Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34576040

ABSTRACT

Takotsubo syndrome (TTS), recognized as stress's cardiomyopathy, or as left ventricular apical balloon syndrome in recent years, is a rare pathology, described for the first time by Japanese researchers in 1990. TTS is characterized by an interindividual heterogeneity in onset and progression, and by strong predominance in postmenopausal women. The clear causes of these TTS features are uncertain, given the limited understanding of this intriguing syndrome until now. However, the increasing frequency of TTS cases in recent years, and particularly correlated to the SARS-CoV-2 pandemic, leads us to the imperative necessity both of a complete knowledge of TTS pathophysiology for identifying biomarkers facilitating its management, and of targets for specific and effective treatments. The suspect of a genetic basis in TTS pathogenesis has been evidenced. Accordingly, familial forms of TTS have been described. However, a systematic and comprehensive characterization of the genetic or epigenetic factors significantly associated with TTS is lacking. Thus, we here conducted a systematic review of the literature before June 2021, to contribute to the identification of potential genetic and epigenetic factors associated with TTS. Interesting data were evidenced, but few in number and with diverse limitations. Consequently, we concluded that further work is needed to address the gaps discussed, and clear evidence may arrive by using multi-omics investigations.


Subject(s)
COVID-19/complications , Epigenesis, Genetic/immunology , Genetic Heterogeneity , Genetic Predisposition to Disease , Takotsubo Cardiomyopathy/genetics , Biomarkers/analysis , COVID-19/immunology , COVID-19/virology , DNA Copy Number Variations/immunology , Genetic Loci/immunology , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Medical History Taking , Polymorphism, Single Nucleotide/immunology , SARS-CoV-2/immunology , Takotsubo Cardiomyopathy/diagnosis , Takotsubo Cardiomyopathy/immunology , Takotsubo Cardiomyopathy/pathology
2.
Eur J Pharmacol ; 909: 174438, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34437885

ABSTRACT

Increasing evidence indicates that patients or experimental animals exposure to endotoxin (lipopolysaccharides, LPS) exert deleterious cardiac functions that greatly contribute to morbidity and mortality. The pathophysiologic processes, including NLRP3 inflammasome overactivation and cardiac inflammatory injury, are complicated. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, is a naturally occurring compound extracted from Salvia miltiorrhiza and has anti-inflammatory and cardioprotective properties. In this study we examined the effect of STS on endotoxin-induced cardiomyopathy and investigated the underlying mechanisms. An endotoxemic mouse model was established by injecting LPS (10 mg/kg). Different doses of STS were administered intraperitoneally (5, 10, or 50 mg/kg) at different time points (2/12 h, 4/12 h, and 8/12 h) after LPS challenge to assess its effect on survival of mice with endotoxemia. In parallel, cardiac function, myocardial inflammatory cytokines, cardiomyocyte pyroptosis and autophagy were evaluated to determine the extent of myocardial damage due to sepsis in the presence and absence of STS at the optimal dose (10 mg/kg) and time-point (2/12 h). The results demonstrated that STS increased the survival rates, improved the compromised cardiac function and reduced myocardial inflammatory injury associated with enhanced autophagy and mitigated NLRP3 inflammasome activation. Moreover, inhibiting of autophagy or blocking the AMPK pathway reversed STS-elicited prevention of cardiomyopathy and activated the NLRP3 inflammasome in endotoxemic mice. Collectively, our study demonstrates that STS attenuates endotoxemia-induced mortality and cardiomyopathy, which may be associated with promotion of autophagy and inhibition of NLRP3 inflammasome overactivation.


Subject(s)
Cardiomyopathies/prevention & control , Endotoxemia/drug therapy , Inflammasomes/antagonists & inhibitors , Phenanthrenes/pharmacology , Animals , Autophagy/drug effects , Autophagy/immunology , Cardiomyopathies/diagnosis , Cardiomyopathies/immunology , Cardiomyopathies/microbiology , Disease Models, Animal , Echocardiography , Endotoxemia/complications , Endotoxemia/immunology , Endotoxemia/microbiology , Endotoxins/blood , Endotoxins/immunology , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Male , Mice , Myocytes, Cardiac , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phenanthrenes/therapeutic use , Pyroptosis/drug effects , Pyroptosis/immunology
3.
Theranostics ; 11(12): 5939-5954, 2021.
Article in English | MEDLINE | ID: mdl-33897891

