Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Biomed Pharmacother ; 143: 112194, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34563949

ABSTRACT

Cardiac pressure overload is a crucial risk factor for cardiac hypertrophy and heart failure. Our previous study showed that depletion of the ß3-adrenergic receptor (ADRB3) induced left ventricular diastolic dysfunction via potential regulation of energy metabolism and cardiac contraction. However, the effects of ADRB3 on pressure overload-induced heart failure remain unclear. In the present study, systemic ADRB3-knockout mice suffering from transverse aortic constriction (TAC) surgery were used to identify the effects of ADRB3 on pressure overload-induced heart failure. Compared to wild-type mice, ADRB3 depletion significantly improved the left ventricular ejection fraction, reduced left ventricular posterior wall thickness and interventricular septum thickness, and decreased the area of cardiomyocytes after TAC. RNA sequencing and bioinformatics analysis showed that ADRB3 depletion up-regulated 275 mRNAs and down-regulated 105 mRNAs in mice suffering TAC surgery. GO analysis, GO-tree analysis, and GSEA showed that ADRB3 depletion mainly enhanced the innate immune response of hearts in cardiac pressure overload mice. In addition, pathway analysis and Pathway-Act analysis presented that innate immune response-related pathways, including RIG-I-like receptor signaling pathway, antigen processing and presentation, Toll-like receptor signaling pathway, and cell adhesion molecules, were significantly enriched in ADRB3-KO-TAC mice. Ten hub genes were identified using protein-protein interaction network, MCODE, and cytoHubba analysis. Furthermore, the depletion and activation of ADRB3 validated the effects of ADRB3 on the innate immune response of hearts after TAC. In conclusion, ADRB3 depletion relieves pressure overload-induced cardiac hypertrophy and heart failure, and these effects could be explained by the enhancement of innate immune response.


Subject(s)
Cardiomegaly/prevention & control , Heart Failure/prevention & control , Immunity, Innate , Myocardium/metabolism , Receptors, Adrenergic, beta-3/deficiency , Animals , Cardiomegaly/immunology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Disease Models, Animal , Gene Regulatory Networks , Heart Failure/immunology , Heart Failure/metabolism , Heart Failure/physiopathology , Immunity, Innate/genetics , Male , Mice, Knockout , Myocardium/immunology , Protein Interaction Maps , Receptors, Adrenergic, beta-3/genetics , Signal Transduction , Stroke Volume , Transcriptome , Ventricular Function, Left , Ventricular Remodeling
2.
Cells ; 10(7)2021 07 03.
Article in English | MEDLINE | ID: mdl-34359851

ABSTRACT

Chronic inflammation, the activation of immune cells and their cross-talk with cardiomyocytes in the pathogenesis and progression of heart diseases has long been overlooked. However, with the latest research developments, it is increasingly accepted that a vicious cycle exists where cardiomyocytes release cardiocrine signaling molecules that spiral down to immune cell activation and chronic state of low-level inflammation. For example, cardiocrine molecules released from injured or stressed cardiomyocytes can stimulate macrophages, dendritic cells, neutrophils and even T-cells, which then subsequently increase cardiac inflammation by co-stimulation and positive feedback loops. One of the key proteins involved in stress-mediated cardiomyocyte signal transduction is a small GTPase RhoA. Importantly, the regulation of RhoA activation is critical for effective immune cell response and is being considered as one of the potential therapeutic targets in many immune-cell-mediated inflammatory diseases. In this review we provide an update on the role of RhoA at the juncture of immune cell activation, inflammation and cardiac disease.


Subject(s)
Heart Diseases/immunology , Leukocytes/immunology , Signal Transduction , rhoA GTP-Binding Protein/metabolism , Animals , Cardiomegaly/immunology , Cardiomegaly/pathology , Heart Diseases/pathology , Heart Failure/immunology , Heart Failure/pathology , Humans
3.
Front Immunol ; 12: 790511, 2021.
Article in English | MEDLINE | ID: mdl-34992607

ABSTRACT

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.


Subject(s)
Cardiomegaly/immunology , Dual-Specificity Phosphatases/deficiency , Heart Failure/immunology , MAP Kinase Signaling System/immunology , Myocardium/pathology , Animals , Blood Pressure , Cardiomegaly/diagnosis , Cardiomegaly/pathology , Cells, Cultured , Disease Models, Animal , Dual-Specificity Phosphatases/genetics , Echocardiography , Fibrosis , Heart/diagnostic imaging , Heart Failure/pathology , Humans , Interleukin-4/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Phosphorylation/immunology , Primary Cell Culture , Ventricular Remodeling/immunology
4.
Circulation ; 142(23): 2240-2258, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33070627

