO2 max, followed by 4-minute intervals at 85% VO2 max (i.e., high-intensity interval training). After the follow-up period, we studied hypertrophy and ventricular geometry, functional alterations in vivo and in vitro, oxidative stress, apoptosis, and cardiac energetic metabolism. Our data showed that both high-intensity interval and continuous low-intensity modalities improved cardiac energetic metabolism variables in comparison with sedentary infarcted animals. In addition, high-intensity interval training decreased cardiac oxidative stress, associated with improved diastolic function. On the other hand, the continuous low-intensity group showed impairment of cardiac function. Therefore, altogether, our data suggest that high-intensity interval training could be the best modality for early physical exercise after MI and should be better studied in this clinical scenario.


Sujet(s)
Infarctus du myocarde/thérapie , Conditionnement physique d'animal/méthodes , Remodelage ventriculaire/immunologie , Animaux , Mâle , Infarctus du myocarde/physiopathologie , Rats , Rat Wistar
10.
Int Immunopharmacol ; 80: 106116, 2020 Mar.
Article de Anglais | MEDLINE | ID: mdl-31978804

RÉSUMÉ

The myocardial inflammatory response is a consequence of myocardial infarction (MI), which may deteriorate cardiac remodeling and lead to dysfunction in the heart post-MI. Dectin-1 is a c-type lectin, which has been shown to regulate innate immune responses to pathogens. However, the role of Dectin-1 in the heart diseases remains largely unknown. In this study, we aimed to investigate the effects of Dectin-1 on cardiac remodeling post-MI. We found that cardiac Dectin-1 mRNA and protein expressions were significantly elevated in C57BL/6 mice after MI. In vitro, hypoxia induced cardiomyocyte injury in parallel with increased Dectin-1 protein expression. Knockdown of Dectin-1 remarkably attenuated cardiomyocyte death under hypoxia and lipopolysaccharide (LPS) stimulation. In vivo administration of adeno-associated virus serotype 9 mediated silencing of Dectin-1, which significantly decreased cardiac fibrosis, dilatation, and improved cardiac function in the mice post-MI. At the molecular level, downregulation of Dectin-1 dramatically suppressed up-regulation of nuclear factor-κB (NF-κB), nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), and the inflammatory genes involved in fibrogenesis and cardiac remodeling after MI. Furthermore, treatment with BAY11-7082, an inhibitor of NF-κB, repressed the activation of NF-κB, and attenuated LPS induced elevation of NLRP3 and cell death in cardiomyocytes. Collectively, upregulation of Dectin-1 in cardiomyocytes post-MI contributes to cardiac remodeling and cardiac dysfunction at least partially by activating NF-κB and NLRP3. This study identified Dectin-1 as a promising therapeutic target for ischemic heart disease.


Sujet(s)
Lectines de type C/métabolisme , Infarctus du myocarde/immunologie , Transduction du signal/immunologie , Remodelage ventriculaire/immunologie , Animaux , Modèles animaux de maladie humaine , Régulation négative , Femelle , Techniques de knock-down de gènes , Humains , Lectines de type C/génétique , Lipopolysaccharides/immunologie , Mâle , Souris , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Myocarde/cytologie , Myocarde/immunologie , Myocarde/anatomopathologie , Myocytes cardiaques , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Facteur de transcription NF-kappa B/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Nitriles/pharmacologie , Nitriles/usage thérapeutique , Culture de cellules primaires , Petit ARN interférent/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Sulfones/pharmacologie , Sulfones/usage thérapeutique , Régulation positive/immunologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Remodelage ventriculaire/génétique
11.
Int Immunopharmacol ; 80: 106156, 2020 Mar.
Article de Anglais | MEDLINE | ID: mdl-31945609