ABSTRACT

Aims: We previously found that complement components are upregulated in the myocardium of patients with arrhythmogenic right ventricular cardiomyopathy (ARVC), and inhibiting the complement receptor C5aR reduces disease severity in desmin knockout (Des-/- ) mice, a model for ARVC. Here, we examined the mechanism underlying complement activation in ARVC, revealing a potential new therapeutic target. Methods: First, immunostaining, RT-PCR and western blot were used to detect the expression levels of complement and coagulation factors. Second, we knocked out the central complement component C3 in Des-/- mice (ARVC model) by crossing Des-/- mice with C3-/- mice to explore whether complement system activation occurs independently of the conventional pathway. Then, we evaluated whether a targeted intervention to coagulation system is effective to reduce myocardium injury. Finally, the plasma sC5b9 level was assessed to investigate the role in predicting adverse cardiac events in the ARVC cohort. Results: The complement system is activated in the myocardium in ARVC. Autoantibodies against myocardial proteins provided a possible mechanism underlying. Moreover, we found increased levels of myocardial C5 and the serum C5a in Des-/-C3-/- mice compared to wild-type mice, indicating that C5 is activated independently from the conventional pathway, presumably via the coagulation system. Crosstalk between the complement and coagulation systems exacerbated the myocardial injury in ARVC mice, and this injury was reduced by using the thrombin inhibitor lepirudin. In addition, we found significantly elevated plasma levels of sC5b9 and thrombin in patients, and this increase was correlated with all-cause mortality. Conclusions: These results suggest that crosstalk between the coagulation and complement systems plays a pathogenic role in cardiac dysfunction in ARVC. Thus, understanding this crosstalk may have important clinical implications with respect to diagnosing and treating ARVC.


Subject(s)
Blood Coagulation/immunology , Complement Activation/immunology , Heart Ventricles/immunology , Myocardium/immunology , Adult , Animals , Arrhythmogenic Right Ventricular Dysplasia/immunology , Autoantibodies/immunology , Female , Hirudins/immunology , Humans , Male , Mice, Knockout , Middle Aged , Recombinant Proteins/immunology , Thrombin/immunology
4.
Int J Mol Sci ; 22(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33578743

ABSTRACT

The pathogenesis of idiopathic pulmonary arterial hypertension (IPAH) is not fully understood, but evidence is accumulating that immune dysfunction plays a significant role. We previously reported that 31-week-old Tnfaip3DNGR1-KO mice develop pulmonary hypertension (PH) symptoms. These mice harbor a targeted deletion of the TNFα-induced protein-3 (Tnfaip3) gene, encoding the NF-κB regulatory protein A20, specifically in type I conventional dendritic cells (cDC1s). Here, we studied the involvement of dendritic cells (DCs) in PH in more detail. We found various immune cells, including DCs, in the hearts of Tnfaip3DNGR1-KO mice, particularly in the right ventricle (RV). Secondly, in young Tnfaip3DNGR1-KO mice, innate immune activation through airway exposure to toll-like receptor ligands essentially did not result in elevated RV pressures, although we did observe significant RV hypertrophy. Thirdly, PH symptoms in Tnfaip3DNGR1-KO mice were not enhanced by concomitant mutation of bone morphogenetic protein receptor type 2 (Bmpr2), which is the most affected gene in PAH patients. Finally, in human IPAH lung tissue we found co-localization of DCs and CD8+ T cells, representing the main cell type activated by cDC1s. Taken together, these findings support a unique role of cDC1s in PAH pathogenesis, independent of general immune activation or a mutation in the Bmpr2 gene.