ABSTRACT

BACKGROUND: Cardiac hypertrophy is a key biological response to injurious stresses such as pressure overload and, when excessive, can lead to heart failure. Innate immune activation by danger signals, through intracellular pattern recognition receptors such as nucleotide-binding oligomerization domain 1 (Nod1) and its adaptor receptor-interacting protein 2 (RIP2), might play a major role in cardiac remodeling and progression to heart failure. We hypothesize that Nod1/RIP2 are major contributors to cardiac hypertrophy, but may not be sufficient to fully express the phenotype alone. METHODS: To elucidate the contribution of Nod1/RIP2 signaling to cardiac hypertrophy, we randomized Nod1-/-, RIP2-/-, or wild-type mice to transverse aortic constriction or sham operations. Cardiac hypertrophy, fibrosis, and cardiac function were examined in these mice. RESULTS: Nod1 and RIP2 proteins were upregulated in the heart after transverse aortic constriction, and this was paralleled by increased expression of mitochondrial proteins, including mitochondrial antiviral signaling protein (MAVS). Nod1-/- and RIP2-/- mice subjected to transverse aortic constriction exhibited better survival, improved cardiac function, and decreased cardiac hypertrophy. Downstream signal transduction pathways that regulate inflammation and fibrosis, including NF (nuclear factor) κB and MAPK (mitogen-activated protein kinase)-GATA4/p300, were reduced in both Nod1-/- and RIP2-/- mice after transverse aortic constriction compared with wild-type mice. Coimmunoprecipitation of extracted cardiac proteins and confocal immunofluorescence microscopy showed that Nod1/RIP2 interaction was robust and that this complex also included MAVS as an essential component. Suppression of MAVS expression attenuated the complex formation, NF κB signaling, and myocyte hypertrophy. Interrogation of mitochondrial function compared in the presence or ablation of MAVS revealed that MAVS serves to suppress mitochondrial energy output and mediate fission/fusion related dynamic changes. The latter is possibly linked to mitophagy during cardiomyocytes stress, which may provide an intriguing link between innate immune activation and mitochondrial energy balance under stress or injury conditions. CONCLUSIONS: We have identified that innate immune Nod1/RIP2 signaling is a major contributor to cardiac remodeling after stress. This process is critically joined by and regulated through the mitochondrial danger signal adapter MAVS. This novel complex coordinates remodeling, inflammatory response, and mitochondrial energy metabolism in stressed cardiomyocytes. Thus, Nod1/RIP2/MAVS signaling complex may represent an attractive new therapeutic approach toward heart failure.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cardiomegaly/immunology , Energy Metabolism/physiology , Immunity, Innate/physiology , Nod1 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Cardiomegaly/metabolism , Cardiomegaly/pathology , Female , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction/physiology
5.
Physiol Rep ; 8(15): e14532, 2020 08.
Article in English | MEDLINE | ID: mdl-32786064

ABSTRACT

Pulmonary arterial hypertension (PAH) is a syndrome diagnosed by increased mean pulmonary artery (PA) pressure and resistance and normal pulmonary capillary wedge pressure. PAH is characterized pathologically by distal pulmonary artery remodeling, increased pulmonary vascular resistance, and plexiform lesions (PLs). Right ventricular fibrosis and hypertrophy, leading to right ventricular failure, are the main determinants of mortality in PAH. Recent work suggests that right ventricular fibrosis results from resident cardiac fibroblast activation and conversion to myofibroblasts, leading to replacement of contractile cardiomyocytes with nondistensible tissue incapable of conductivity or contractility. However, the origins, triggers, and consequences of myofibroblast expansion and its pathophysiological relationship with PAH are unclear. Recent advances indicate that signals generated by adaptive and innate immune cells may play a role in right ventricular fibrosis and remodeling. This review summarizes recent insights into the mechanisms by which adaptive and innate immune signals participate in the transition of cardiac fibroblasts to activated myofibroblasts and highlights the existing gaps of knowledge as relates to the development of right ventricular fibrosis.


Subject(s)
Adaptive Immunity , Cardiomegaly/immunology , Hypertension, Pulmonary/complications , Immunity, Innate , Animals , Cardiomegaly/etiology , Cardiomegaly/pathology , Cell Transdifferentiation , Fibrosis , Humans , Macrophages/immunology , Macrophages/pathology , Myofibroblasts/immunology , Myofibroblasts/pathology
6.
Exp Physiol ; 105(3): 489-501, 2020 03.
Article in English | MEDLINE | ID: mdl-31957919

ABSTRACT

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.