RÉSUMÉ

OBJECTIVE: Recently, the function of microRNAs (miRNAs) has been clarified in human diseases, we aimed to identify the role of miR-185 in myocardial infarction (MI). METHODS: Bone marrow mesenchymal stem cells (BMSCs) were cultured, from which the exosomes were extracted. MI mice models were established by coronary artery ligation and injected with transfected BMSCs. The echocardiographic and ventricle indicators, and hemodynamics of mice were measured. Moreover, the ultrastructure and apoptosis of cardiomyocytes were determined, and expression of miR-185, suppressor of cytokine signaling 2 (SOCS2), collagens, and apoptotic proteins in myocardial tissues were evaluated. RESULTS: MiR-185 was poorly expressed in myocardial tissues of MI mice. BMSCs-Exo could shuttle miR-185 to promote cardiac function and attenuate myocardial injury of myocardial tissues in MI mice, and also could protect cardiomyocytes from apoptosis in MI mice by reducing the expression of SOCS2. SOCS2 was determined to be the direct target gene of miR-185. Overexpressed SOCS2 could block the cardioprotective effect of BMSCs-derived exosomal miR-185 in MI mice. CONCLUSION: We have found in this study that BMSCs-derived exosomal miR-185 could repress ventricular remolding of MI mice by inhibiting SOCS2. This study may provide new method for MI treatment.


Sujet(s)
Cellules souches mésenchymateuses/métabolisme , microARN/métabolisme , Infarctus du myocarde/génétique , Protéines SOCS/génétique , Remodelage ventriculaire/génétique , Animaux , Apoptose/génétique , Apoptose/immunologie , Lignée cellulaire , Modèles animaux de maladie humaine , Échocardiographie , Exosomes/métabolisme , Exosomes/transplantation , Ventricules cardiaques/cytologie , Ventricules cardiaques/imagerie diagnostique , Ventricules cardiaques/immunologie , Ventricules cardiaques/anatomopathologie , Humains , Mâle , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/cytologie , Souris , Infarctus du myocarde/diagnostic , Infarctus du myocarde/immunologie , Infarctus du myocarde/thérapie , Myocytes cardiaques , Remodelage ventriculaire/immunologie
12.
Exp Physiol ; 105(3): 489-501, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31957919

RÉSUMÉ

NEW FINDINGS: What is the central question of this study? We questioned whether the disruption of invariant natural killer T (iNKT) cells exacerbates left ventricular (LV) remodelling and heart failure after transverse aortic constriction in mice. What are the main findings and their importance? Pressure overload induced by transverse aortic constriction increased the infiltration of iNKT cells in mouse hearts. The disruption of iNKT cells exacerbated LV remodelling and hastened the transition from hypertrophy to heart failure, in association with the activation of mitogen-activated protein kinase signalling. Activation of iNKT cells modulated the immunological balance in this process and played a protective role against LV remodelling and failure. ABSTRACT: Chronic inflammation is involved in the development of cardiac remodelling and heart failure (HF). Invariant natural killer T (iNKT) cells, a subset of T lymphocytes, have been shown to produce various cytokines and orchestrate tissue inflammation. The pathophysiological role of iNKT cells in HF caused by pressure overload has not been studied. In the present study, we investigated whether the disruption of iNKT cells affected this process in mice. Transverse aortic constriction (TAC) and a sham operation were performed in male C57BL/6J wild-type (WT) and iNKT cell-deficient Jα18 knockout (KO) mice. The infiltration of iNKT cells was increased after TAC. The disruption of iNKT cells exacerbated left ventricular (LV) remodelling and hastened the transition to HF after TAC. Histological examinations also revealed that the disruption of iNKT cells induced greater myocyte hypertrophy and a greater increase in interstitial fibrosis after TAC. The expressions of interleukin-10 and tumour necrosis factor-α mRNA and their ratio in the LV after TAC were decreased in the KO compared with WT mice, which might indicate that the disruption of iNKT cells leads to an imbalance between T-helper type 1 and type 2 cytokines. The phosphorylation of extracellular signal-regulated kinase was significantly increased in the KO mice. The disruption of iNKT cells exacerbated the development of cardiac remodelling and HF after TAC. The activation of iNKT cells might play a protective role against HF caused by pressure overload. Targeting the activation of iNKT cells might thus be a promising candidate as a new therapeutic strategy for HF.