Subject(s)
Dendritic Cells/immunology , Familial Primary Pulmonary Hypertension/immunology , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Dendritic Cells/pathology , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/pathology , Gene Deletion , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Immunity, Innate , Mice , Mutation , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/genetics
5.
Int J Cardiovasc Imaging ; 36(8): 1455-1464, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32297099

ABSTRACT

To investigate the value of tissue Doppler velocities for ruling out treatment-requiring acute cellular rejection (TR-ACR), in the context of myocardial deformation analysis performed by means of speckle tracking echocardiography. We performed serial echocardiograms in 37 heart transplant recipients in their first year post-transplantation within 3 h of the routine surveillance endomyocardial biopsies (EMB). The association of the sum of lateral mitral annulus systolic (s') and early diastolic (e') velocities, in absolute values, measured by tissue Doppler echocardiography (s'+ e'), with TR-ACR (ACR grade ≥ 2R) was investigated by multivariate analysis, including classic echocardiographic parameters and myocardial deformation variables. A total of 251 pairs of EMB and echo exams were performed, 35 (14%) with rejection grade ≥ 2R (TR-ACR). s' + e' was independently associated to TR-ACR (OR 0.80, 95%CI 0.72-0.89, p < 0.0005), with a C statistic of 0.79 (95%CI 0.71-0.87, p < 0.0005) by ROC curve analysis. An s'+ e' value ≥ 23 cm/s, present in 43% of studies, had a negative predictive value of 98% for ruling out TR-ACR. Moreover, in the same patients, s'+ e' significantly decreased when TR-ACR occurred after a study without this condition (- 3.7 ± 3.3 cm/s, p = 0.003), but it was similar when rejection status was the same in the present versus the previous study. A drop in s'+ e' value < 2.7 cm/s from the previous echocardiogram, had a 99% negative predictive value for ruling out TR-ACR. Tissue Doppler velocities, a widely available echo parameter, were found to be a valuable marker for ruling out TR-ACR in this multivariate study which included myocardial deformation variables.


Subject(s)
Echocardiography, Doppler , Graft Rejection/diagnostic imaging , Heart Transplantation/adverse effects , Heart Ventricles/diagnostic imaging , Mitral Valve/diagnostic imaging , Acute Disease , Adult , Aged , Female , Graft Rejection/immunology , Graft Rejection/physiopathology , Heart Ventricles/immunology , Heart Ventricles/physiopathology , Hemodynamics , Humans , Immunity, Cellular , Male , Middle Aged , Mitral Valve/immunology , Mitral Valve/physiopathology , Multivariate Analysis , Myocardium/immunology , Predictive Value of Tests , Prospective Studies , Risk Assessment , Risk Factors , Treatment Outcome , Ventricular Function, Left
6.
Int Immunopharmacol ; 80: 106156, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31945609

ABSTRACT

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.


Subject(s)
Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Myocardial Infarction/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Ventricular Remodeling/genetics , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Disease Models, Animal , Echocardiography , Exosomes/metabolism , Exosomes/transplantation , Heart Ventricles/cytology , Heart Ventricles/diagnostic imaging , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Myocardial Infarction/diagnosis , Myocardial Infarction/immunology , Myocardial Infarction/therapy , Myocytes, Cardiac , Ventricular Remodeling/immunology
7.
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
8.
Int Immunopharmacol ; 78: 106055, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31816575

ABSTRACT

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.


Subject(s)
Cardiotonic Agents/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/prevention & control , Protein-Arginine Deiminase Type 4/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/immunology , Cardiotonic Agents/therapeutic use , Disease Models, Animal , Extracellular Traps/drug effects , Extracellular Traps/immunology , Extracellular Traps/metabolism , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Male , Mice , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/immunology , Neutrophil Infiltration/drug effects , Protein-Arginine Deiminase Type 4/immunology , Protein-Arginine Deiminase Type 4/metabolism , Ventricular Remodeling/drug effects , Ventricular Remodeling/immunology
9.
Sci Rep ; 9(1): 18765, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822739

ABSTRACT

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.


Subject(s)
Hypertension/drug therapy , Hypertrophy, Left Ventricular/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Peroxidase/antagonists & inhibitors , Thioxanthenes/administration & dosage , Angiotensin II/administration & dosage , Angiotensin II/toxicity , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Hypertension/blood , Hypertension/diagnosis , Hypertension/etiology , Hypertrophy, Left Ventricular/blood , Hypertrophy, Left Ventricular/diagnosis , Hypertrophy, Left Ventricular/etiology , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/etiology , Obesity/blood , Obesity/diagnosis , Obesity/etiology , Peroxidase/blood , Peroxidase/metabolism , Severity of Illness Index , Ventricular Remodeling/drug effects , Ventricular Remodeling/immunology
10.
JCI Insight ; 4(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31534054

ABSTRACT

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.