Subject(s)
Cardiomegaly/immunology , Heart Failure/immunology , Natural Killer T-Cells/immunology , Animals , Fibrosis/immunology , Heart Ventricles/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/immunology , Myocytes, Cardiac/immunology , Phosphorylation/immunology , Signal Transduction/immunology , Ventricular Remodeling/immunology
7.
Sci Rep ; 9(1): 6055, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988323

ABSTRACT

Heart failure (HF) is the leading cause of death in the Western world. Pathophysiological processes underlying HF development, including cardiac hypertrophy, fibrosis and inflammation, are controlled by specific microRNAs (miRNAs). Whereas most studies investigate miRNA function in one particular cardiac cell type, their multicellular function is poorly investigated. The present study probed 194 miRNAs -differentially expressed in cardiac inflammatory disease - for regulating cardiomyocyte size, cardiac fibroblasts collagen content, and macrophage polarization. Of the tested miRNAs, 13%, 26%, and 41% modulated cardiomyocyte size, fibroblast collagen production, and macrophage polarization, respectively. Seventeen miRNAs affected all three cellular processes, including miRNAs with established (miR-210) and unknown roles in cardiac pathophysiology (miR-145-3p). These miRNAs with a multi-cellular function commonly target various genes. In-depth analysis in vitro of previously unstudied miRNAs revealed that the observed phenotypical alterations concurred with changes in transcript and protein levels of hypertrophy-, fibrosis- and inflammation-related genes. MiR-145-3p and miR-891a-3p were identified to regulate the fibrotic response, whereas miR-223-3p, miR-486-3p, and miR-488-5p modulated macrophage activation and polarisation. In conclusion, miRNAs are multi-cellular regulators of different cellular processes underlying cardiac disease. We identified previously undescribed roles of miRNAs in hypertrophy, fibrosis, and inflammation, and attribute new cellular effects to various well-known miRNAs.


Subject(s)
Cardiomegaly/pathology , Heart Failure/genetics , MicroRNAs/metabolism , Myocarditis/immunology , Myocardium/pathology , Animals , Animals, Newborn , Cardiomegaly/genetics , Cardiomegaly/immunology , Cells, Cultured , Fibroblasts , Fibrosis , Gene Expression Profiling , Gene Expression Regulation , Heart Failure/immunology , Heart Failure/pathology , Humans , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages , Mice , Myocarditis/genetics , Myocarditis/pathology , Myocardium/cytology , Myocardium/immunology , Myocytes, Cardiac , Primary Cell Culture , Rats
8.
Sci Rep ; 9(1): 6047, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988334

ABSTRACT

Early adaptive cardiac hypertrophy (EACH) is initially a compensatory process to optimize pump function. We reported the emergence of Orai3 activity during EACH. This study aimed to characterize how inflammation regulates store-independent activation of Orai3-calcium influx and to evaluate the functional role of this influx. Isoproterenol infusion or abdominal aortic banding triggered EACH. TNFα or conditioned medium from cardiac CD11b/c cells activated either in vivo [isolated from rats displaying EACH], or in vitro [isolated from normal rats and activated with lipopolysaccharide], were added to adult cardiomyocytes before measuring calcium entry, cell hypertrophy and cell injury. Using intramyocardial injection of siRNA, Orai3 was in vivo knockdown during EACH to evaluate its protective activity in heart failure. Inflammatory CD11b/c cells trigger a store-independent calcium influx in hypertrophied cardiomyocytes, that is mimicked by TNFα. Pharmacological or molecular (siRNA) approaches demonstrate that this calcium influx, depends on TNFR2, is Orai3-driven, and elicits cardiomyocyte hypertrophy and resistance to oxidative stress. Neutralization of Orai3 inhibits protective GSK3ß phosphorylation, impairs EACH and accelerates heart failure. Orai3 exerts a pathophysiological protective impact in EACH promoting hypertrophy and resistance to oxidative stress. We highlight inflammation arising from CD11b/c cells as a potential trigger of TNFR2- and Orai3-dependent signaling pathways.


Subject(s)
Calcium Channels/metabolism , Cardiomegaly/immunology , Heart Failure/immunology , Myocytes, Cardiac/immunology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Animals , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Calcium/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Culture Media, Conditioned/metabolism , Disease Models, Animal , Disease Progression , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Isoproterenol/toxicity , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Oxidative Stress/immunology , Phosphorylation/immunology , RNA, Small Interfering/metabolism , Rats , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
Am J Physiol Heart Circ Physiol ; 316(5): H1027-H1038, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30793936