Sujet(s)
Cardiomégalie/immunologie , Défaillance cardiaque/immunologie , Cellules T tueuses naturelles/immunologie , Animaux , Fibrose/immunologie , Ventricules cardiaques/immunologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Myocarde/immunologie , Myocytes cardiaques/immunologie , Phosphorylation/immunologie , Transduction du signal/immunologie , Remodelage ventriculaire/immunologie
13.
Int Immunopharmacol ; 78: 106055, 2020 Jan.
Article de Anglais | MEDLINE | ID: mdl-31816575

RÉSUMÉ

Peptidyl arginine deiminase-4 (PAD4), a PAD enzyme family member, catalyzes the posttranslational conversion of arginine residues to citrulline in target proteins. Although PAD4 is believed to play a crucial role in various pathological conditions such as infectious diseases, autoimmune diseases, and ischemic conditions, the effect of PAD4 in myocardial infarction (MI)-induced cardiac injury remains to be examined. Here, we hypothesize that PAD4 contributes to cardiac ischemic injury by exacerbating the inflammatory response and promoting neutrophil extracellular trap (NET) formation after MI. Permanent left coronary artery ligation, a condition that mimics MI, was performed on male C57BL/6 mice. [(3S,4R)-3-amino-4-hydroxy-1-piperidinyl] [2-[1-(cyclopropylmethyl)-1H-indol-2-yl]-7-methoxy-1-methyl-1H-benzimidazol-5-yl]-methanone (GSK484), an inhibitor of PAD4, was delivered via intraperitoneal injection to inhibit PAD4 activity. Cardiac PAD4 expression, tissue injury scoring, neutrophil infiltration, cit-H3 expression, NET formation, inflammatory cytokine secretion, apoptosis, and cardiac function were analyzed. In the current study, we discovered the protective effect of PAD4 inhibition using the PAD4-specific inhibitor GSK484 in cardiomyocytes challenged by MI. GSK484-mediated PAD4 inhibition can moderately preserve ventricle histological structure and myocardium integrity after MI, thereby reducing the infarct size and decreasing myocardial enzyme levels in serum. PAD4 inhibition also effectively protects cardiomyocytes from MI-induced NET formation and inflammatory cytokine secretion, in turn alleviating cardiac ischemia-induced apoptosis of cardiomyocytes. Collectively, these findings demonstrate the efficacy of specific PAD4 inhibition in reducing MI-induced neutrophil infiltration, NET formation, inflammatory reaction, and cardiomyocyte apoptosis, thereby increasing overall cardiac function improvement. These results provide novel insights for the development of new strategies to treat cardiovascular dysfunction in MI patients.


Sujet(s)
Cardiotoniques/pharmacologie , Infarctus du myocarde/traitement médicamenteux , Lésion de reperfusion myocardique/prévention et contrôle , Protein-arginine deiminase Type 4/antagonistes et inhibiteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Cardiotoniques/usage thérapeutique , Modèles animaux de maladie humaine , Pièges extracellulaires/effets des médicaments et des substances chimiques , Pièges extracellulaires/immunologie , Pièges extracellulaires/métabolisme , Ventricules cardiaques/immunologie , Ventricules cardiaques/anatomopathologie , Humains , Mâle , Souris , Infarctus du myocarde/immunologie , Infarctus du myocarde/anatomopathologie , Lésion de reperfusion myocardique/immunologie , Lésion de reperfusion myocardique/anatomopathologie , Myocarde/immunologie , Myocarde/anatomopathologie , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/immunologie , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques , Protein-arginine deiminase Type 4/immunologie , Protein-arginine deiminase Type 4/métabolisme , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Remodelage ventriculaire/immunologie
14.
Sci Rep ; 9(1): 18765, 2019 12 10.
Article de Anglais | MEDLINE | ID: mdl-31822739

RÉSUMÉ

Lifestyle factors are important drivers of chronic diseases, including cardiovascular syndromes, with low grade inflammation as a central player. Attenuating myeloperoxidase (MPO) activity, an inflammatory enzyme associated with obesity, hypertension and heart failure, could have protective effects on multiple organs. Herein, the effects of the novel oral available MPO inhibitor AZM198 were studied in an obese/hypertensive mouse model which displays a cardiac phenotype. Eight week old male C57BL6/J mice received 16 weeks of high fat diet (HFD) combined with angiotensin II (AngII) infusion during the last 4 weeks, with low fat diet and saline infusion as control. Treated animals showed therapeutic AZM198 levels (2.1 µM), corresponding to 95% MPO inhibition. AZM198 reduced elevated circulating MPO levels in HFD/AngII mice to normal values. Independent of food intake, bodyweight increase and fat accumulation were attenuated by AZM198, alongside with reduced visceral adipose tissue (VAT) inflammation and attenuated severity of nonalcoholic steatohepatitis. The HFD/AngII perturbation caused impaired cardiac relaxation and contraction, and increased cardiac hypertrophy and fibrosis. AZM198 treatment did, however, not improve these cardiac parameters. Thus, AZM198 had positive effects on the main lipid controlling tissues in the body, namely adipose tissue and liver. This did, however, not directly result in improved cardiac function.