Subject(s)
Cadherins/metabolism , Myocardial Infarction/complications , Myocardium/pathology , Ventricular Remodeling/drug effects , Animals , Bone Marrow Transplantation , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Adhesion/immunology , Disease Models, Animal , Echocardiography , Fibrosis , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/prevention & control , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Male , Mice , Mice, Knockout , Myeloid Cells/immunology , Myocardial Infarction/diagnosis , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardium/immunology , Ventricular Remodeling/immunology
11.
Nat Rev Cardiol ; 15(10): 601-616, 2018 10.
Article in English | MEDLINE | ID: mdl-30181596

ABSTRACT

Cardiac regeneration, that is, restoration of the original structure and function in a damaged heart, differs from tissue repair, in which collagen deposition and scar formation often lead to functional impairment. In both scenarios, the early-onset inflammatory response is essential to clear damaged cardiac cells and initiate organ repair, but the quality and extent of the immune response vary. Immune cells embedded in the damaged heart tissue sense and modulate inflammation through a dynamic interplay with stromal cells in the cardiac interstitium, which either leads to recapitulation of cardiac morphology by rebuilding functional scaffolds to support muscle regrowth in regenerative organisms or fails to resolve the inflammatory response and produces fibrotic scar tissue in adult mammals. Current investigation into the mechanistic basis of homeostasis and restoration of cardiac function has increasingly shifted focus away from stem cell-mediated cardiac repair towards a dynamic interplay of cells composing the less-studied interstitial compartment of the heart, offering unexpected insights into the immunoregulatory functions of cardiac interstitial components and the complex network of cell interactions that must be considered for clinical intervention in heart diseases.


Subject(s)
Cell Communication , Cell Proliferation , Heart Diseases/therapy , Heart Ventricles/pathology , Myocytes, Cardiac/pathology , Regeneration , Stromal Cells/pathology , Ventricular Remodeling , Animals , Heart Diseases/immunology , Heart Diseases/pathology , Heart Diseases/physiopathology , Heart Ventricles/immunology , Humans , Immunotherapy/methods , Myocytes, Cardiac/immunology , Phenotype , Recovery of Function , Regenerative Medicine/methods , Signal Transduction , Stromal Cells/immunology
12.
Am J Physiol Heart Circ Physiol ; 315(5): H1091-H1100, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30074834

ABSTRACT

Doxorubicin (DOX) is a widely used drug for cancer treatment as a chemotherapeutic agent. However, the cellular and integrative mechanism of DOX-induced immunometabolism is unclear. Two-month-old male C57BL/6J mice were divided into high- and low-dose DOX-treated groups with a maintained saline control group. The first group was injected with a high dose of DOX (H-DOX; 15 mg·kg-1·wk-1), and the second group was injected with 7.5 mg·kg-1·wk-1 as a latent low dose of DOX (LL-DOX). H-DOX treatment led to complete mortality in 2 wk and 70% survival in the LL-DOX group compared with the saline control group. Therefore, an additional group of mice was injected with an acute high dose of DOX (AH-DOX) and euthanized at 24 h to compare with LL-DOX and saline control groups. The LL-DOX and AH-DOX groups showed obvious apoptosis and dysfunctional and structural changes in cardiac tissue. Splenic contraction was evident in AH-DOX- and LL-DOX-treated mice, indicating the systems-wide impact of DOX on integrative organs of the spleen, which is essential for cardiac homeostasis and repair. DOX dysregulated splenic-enriched immune-sensitive lipoxygenase and cyclooxygenase in the spleen and left ventricle compared with the saline control group. As a result, lipoxygenase-dependent D- and E-series resolvin precursors, such as 16HDoHE, 4HDoHE, and 12-HEPE, as well as cyclooxygenase-mediated PG species (PGD2, PGE2, and 6-keto-PG2α) were decreased in the left ventricle, suggestive of defective immunometabolism. Both AH-DOX and LL-DOX induced splenic contraction and expansion of red pulp with decreased CD169+ metallophilic macrophages. AH-DOX intoxicated macrophages in the spleen by depleting CD169+ cells in the acute setting and sustained the splenic macrophage loss in the chronic phase in the LL-DOX group. Thus, DOX triggers a vicious cycle of splenocardiac cachexia to facilitate defective immunometabolism and irreversible macrophage toxicity and thereby impaired the inflammation-resolution program. NEW & NOTEWORTHY Doxorubicin (DOX) triggered splenic mass loss and decreased CD169 with germinal center contraction in acute and chronic exposure. Cardiac toxicity of DOX is marked with dysregulation of immunometabolism and thereby impaired resolution of inflammation. DOX suppressed physiological levels of cytokines and chemokines with signs of splenocardiac cachexia.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cachexia/chemically induced , Doxorubicin/toxicity , Heart Diseases/chemically induced , Heart Ventricles/drug effects , Lipoxygenase/metabolism , Macrophages/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Spleen/drug effects , Splenic Diseases/chemically induced , Animals , Apoptosis/drug effects , Cachexia/enzymology , Cachexia/immunology , Cachexia/pathology , Cardiotoxicity , Cytokines/genetics , Cytokines/metabolism , Dose-Response Relationship, Drug , Fibrosis , Gene Expression Regulation, Enzymologic , Heart Diseases/enzymology , Heart Diseases/immunology , Heart Diseases/pathology , Heart Ventricles/enzymology , Heart Ventricles/immunology , Heart Ventricles/pathology , Lipoxygenase/genetics , Macrophages/enzymology , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred C57BL , Myocardium/enzymology , Myocardium/immunology , Myocardium/pathology , Organ Size , Prostaglandin-Endoperoxide Synthases/genetics , Signal Transduction/drug effects , Spleen/enzymology , Spleen/immunology , Spleen/pathology , Splenic Diseases/enzymology , Splenic Diseases/immunology , Splenic Diseases/pathology , Time Factors , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
13.
J Leukoc Biol ; 104(6): 1173-1186, 2018 12.
Article in English | MEDLINE | ID: mdl-30145840