ABSTRACT

Toll-like receptors (TLR) are key components of the innate immune system that elicit inflammatory responses through the adaptor proteins myeloid differentiation protein 88 (MyD88) and Toll-interleukin receptor domain-containing adaptor protein-inducing interferon-ß (TRIF). Previously, we demonstrated that TRIF mediates the signaling of angiotensin II (ANG II)- induced hypertension and cardiac hypertrophy. Since TRIF is activated selectively by TLR3 and TLR4, our goals in this study were to determine the roles of TLR3 and TLR4 in mediating ANG II-induced hypertension and cardiac hypertrophy, and associated changes in proinflammatory gene expression in heart and kidney. In wild-type (WT) mice, ANG II infusion (1,000 ng·kg-1·min-1 for 3 wk) increased systolic blood pressure and caused cardiac hypertrophy. In ANG II-infused TLR4-deficient mice (Tlr4del), hypertrophy was significantly attenuated despite a preserved or enhanced hypertensive response. In contrast, in TLR3-deficient mice (Tlr3-/-), both ANG II-induced hypertension and hypertrophy were abrogated. In WT mice, ANG II increased the expression of several proinflammatory genes in hearts and kidneys that were attenuated in both TLR4- and TLR3-deficient mice compared with WT. We conclude that ANG II activates both TLR4-TRIF and TLR3-TRIF pathways in a nonredundant manner whereby hypertension is dependent on activation of the TLR3-TRIF pathway and cardiac hypertrophy is dependent on both TLR3-TRIF and TLR4-TRIF pathways. NEW & NOTEWORTHY Angiotensin II (ANG II)-induced hypertension is dependent on the endosomal Toll-like receptor 3 (TLR3)-Toll-interleukin receptor domain-containing adaptor protein-inducing interferon-ß (TRIF) pathway of the innate immune system but not on cell membrane localized TLR4. However, ANG II-induced cardiac hypertrophy is regulated by both TLR4-TRIF and TLR3-TRIF pathways. Thus, ANG II-induced rise in systolic blood pressure is independent of TLR4-TRIF effect on cardiac hypertrophy. The TLR3-TRIF pathway may be a potential target of therapeutic intervention.


Subject(s)
Angiotensin II , Cardiomegaly/metabolism , Hypertension/metabolism , Immunity, Innate , Kidney/metabolism , Myocardium/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/genetics , Cardiomegaly/immunology , Disease Models, Animal , Hypertension/chemically induced , Hypertension/genetics , Hypertension/immunology , Inflammation Mediators/metabolism , Kidney/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardium/immunology , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
10.
J Mol Cell Cardiol ; 129: 293-302, 2019 04.
Article in English | MEDLINE | ID: mdl-30641087

ABSTRACT

Pressure overload-induced cardiac hypertrophy and remodeling are not simply mechanical responses to overloaded stress. They also involve participation of various immune cells, especially T cells. In this review, we summarized recent advances in understanding the roles of T cells in this process and the possible mechanisms underlying T cells involvement and modulation. In this pathological process, αßT cells play an indispensable role, with contribution of NKT cells and γδT cells. Moreover, among the αßT cells, CD4+ T cells rather than CD8+ T cells are dominant in the process with different subsets exerting diverse influences. Th1 and Th17 cells mainly promote the pathological development, while Treg cells are negative modulators that alleviate cardiac hypertrophy and remodeling. Even though the involvement of T cells has been reported extensively, the detailed modulating mechanism remains to be elucidated. Pressure overload exerting on heart stimulates cytokines secretion from resident cardiac cells and upregulates cell adhesion molecules on cardiac endothelial cells, which together might mediate T cells infiltration into the heart. Infiltrating T cells modulate pro-hypertrophic pathways and the transition from cardiac fibroblasts to myofibroblasts, which might represent mechanisms underlying their effects. Considering the vital participation of T cells, immune regulation is a promising treatment direction for this class of diseases and more work is needed to reveal the detailed mechanism.


Subject(s)
Cardiomegaly/immunology , Cardiomegaly/physiopathology , Pressure , T-Lymphocytes/immunology , Ventricular Remodeling , Animals , Humans , Models, Cardiovascular
11.
Cardiovasc Res ; 115(1): 83-93, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29939225

ABSTRACT

Aims: CD1d is a member of the cluster of differentiation 1 (CD1) family of glycoproteins expressed on the surface of various antigen-presenting cells, which is recognized by natural killer T (NKT) cells. CD1d-dependent NKT cells play an important role in immune-mediated diseases; but the role of these cells in regulating cardiac remodelling remains unknown. Methods and results: Cardiac remodelling was induced by angiotensin (Ang) II infusion for 2 weeks. Ang II-induced increase in hypertension, cardiac performance, hypertrophy and fibrosis, inflammatory response, and activation of the NF-kB and TGF-ß1/Smad2/3 pathways was significantly aggravated in CD1d knockout (CD1dko) mice compared with wild-type (WT) mice, but these effects were markedly abrogated in WT mice treated with α-galactosylceramide (αGC), a specific activator of NKT cells. Adoptive transfer of CD1dko bone marrow cells to WT mice further confirmed the deleterious effect of CD1dko. Moreover, IL-10 expression was significantly decreased in CD1dko hearts but increased in αGC-treated mice. Co-culture experiments revealed that CD1dko dendritic cells significantly reduced IL-10 mRNA expression from NKT cells. Administration of recombinant murine IL-10 to CD1dko mice improved hypertension, cardiac performance, and adverse cardiac remodelling induced by Ang II, and its cardioprotective effect was possibly associated with activation of STAT3, and inhibition of the TGF-ß1 and NF-kB pathways. Conclusion: These findings revealed a previously undefined role for CD1d-dependent NKT cells in Ang II-induced cardiac remodelling, hence activation of NKT cells may be a novel therapeutic target for hypertensive cardiac disease.