Sujet(s)
Hypertension artérielle/traitement médicamenteux , Hypertrophie ventriculaire gauche/traitement médicamenteux , Stéatose hépatique non alcoolique/traitement médicamenteux , Obésité/traitement médicamenteux , Myeloperoxidase/antagonistes et inhibiteurs , Thioxanthènes/administration et posologie , Angiotensine-II/administration et posologie , Angiotensine-II/toxicité , Animaux , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Ventricules cardiaques/effets des médicaments et des substances chimiques , Ventricules cardiaques/immunologie , Ventricules cardiaques/anatomopathologie , Humains , Hypertension artérielle/sang , Hypertension artérielle/diagnostic , Hypertension artérielle/étiologie , Hypertrophie ventriculaire gauche/sang , Hypertrophie ventriculaire gauche/diagnostic , Hypertrophie ventriculaire gauche/étiologie , Graisse intra-abdominale/effets des médicaments et des substances chimiques , Graisse intra-abdominale/immunologie , Foie/effets des médicaments et des substances chimiques , Foie/immunologie , Foie/anatomopathologie , Mâle , Souris , Stéatose hépatique non alcoolique/sang , Stéatose hépatique non alcoolique/diagnostic , Stéatose hépatique non alcoolique/étiologie , Obésité/sang , Obésité/diagnostic , Obésité/étiologie , Myeloperoxidase/sang , Myeloperoxidase/métabolisme , Indice de gravité de la maladie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Remodelage ventriculaire/immunologie
16.
JCI Insight ; 4(18)2019 09 19.
Article de Anglais | MEDLINE | ID: mdl-31534054

RÉSUMÉ

Over one million Americans experience myocardial infarction (MI) annually, and the resulting scar and subsequent cardiac fibrosis gives rise to heart failure. A specialized cell-cell adhesion protein, cadherin-11 (CDH11), contributes to inflammation and fibrosis in rheumatoid arthritis, pulmonary fibrosis, and aortic valve calcification but has not been studied in myocardium after MI. MI was induced by ligation of the left anterior descending artery in mice with either heterozygous or homozygous knockout of CDH11, wild-type mice receiving bone marrow transplants from Cdh11-deficient animals, and wild-type mice treated with a functional blocking antibody against CDH11 (SYN0012). Flow cytometry revealed significant CDH11 expression in noncardiomyocyte cells after MI. Animals given SYN0012 had improved cardiac function, as measured by echocardiogram, reduced tissue remodeling, and altered transcription of inflammatory and proangiogenic genes. Targeting CDH11 reduced bone marrow-derived myeloid cells and increased proangiogenic cells in the heart 3 days after MI. Cardiac fibroblast and macrophage interactions increased IL-6 secretion in vitro. Our findings suggest that CDH11-expressing cells contribute to inflammation-driven fibrotic remodeling after MI and that targeting CDH11 with a blocking antibody improves outcomes by altering recruitment of bone marrow-derived cells, limiting the macrophage-induced expression of IL-6 by fibroblasts and promoting vascularization.


Sujet(s)
Cadhérines/métabolisme , Infarctus du myocarde/complications , Myocarde/anatomopathologie , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Animaux , Transplantation de moelle osseuse , Cadhérines/antagonistes et inhibiteurs , Cadhérines/génétique , Adhérence cellulaire/effets des médicaments et des substances chimiques , Adhérence cellulaire/génétique , Adhérence cellulaire/immunologie , Modèles animaux de maladie humaine , Échocardiographie , Fibrose , Défaillance cardiaque/étiologie , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/prévention et contrôle , Ventricules cardiaques/imagerie diagnostique , Ventricules cardiaques/effets des médicaments et des substances chimiques , Ventricules cardiaques/immunologie , Ventricules cardiaques/anatomopathologie , Humains , Mâle , Souris , Souris knockout , Cellules myéloïdes/immunologie , Infarctus du myocarde/diagnostic , Infarctus du myocarde/immunologie , Infarctus du myocarde/anatomopathologie , Myocarde/immunologie , Remodelage ventriculaire/immunologie
17.
JCI Insight ; 52019 07 09.
Article de Anglais | MEDLINE | ID: mdl-31287805