ABSTRACT

Inflammation-limiting nonsteroidal pain relievers magnify myocardial infarction (MI) incidences and increase re-admission events in heart failure (HF) patients. However, the molecular and cellular mechanism of this provocative adverse effect is unclear. Our goal was to determine whether carprofen (CAP) impedes splenic leukocyte-directed acute inflammation-resolving response in cardiac injury. After subacute CAP treatment, mice were subjected to permanent coronary ligation maintaining MI- and naïve-controls. Spleen and left ventricle (LV) leukocytes were quantitated using flow cytometry pre- and 24 h post-MI. The inflammation resolution mediators were quantified using mass spectrometry while splenocardiac apoptosis and leukocyte phagocytosis were measured by immunofluorescence and ImageStream, respectively. Subacute CAP treatment promoted strain and cardiac dysfunction before MI and coronary occlusion showed signs of acute HF in CAP and MI-controls. Subacute CAP-injected mice had pre-activated splenic neutrophils, an over activated "don't eat me" signal (CD47) with reduced total Mϕs (F4/80+ ) and reparative Mϕs (F4/80/Ly6Clo /CD206) compared with control in LV and spleen. Post-MI, CAP pre-activated neutrophils (Ly6G+ ) were intensified and reduced reparative neutrophils (Ly6G+ /CD206+ ) and Mϕs (F4/80/Ly6Clo ) in LV was indicative of non-resolving inflammation compared with MI-control. Subacute CAP treatment deferred neutrophil phagocytosis functions in the spleen and LV and was more evident post-MI compared with MI-control. CAP pre-activated splenic neutrophils that tailored the Mϕ phagocytosis thereby increased splenocardiac leukocyte death. CAP over amplified COX-1 and COX-2 compared with MI-control and failed to limit prostaglandins and thromboxane in post-MI setting. Further, CAP reduced cardiac-protective epoxyeicosatrienoic acids and over amplified pyrogenic inflammatory cytokines and reduced reparative cytokines, thereby non-resolving inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Carbazoles/toxicity , Heart Ventricles/drug effects , Inflammation/chemically induced , Leukocytes/drug effects , Myocardial Infarction/physiopathology , Spleen/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/physiology , Eicosanoids/metabolism , Heart Failure/etiology , Heart Ventricles/immunology , Heart Ventricles/physiopathology , Inflammation/etiology , Inflammation Mediators/metabolism , Leukocytes/immunology , Macrophage Activation/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Neutrophil Activation/drug effects , Phagocytosis/drug effects , Prostaglandins/metabolism , Spleen/immunology , Spleen/physiopathology
14.
JCI Insight ; 3(11)2018 06 07.
Article in English | MEDLINE | ID: mdl-29875326