Subject(s)
Angiotensin II , Antigens, CD1d/metabolism , Cardiomegaly/metabolism , Hypertension/physiopathology , Interleukin-10/metabolism , Myocytes, Cardiac/metabolism , Natural Killer T-Cells/metabolism , Ventricular Remodeling , Adoptive Transfer , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Cardiomegaly/chemically induced , Cardiomegaly/immunology , Cardiomegaly/physiopathology , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Fibrosis , Galactosylceramides/pharmacology , Hypertension/chemically induced , Hypertension/immunology , Hypertension/metabolism , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/transplantation , STAT3 Transcription Factor/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Ventricular Remodeling/drug effects
12.
Biochem Biophys Res Commun ; 509(1): 16-23, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30581005

ABSTRACT

Pathological cardiac hypertrophy is a leading cause of morbidity and mortality in the world. However, it is still unclear the molecular mechanism revealing the progression of the disease. In the study, we illustrated that the expression of leukocyte immunoglobulin-like receptor B4 (LILRB4), associated with the pathological development of various inflammatory diseases, was down-regulated in pressure overload-induced hearts of patients and mice. LILRB4-knockout mice developed cardiac hypertrophy and heart failure by promoting cardiac dysfunction, fibrosis, inflammation and apoptosis. Mechanistically, transforming growth factor ß1 (TGF-ß1) expression was significantly promoted by LILRB4 deficiency in hearts of mice after aortic banding (AB) surgery. AB-induced inflammation in cardiac tissues was accelerated by LILRB4 deletion through elevating nuclear factor κB (NF-κB) signaling pathway. Furthermore, apoptosis triggered by AB operation in heart tissues was markedly enhanced in LILRB4-KO mice through promoting Caspase-3 activation. Importantly, the in vitro study indicated that LILRB4 knockdown-promoted fibrosis; inflammation and apoptosis were largely via the NF-κB signaling. Therefore, the findings above identified LILRB4 might be a negative regulator of cardiac remodeling, illustrating that LILRB4 represented as a therapeutic target for the prevention of cardiac hypertrophy and heart failure.


Subject(s)
Cardiomegaly/pathology , Inflammation/pathology , Membrane Glycoproteins/immunology , Myocardium/pathology , NF-kappa B/immunology , Receptors, Cell Surface/immunology , Receptors, Immunologic/immunology , Animals , Apoptosis , Cardiomegaly/genetics , Cardiomegaly/immunology , Cells, Cultured , Down-Regulation , Fibrosis , Humans , Inflammation/genetics , Inflammation/immunology , Male , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics
13.
Circulation ; 139(11): 1407-1421, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30586752

ABSTRACT

BACKGROUND: Arterial hypertension and its organ sequelae show characteristics of T cell-mediated inflammatory diseases. Experimental anti-inflammatory therapies have been shown to ameliorate hypertensive end-organ damage. Recently, the CANTOS study (Canakinumab Antiinflammatory Thrombosis Outcome Study) targeting interleukin-1ß demonstrated that anti-inflammatory therapy reduces cardiovascular risk. The gut microbiome plays a pivotal role in immune homeostasis and cardiovascular health. Short-chain fatty acids (SCFAs) are produced from dietary fiber by gut bacteria and affect host immune homeostasis. Here, we investigated effects of the SCFA propionate in 2 different mouse models of hypertensive cardiovascular damage. METHODS: To investigate the effect of SCFAs on hypertensive cardiac damage and atherosclerosis, wild-type NMRI or apolipoprotein E knockout-deficient mice received propionate (200 mmol/L) or control in the drinking water. To induce hypertension, wild-type NMRI mice were infused with angiotensin II (1.44 mg·kg-1·d-1 subcutaneous) for 14 days. To accelerate the development of atherosclerosis, apolipoprotein E knockout mice were infused with angiotensin II (0.72 mg·kg-1·d-1 subcutaneous) for 28 days. Cardiac damage and atherosclerosis were assessed using histology, echocardiography, in vivo electrophysiology, immunofluorescence, and flow cytometry. Blood pressure was measured by radiotelemetry. Regulatory T cell depletion using PC61 antibody was used to examine the mode of action of propionate. RESULTS: Propionate significantly attenuated cardiac hypertrophy, fibrosis, vascular dysfunction, and hypertension in both models. Susceptibility to cardiac ventricular arrhythmias was significantly reduced in propionate-treated angiotensin II-infused wild-type NMRI mice. Aortic atherosclerotic lesion area was significantly decreased in propionate-treated apolipoprotein E knockout-deficient mice. Systemic inflammation was mitigated by propionate treatment, quantified as a reduction in splenic effector memory T cell frequencies and splenic T helper 17 cells in both models, and a decrease in local cardiac immune cell infiltration in wild-type NMRI mice. Cardioprotective effects of propionate were abrogated in regulatory T cell-depleted angiotensin II-infused mice, suggesting the effect is regulatory T cell-dependent. CONCLUSIONS: Our data emphasize an immune-modulatory role of SCFAs and their importance for cardiovascular health. The data suggest that lifestyle modifications leading to augmented SCFA production could be a beneficial nonpharmacological preventive strategy for patients with hypertensive cardiovascular disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aortic Diseases/drug therapy , Arrhythmias, Cardiac/prevention & control , Atherosclerosis/drug therapy , Cardiomegaly/prevention & control , Hypertension/drug therapy , Propionates/pharmacology , Angiotensin II , Animals , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/physiopathology , Arterial Pressure/drug effects , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Cardiomegaly/immunology , Cardiomegaly/physiopathology , Disease Models, Animal , Hypertension/chemically induced , Hypertension/immunology , Hypertension/physiopathology , Male , Mice, Knockout, ApoE , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
14.
Int J Mol Sci ; 19(12)2018 Nov 22.
Article in English | MEDLINE | ID: mdl-30467294

ABSTRACT

Heart failure (HF) is a cardiovascular syndrome characterized by maladaptive changes with an underlying inflammatory mediated pathogenesis. Nevertheless, current therapy is aimed at the heart workload and neurohormonal axis; thus, prognosis remains poor. To continue improving treatment, we rely on murine models for a better understanding of HF pathophysiology. Among them, pressure overload HF (PO-HF) animal models are a common strategy. Development of PO-HF is characterized by monocyte infiltration, which orchestrates a cascade of events leading to sustained inflammation and maladaptive changes. Here, we divide the PO-HF model progression into four phases and describe the inflammatory, structural, and gene expression profiles. This division is relevant due to its similarities with clinical hypertensive heart disease progression to HF. Evidence shows improvement in hemodynamic and other local parameters by altering the inflammatory response in a specific immune response at a specific point of time. Thus, it is relevant to focus on the time-dependent immune response interaction in order to provide more effective therapy. This review summarizes the pathogenesis of PO-HF murine models, highlighting the inflammatory events in a time frame view. By this approach, we expect to provide researchers with a better understanding of the intertwining time-dependent events that occur in PO-HF.


Subject(s)
B-Lymphocytes/immunology , Heart Failure/immunology , Hypertension/immunology , Monocytes/immunology , T-Lymphocytes/immunology , Angiotensin II/administration & dosage , Angiotensin II/adverse effects , Animals , Aorta/immunology , Aorta/pathology , B-Lymphocytes/pathology , Cardiomegaly/immunology , Cardiomegaly/pathology , Cell Movement , Constriction, Pathologic/immunology , Constriction, Pathologic/pathology , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Endomyocardial Fibrosis/immunology , Endomyocardial Fibrosis/pathology , Heart Failure/chemically induced , Heart Failure/etiology , Heart Failure/pathology , Humans , Hypertension/complications , Hypertension/pathology , Mice , Monocytes/pathology , T-Lymphocytes/pathology , Time Factors , Ventricular Dysfunction, Left/immunology , Ventricular Dysfunction, Left/pathology
15.
Proc Natl Acad Sci U S A ; 115(20): E4661-E4669, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29712858

ABSTRACT

Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macrophage proliferation occurs within the first week following pressure overload hypertrophy (POH; a model of heart failure) and is requisite for the heart's adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription factor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macrophages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappreciated temporal and spatial roles for resident and nonresident macrophages in the development of heart failure.


Subject(s)
Cardiomegaly/pathology , Cardiomyopathies/pathology , Heart Failure/pathology , Kruppel-Like Transcription Factors/metabolism , Macrophages/pathology , Myocardium/pathology , Animals , Cardiomegaly/immunology , Cardiomegaly/metabolism , Cardiomyopathies/immunology , Cardiomyopathies/metabolism , Cells, Cultured , Heart Failure/immunology , Heart Failure/metabolism , Kruppel-Like Factor 4 , Macrophages/immunology , Macrophages/metabolism , Mice , Myocardium/immunology , Myocardium/metabolism , Pressure
16.
Basic Res Cardiol ; 113(3): 19, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29564567

ABSTRACT

Previous studies have suggested the involvement of CD4 + T lymphocytes in cardiac remodelling. T-bet can direct Th1 lineage commitment. This study aimed to investigate the functional significance of T-bet in cardiac remodelling induced by pressure overload using T-bet global knockout rats. Increased T-bet levels were observed in rodent and human hypertrophied hearts. T-bet deficiency resulted in a less severe hypertrophic phenotype in rats. CD4 + T-lymphocyte reconstitution in T-bet-/- rats resulted in aggravated cardiac remodelling. T-cell homing molecule expression and cytokine secretion were altered in T-bet-deficient rat hearts. Administration of exogenous interferon-γ (IFN-γ) offset T-bet deficiency-mediated cardioprotection. Cardiomyocytes cultured in T-bet-/- CD4 + T-cell-conditioned media showed a reduced hypertrophic response after hypertrophic stimuli, which was abolished by an IFN-γ-neutralizing antibody. Taken together, our findings show that T-bet deficiency attenuates pressure overload-induced cardiac remodelling in rats. Specifically, targeting T-bet in T cells may be of great importance for the treatment of pathological cardiac remodelling and heart failure.


Subject(s)
Cardiomegaly/metabolism , Cardiomyopathy, Dilated/metabolism , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/deficiency , Th1 Cells/metabolism , Ventricular Remodeling , Adoptive Transfer , Animals , Cardiomegaly/immunology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/prevention & control , Cells, Cultured , Chemotaxis, Leukocyte , Cytokines/immunology , Cytokines/metabolism , Gene Knockdown Techniques , Genotype , Humans , Interferon-gamma/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Paracrine Communication , Phenotype , Rats, Sprague-Dawley , Rats, Transgenic , Signal Transduction , T-Box Domain Proteins/genetics , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/transplantation , Ventricular Remodeling/drug effects , Ventricular Remodeling/genetics
17.
Front Immunol ; 9: 3029, 2018.
Article in English | MEDLINE | ID: mdl-30619368

ABSTRACT

The transcription factor STAT3 has a protective function in the heart. Until recently, the role of STAT3 in hypertension-induced cardiac hypertrophy was unsettled. Earlier studies revealed that global reduction of STAT3 activity reduced cardiac hypertrophy with hypertension, but caused a disruption of myofilaments and increased contractile dysfunction. However, newer studies with cardiomyocyte-specific deletion of STAT3 indicate that STAT3 does not cause cardiac hypertrophy with increased blood pressure. Rather, cardiac STAT3 is important for maintaining metabolic homeostasis, and loss of STAT3 in cardiomyocytes makes the heart more susceptible to chronic pathological insult, for example by disrupting glucose metabolism and protective signaling networks via the upregulation of certain microRNAs. This scenario has implications for understanding peripartum cardiomyopathy as well. In viral myocarditis, STAT3 opposes the initiation of the dilated phenotype by maintaining membrane integrity via the expression of dystrophin. STAT3 signaling was also found to attenuate myocarditis by polarizing macrophages to a less inflammatory phenotype. On the other hand, STAT3 contributes to immune-mediated myocarditis due to IL-6-induced complement component C3 production in the liver, as well as the differentiation of Th17 cells, which play a role in initiation and development of myocarditis. Besides canonical signaling pathways, unphosphorylated STAT3 (U-STAT3) and redox-activated STAT3 have been shown to couple to transcription in the heart. In addition, tissue signaling cytokines such as IL-22 and IL-17 have been proposed to have actions on the heart that involve STAT3, but are not fully defined. Understanding the novel and often protective aspects of STAT3 in the myocardium could lead to new therapeutic approaches to treat heart disease.


Subject(s)
Cardiomegaly/immunology , Heart/physiology , Myocarditis/immunology , Myocardium/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cardiomegaly/physiopathology , Complement C3/immunology , Disease Models, Animal , Humans , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukins/immunology , Interleukins/metabolism , Macrophages/immunology , Macrophages/metabolism , Myocarditis/physiopathology , Myocardium/cytology , Myocardium/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , Interleukin-22
18.
Cardiovasc Res ; 114(3): 409-416, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29244142