RÉSUMÉ

Podoplanin, a small mucine-type transmembrane glycoprotein, has been recently shown to be expressed by lymphangiogenic, fibrogenic and mesenchymal progenitor cells in the acutely and chronically infarcted myocardium. Podoplanin binds to CLEC-2, a C-type lectin-like receptor 2 highly expressed by CD11bhigh cells following inflammatory stimuli. Why podoplanin expression appears only after organ injury is currently unknown. Here, we characterize the role of podoplanin in different stages of myocardial repair after infarction and propose a podoplanin-mediated mechanism in the resolution of post-MI inflammatory response and cardiac repair. Neutralization of podoplanin led to significant improvements in the left ventricular functions and scar composition in animals treated with podoplanin neutralizing antibody. The inhibition of the interaction between podoplanin and CLEC-2 expressing immune cells in the heart enhances the cardiac performance, regeneration and angiogenesis post MI. Our data indicates that modulating the interaction between podoplanin positive cells with the immune cells after myocardial infarction positively affects immune cell recruitment and may represent a novel therapeutic target to augment post-MI cardiac repair, regeneration and function.


Sujet(s)
Cicatrice/métabolisme , Défaillance cardiaque/métabolisme , Hypertrophie ventriculaire gauche/métabolisme , Glycoprotéines membranaires/métabolisme , Infarctus du myocarde/métabolisme , Remodelage ventriculaire/génétique , Angiotensine-II/toxicité , Animaux , Anticorps neutralisants , Cardiomyopathies/immunologie , Cardiomyopathies/métabolisme , Cardiomyopathies/chirurgie , Survie cellulaire/immunologie , Cicatrice/immunologie , Échocardiographie , Fibrose , Défaillance cardiaque/induit chimiquement , Défaillance cardiaque/immunologie , Transplantation cardiaque , Hémodynamique , Humains , Hypertrophie ventriculaire gauche/induit chimiquement , Hypertrophie ventriculaire gauche/immunologie , Inflammation/immunologie , Macrophages/immunologie , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/immunologie , Souris , Monocytes/immunologie , Infarctus du myocarde/immunologie , Ischémie myocardique/immunologie , Ischémie myocardique/métabolisme , Ischémie myocardique/chirurgie , Myocytes cardiaques , Régénération/immunologie , Vasoconstricteurs/toxicité , Fonction ventriculaire gauche , Remodelage ventriculaire/immunologie
18.
Heart Fail Rev ; 24(6): 1019-1030, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31049750

RÉSUMÉ

Myocarditis (MCD) is a type of inflammatory disease in which inflammatory cells infiltrate the myocardium, leading to cardiac dysfunction, myocardial necrosis, and fibrosis. Although it has been reported that MCD is mediated by T cells, the immune system is complex and includes many types of immune cells that interact with one another. Through investigations of the inflammatory responses in MCD including myocardial necrosis, fibrosis, and arrhythmia, we have gained further insight into the pathogenesis of MCD. This article aims to discuss the diversity and the roles of immune cells involved in the pathogenesis of MCD. Moreover, immunotherapy for the treatment of MCD remains controversial, and further investigation is required to identify accurate immunotherapies for special cell types.


Sujet(s)
Système immunitaire/physiologie , Myocardite/immunologie , Myocarde/immunologie , Immunité acquise/immunologie , Animaux , Lymphocytes B/immunologie , Femelle , Fibrose , Antigènes d'histocompatibilité de classe II/immunologie , Humains , Système immunitaire/cytologie , Immunothérapie/méthodes , Cellules tueuses naturelles/immunologie , Macrophages/immunologie , Mâle , Souris , Modèles animaux , Myocardite/physiopathologie , Myocardite/thérapie , Myocarde/anatomopathologie , Récepteurs CCR2/métabolisme , Lymphocytes T régulateurs/immunologie , Remodelage ventriculaire/immunologie
19.
Medicine (Baltimore) ; 98(17): e15194, 2019 Apr.
Article de Anglais | MEDLINE | ID: mdl-31027064