ABSTRACT

Despite the long-standing recognition that the immune response to acute myocardial injury contributes to adverse left ventricular (LV) remodeling, it has not been possible to effectively target this clinically. Using 2 different in vivo models of acute myocardial injury, we show that pirfenidone confers beneficial effects in the murine heart through an unexpected mechanism that depends on cardiac B lymphocytes. Naive hearts contained a large population of CD19+CD11b-CD23-CD21-IgD+IgMlo lymphocytes, and 2 smaller populations of CD19+CD11b+ B1a and B1b cells. In response to tissue injury, there was an increase in neutrophils, monocytes, macrophages, as well as an increase in CD19+ CD11b- B lymphocytes. Treatment with pirfenidone had no effect on the number of neutrophils, monocytes, or macrophages, but decreased CD19+CD11b- lymphocytes. B cell depletion abrogated the beneficial effects of pirfenidone. In vitro studies demonstrated that stimulation with lipopolysaccharide and extracts from necrotic cells activated CD19+ lymphocytes through a TIRAP-dependent pathway. Treatment with pirfenidone attenuated this activation of B cells. These findings reveal a previously unappreciated complexity of myocardial B lymphocytes within the inflammatory infiltrate triggered by cardiac injury and suggest that pirfenidone exerts beneficial effects in the heart through a unique mechanism that involves modulation of cardiac B lymphocytes.


Subject(s)
B-Lymphocyte Subsets/immunology , Heart Ventricles/drug effects , Myocardial Infarction/immunology , Pyridones/administration & dosage , Ventricular Remodeling/drug effects , Animals , B-Lymphocyte Subsets/drug effects , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/immunology , Disease Models, Animal , Female , Heart Ventricles/immunology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Lymphocyte Activation/drug effects , Lymphocyte Depletion/methods , Mice , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/cytology , Myocardium/immunology , Myocardium/pathology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Ventricular Remodeling/immunology
15.
J Biochem Mol Toxicol ; 32(2)2018 Feb.
Article in English | MEDLINE | ID: mdl-29315967

ABSTRACT

Doxorubicin (DXR) is a highly effective drug for chemotherapy. However, cardiotoxicity reduces its clinical utility in humans. The present study aimed to assess the ameliorative effect of curcumin against DXR-induced cardiotoxicity in rats. Rats were subjected to oral treatment of curcumin (100 and 200 mg/kg body weight) for 7 days. Cardiotoxicity was induced by single intraperitoneal injection of DXR (40 mg/kg body weight) on the 5th day and the rats sacrificed on 8th day. Curcumin ameliorated DXR-induced lipid peroxidation, glutathione depletion, decrease in antioxidant (superoxide dismutase, catalase, and glutathione peroxidase) enzyme activities, and cardiac toxicity markers (CK-MB, LDH, and cTn-I). Curcumin also attenuated activities of Caspase-3, cyclooxygenase-2, inducible nitric oxide synthase, and levels of nuclear factor kappa-B, tumor necrosis factor-α, and interleukin-1ß, and cardiac tissue damages that were induced by DXR. Moreover, curcumin decreased the expression of 8-OHdG and 3,3'-dityrosine. This study demonstrated that curcumin has a multi-cardioprotective effect due to its antioxidant, anti-inflammatory, and antiapoptotic properties.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Cardiotonic Agents/therapeutic use , Cardiotoxicity/prevention & control , Curcumin/therapeutic use , Heart Ventricles/drug effects , Oxidative Stress/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antibiotics, Antineoplastic/adverse effects , Antioxidants/administration & dosage , Antioxidants/adverse effects , Antioxidants/therapeutic use , Biomarkers/metabolism , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/adverse effects , Cardiotoxicity/immunology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Curcumin/administration & dosage , Curcumin/adverse effects , DNA Damage/drug effects , Dose-Response Relationship, Drug , Doxorubicin/adverse effects , Glutathione/metabolism , Heart Ventricles/immunology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Lipid Peroxidation/drug effects , Male , Oxidation-Reduction , Random Allocation , Rats, Wistar
16.
J Immunol ; 200(2): 523-537, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29229678

ABSTRACT

Sarcoplasmic/endoplasmic reticulum Ca2+ adenosine triphosphatase (SERCA)2a, a critical regulator of calcium homeostasis, is known to be decreased in heart failure. Patients with myocarditis or dilated cardiomyopathy develop autoantibodies to SERCA2a suggesting that they may have pathogenetic significance. In this report, we describe epitope mapping analysis of SERCA2a in A/J mice that leads us to make five observations: 1) SERCA2a contains multiple T cell epitopes that induce varying degrees of myocarditis. One epitope, SERCA2a 971-990, induces widespread atrial inflammation without affecting noncardiac tissues; the cardiac abnormalities could be noninvasively captured by echocardiography, electrocardiography, and magnetic resonance microscopy imaging. 2) SERCA2a 971-990-induced disease was associated with the induction of CD4 T cell responses and the epitope preferentially binds MHC class II/IAk rather than IEk By creating IAk/and IEk/SERCA2a 971-990 dextramers, the T cell responses were determined by flow cytometry to be Ag specific. 3) SERCA2a 971-990-sensitized T cells produce both Th1 and Th17 cytokines. 4) Animals immunized with SERCA2a 971-990 showed Ag-specific Abs with enhanced production of IgG2a and IgG2b isotypes, suggesting that SERCA2a 971-990 can potentially act as a common epitope for both T cells and B cells. 5) Finally, SERCA2a 971-990-sensitized T cells were able to transfer disease to naive recipients. Together, these data indicate that SERCA2a is a critical autoantigen in the mediation of atrial inflammation in mice and that our model may be helpful to study the inflammatory events that underlie the development of conditions such as atrial fibrillation in humans.


Subject(s)
Epitope Mapping , Epitopes/immunology , Myocarditis/immunology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology , Alleles , Animals , Bacterial Proteins , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Epitope Mapping/methods , Epitopes, B-Lymphocyte/immunology , Fluorescent Antibody Technique , Gene Expression , Heart Atria/immunology , Heart Atria/metabolism , Heart Atria/pathology , Heart Ventricles/immunology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunodominant Epitopes/immunology , Immunohistochemistry , Mice , Mice, Inbred Strains , Myocarditis/diagnostic imaging , Myocarditis/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Peptides/immunology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
17.
Cardiovasc Res ; 114(2): 291-299, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29186414

ABSTRACT

Aims: Cardiac inflammation has been suggested to be regulated by the sympathetic nervous system (SNS). However, due to the lack of methodology to surgically eliminate the myocardial SNS in mice, neuronal control of cardiac inflammation remains ill-defined. Here, we report a procedure for local cardiac sympathetic denervation in mice and tested its effect in a mouse model of heart failure post-myocardial infarction. Methods and results: Upon preparation of the carotid bifurcation, the right and the left superior cervical ganglia were localized and their pre- and postganglionic branches dissected before removal of the ganglion. Ganglionectomy led to an almost entire loss of myocardial sympathetic innervation in the left ventricular anterior wall. When applied at the time of myocardial infarction (MI), cardiac sympathetic denervation did not affect acute myocardial damage and infarct size. In contrast, cardiac sympathetic denervation significantly attenuated chronic consequences of MI, including myocardial inflammation, myocyte hypertrophy, and overall cardiac dysfunction. Conclusion: These data suggest a critical role for local sympathetic control of cardiac inflammation. Our model of myocardial sympathetic denervation in mice should prove useful to further dissect the molecular mechanisms underlying cardiac neural control.


Subject(s)
Ganglionectomy , Heart Failure/prevention & control , Heart Ventricles/innervation , Myocardial Infarction/complications , Myocarditis/prevention & control , Myocardium , Superior Cervical Ganglion/surgery , Animals , Heart Failure/immunology , Heart Failure/pathology , Heart Failure/physiopathology , Heart Ventricles/immunology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , Male , Mice, Inbred C57BL , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/physiopathology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Neuroimmunomodulation , Superior Cervical Ganglion/physiopathology , Ventricular Function, Left
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.
Clin Cardiol ; 40(9): 626-632, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28475816

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of confirmed HF cases in the United States. However, there are no highly effective evidence-based treatments currently available for these patients. Inflammation correlates positively with adverse outcomes in HF patients. Interleukin (IL)-1, a prototypical inflammatory cytokine, has been implicated as a driver of diastolic dysfunction in preclinical animal models and a pilot clinical trial. The Diastolic Heart Failure Anakinra Response Trial 2 (D-HART2) is a phase 2, 2:1 randomized, double-blind, placebo-controlled clinical trial that will test the hypothesis that IL-1 blockade with anakinra (recombinant human IL-1 receptor antagonist) improves (1) cardiorespiratory fitness, (2) objective evidence of diastolic dysfunction, and (3) elevated inflammation in patients with HFpEF (http://www.ClinicalTrials.gov NCT02173548). The co-primary endpoints will be placebo-corrected interval changes in peak oxygen consumption and ventilatory efficiency at week 12. In addition, secondary and exploratory analyses will investigate the effects of IL-1 blockade on cardiac structure and function, systemic inflammation, endothelial function, quality of life, body composition, nutritional status, and clinical outcomes. The D-HART2 clinical trial will add to the growing body of evidence on the role of inflammation in cardiovascular disease, specifically focusing on patients with HFpEF.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiovascular Agents/therapeutic use , Heart Failure/drug therapy , Heart Ventricles/drug effects , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1/antagonists & inhibitors , Ventricular Function, Left , Anti-Inflammatory Agents/adverse effects , Cardiorespiratory Fitness , Cardiovascular Agents/adverse effects , Clinical Protocols , Cross-Over Studies , Double-Blind Method , Exercise Tolerance/drug effects , Heart Failure/diagnosis , Heart Failure/immunology , Heart Failure/physiopathology , Heart Ventricles/immunology , Heart Ventricles/physiopathology , Humans , Interleukin 1 Receptor Antagonist Protein/adverse effects , Interleukin-1/immunology , Pilot Projects , Recovery of Function , Research Design , Stroke Volume , Time Factors , Treatment Outcome , Virginia
20.
Biochem Pharmacol ; 138: 193-204, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28450224

ABSTRACT

Pulmonary hypertension is still not curable and the available current therapies can only alleviate symptoms without hindering the progression of disease. The present study was directed to investigate the possible modulatory effect of pinocembrin on endothelial progenitor cells transplanted in monocrotaline-induced pulmonary hypertension in rats. Pulmonary hypertension was induced by a single subcutaneous injection of monocrotaline (60mg/kg). Endothelial progenitor cells were in vitro preconditioned with pinocembrin (25mg/L) for 30min before being i.v. injected into rats 2weeks after monocrotaline administration. Four weeks after monocrotaline administration, blood pressure, electrocardiography and right ventricular systolic pressure were recorded. Rats were sacrificed and serum was separated for determination of endothelin-1 and asymmetric dimethylarginine levels. Right ventricles and lungs were isolated for estimation of tumor necrosis factor-alpha and transforming growth factor-beta contents as well as caspase-3 activity. Moreover, protein expression of matrix metalloproteinase-9 and endothelial nitric oxide synthase in addition to myocardial connexin-43 was assessed. Finally, histological analysis of pulmonary arteries, cardiomyocyte cross-sectional area and right ventricular hypertrophy was performed and cryosections were done for estimation of cell homing. Preconditioning with pinocembrin provided a significant improvement in endothelial progenitor cells' effect towards reducing monocrotaline-induced elevation of inflammatory, fibrogenic and apoptotic markers. Furthermore, preconditioned cells induced a significant amelioration of endothelial markers and cell homing and prevented monocrotaline-induced changes in right ventricular function and histological analysis compared with native cells alone. In conclusion, pinocembrin significantly improves the therapeutic efficacy of endothelial progenitor cells in monocrotaline-induced pulmonary hypertension in rats.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bone Marrow Cells/drug effects , Disease Models, Animal , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/transplantation , Flavanones/therapeutic use , Hypertension, Pulmonary/surgery , Animals , Apoptosis , Biomarkers/blood , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Transplantation/adverse effects , Cells, Cultured , Cytokines/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Graft Rejection/prevention & control , Heart Ventricles/immunology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Pulmonary Artery/pathology , Random Allocation , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...