ABSTRACT

AIMS: The aim of our study was to set up a simple and reliable isolation method of living ventricular cardiomyocytes (vCMs) for molecular and biological studies. METHODS AND RESULTS: A standard technique for the retrograde perfusion of an enzymatic solution was used to isolate cardiac cells from adult mouse heart. Fluorescence-activated cell sorting (FACS) on adult murine cardiac ventricle cells was performed, comparing the intrinsic autofluorescence in the FITC channel and the forward scatter (FSC) parameter in order to isolate highly fluorescent cells. The expression of cell-specific mRNAs was assessed with real-time PCR in cells sorted on the basis of their FITC and FSC characteristics. We identified two distinct subpopulations of cells harvested after retrograde perfusion of wild-type heart: FITChigh/FSCdim and FITCdim/FSChigh. Immunophenotyping and mRNA analysis (qPCR and RNA sequencing) revealed that only FITChigh/FSCdim cells were highly enriched in CM markers. Genes with high expression in endothelial cells and fibroblasts were enriched in the FITCdim/FSChigh subpopulation. With the use of tdTomatofl/fl-α-myosin heavy chain MerCreMer+/-mouse heart, we found that tdTomato-positive vCMs were present in the FITChigh/FSCdim region but were only rare in the FITCdim/FSChigh fraction. CONCLUSION: We have developed a simple and reliable method for the isolation of highly purified vCMs from the adult murine myocardium, avoiding fixation and permeabilization steps. These isolated vCMs can be used in particular for detailed molecular studies, avoiding contamination with other myocardial cell types.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Heart Ventricles/metabolism , Myocytes, Cardiac/metabolism , Animals , Biomarkers/metabolism , Cardiomegaly/immunology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Heart Ventricles/immunology , Heart Ventricles/pathology , Luminescent Measurements , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Cardiovasc Res ; 113(13): 1538-1550, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29088374

ABSTRACT

Canonical innate immune signalling involves complex cascades: multiple germline-encoded pattern recognition receptors rapidly recognize pathogen-associated or damage-associated molecular patterns to induce the production of cytokines, which bind to their corresponding receptors to orchestrate subsequent host defense phases. Inflammation is a healthy response to pathogenic signals, which are typically rapid and specific, and they terminate once the threat has passed. However, excessive activation or suppression of innate immune or inflammatory responses can lead to considerable human suffering, such as cardiac remodelling. Interestingly, recent studies have revealed that innate immune molecules in the parenchymal cells of the heart influence cardiac homeostasis not only by directly regulating innate immune responses but also through reprogrammed signalling pathways, which are independent of conventional innate immune signalling. Elucidating 'innate immune signalling reprogramming' events will help us better understand the functions of innate immune molecules and, moreover, the pathogenesis of cardiac diseases.


Subject(s)
Cardiomegaly/immunology , Heart Failure/immunology , Hypertension/immunology , Immunity, Innate , Myocardial Ischemia/immunology , Myocardium/immunology , Ventricular Remodeling , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Hypertension/metabolism , Hypertension/pathology , Hypertension/physiopathology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardium/metabolism , Myocardium/pathology , Protein Kinases/immunology , Protein Kinases/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction
20.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1568-1574, 2017 06.
Article in English | MEDLINE | ID: mdl-28254494

ABSTRACT

In the view of the relationships between excessive sodium intake, immunity and target organ damage, we hypothesized that reduction in dietary sodium would be beneficial in the prevention of cardiac alterations through a restrained local immunity response in a rat model of metabolic syndrome. Sprague-Dawley rats were fed a 60% fructose diet with either a normal sodium (0.64% NaCl) or a low sodium content (<0.01% NaCl) for 8weeks. After 4weeks, rats were infused or not with angiotensin II (200ng·kg-1·min-1, sc) for 4weeks. Tail-cuff blood pressure was determined in conscious rats. Heart and left ventricle weight, cardiomyocyte size, and cardiac fibrosis were evaluated. We performed a transcriptomic analysis in order to identify differentially regulated cardiac mRNAs between normal and low sodium diets. We validated those results using qPCR and immunohistochemistry. Angiotensin II-induced blood pressure rise was blunted (~50%) in the low-sodium fed rats while cardiac hypertrophy and fibrosis were prevented. Transcriptomic analysis revealed 66 differentially regulated genes including 13 downregulated genes under the low sodium diet and implicated in the innate immune response. This was confirmed by reduced cardiac macrophages infiltration under the low sodium diet. Dietary sodium restriction prevents structural alterations of the heart of rats with fructose-induced insulin resistance and angiotensin II-hypertension. The reduction of cardiac inflammation and macrophage infiltration suggests that innate immunity has an important role in the beneficial effect of sodium restriction on cardiac remodeling.


Subject(s)
Cardiomegaly , Diet, Sodium-Restricted , Dietary Carbohydrates/adverse effects , Fructose/adverse effects , Immunity, Innate , Metabolic Syndrome , Animals , Cardiomegaly/diet therapy , Cardiomegaly/immunology , Dietary Carbohydrates/pharmacology , Disease Models, Animal , Fibrosis , Fructose/pharmacology , Metabolic Syndrome/chemically induced , Metabolic Syndrome/diet therapy , Metabolic Syndrome/immunology , Rats , Rats, Sprague-Dawley , Sodium Chloride, Dietary/adverse effects , Sodium Chloride, Dietary/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...