RÉSUMÉ

INTRODUCTION: While the role of inflammation in acute coronary events is well established, the impact of inflammatory-mediated vulnerability of coronary plaques from the entire coronary tree, on the extension of ventricular remodeling and scaring, has not been clarified yet. MATERIALS AND METHODS: The present manuscript describes the procedures of the VIABILITY trial, a descriptive prospective single-center cohort study. The main purpose of this trial is to assess the link between systemic inflammation, pan-coronary plaque vulnerability (referring to the plaque vulnerability within the entire coronary tree), myocardial viability and ventricular remodeling in patients who had suffered a recent ST-segment elevation acute myocardial infarction (STEMI). One hundred patients with STEMI who underwent successful revascularization of the culprit lesion in the first 12 hours after the onset of symptoms will be enrolled in the study. The level of systemic inflammation will be evaluated based on the serum biomarker levels (hs-CRP, matrix metalloproteinases, interleukin-6) in the acute phase of the myocardial infarction (MI) and at 1 month. Pan-coronary plaque vulnerability will be assessed based on serum biomarkers known to be associated with increased plaque vulnerability (V-CAM or I-CAM) and at 1 month after infarction, based on computed tomographic angiography analysis of vulnerability features of all coronary plaques. Myocardial viability and remodeling will be assessed based on 3D speckle tracking echocardiography associated with dobutamine infusion and LGE-CMR associated with post-processing imaging methods. The study population will be categorized in 2 subgroups: subgroup 1 - subjects with STEMI and increased inflammatory response at 7 days after the acute event (hs-CRP ≥ 3 mg/dl), and subgroup 2 - subjects with STEMI and no increased inflammatory response at 7 days (hs-CRP < 3 mg/dl). Study outcomes will consist in the rate of post-infarction heart failure development and the major adverse events (MACE) rate. CONCLUSION: VIABILITY is the first prospective study designed to evaluate the influence of infarct-related inflammatory response on several major determinants of post-infarction outcomes, such as coronary plaque vulnerability, myocardial viability, and ventricular remodeling.


Sujet(s)
Maladie des artères coronaires/immunologie , Inflammation/immunologie , Plaque d'athérosclérose/immunologie , Infarctus du myocarde avec sus-décalage du segment ST/immunologie , Remodelage ventriculaire/immunologie , Marqueurs biologiques/sang , Maladie des artères coronaires/sang , Maladie des artères coronaires/imagerie diagnostique , Humains , Inflammation/sang , Inflammation/imagerie diagnostique , Plaque d'athérosclérose/sang , Plaque d'athérosclérose/imagerie diagnostique , Infarctus du myocarde avec sus-décalage du segment ST/sang , Infarctus du myocarde avec sus-décalage du segment ST/imagerie diagnostique , Infarctus du myocarde avec sus-décalage du segment ST/chirurgie
20.
Front Immunol ; 10: 300, 2019.
Article de Anglais | MEDLINE | ID: mdl-30873166

RÉSUMÉ

The glycosciences aim to understand the impact of extracellular and intracellular carbohydrate structures on biological function. These glycans primarily fall into three major groups: lipid-linked carbohydrates that are referred to as glycosphingolipids or simply glycolipids; relatively short carbohydrate chains that are often O- or N-linked to proteins yielding common glycoproteins; and extended linear polymeric carbohydrate structures that are referred to as glycosaminoglycans (GAGs). Whereas, the impact of such carbohydrate structures has been extensively examined in cancer biology, their role in acute and chronic heart disease is less studied. In this context, a growing body of evidence indicates that glycans play an important role in immune mediated cell recruitment to damaged heart tissue to initiate wound healing and repair after injury. This is particularly important following ischemia and reperfusion that occurs in the heart in the setting of acute myocardial infarction. Here, immune system-mediated repair of the damaged myocardium plays a critical role in determining post-infarction ventricular remodeling, cardiac function, and patient outcome. Further, alterations in immune cell activity can promote the development of heart failure. The present review summarizes our current understanding of the phases of immune-mediated repair following myocardial infarction. It discusses what is known regarding glycans in mediating the recruitment of circulating immune cells during the early inflammatory stage of post-infarction repair, with focus on the selectin family of adhesion molecules. It offers future directions for research aimed at utilizing our knowledge of mechanisms underlying immune cell recruitment to either modulate leukocyte recruitment to the injured tissue or enhance the targeted delivery of biologic therapeutics such as stem cells in an attempt to promote repair of the damaged heart.


Sujet(s)
Infarctus du myocarde/immunologie , Sélectines/immunologie , Animaux , Humains , Infarctus du myocarde/thérapie , Transplantation de cellules souches , Remodelage ventriculaire/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE