Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
Nat Commun ; 15(1): 3637, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38684665

In contrast to adult mammals, adult zebrafish can fully regenerate injured cardiac tissue, and this regeneration process requires an adequate and tightly controlled immune response. However, which components of the immune response are required during regeneration is unclear. Here, we report positive roles for the antigen presentation-adaptive immunity axis during zebrafish cardiac regeneration. We find that following the initial innate immune response, activated endocardial cells (EdCs), as well as immune cells, start expressing antigen presentation genes. We also observe that T helper cells, a.k.a. Cd4+ T cells, lie in close physical proximity to these antigen-presenting EdCs. We targeted Major Histocompatibility Complex (MHC) class II antigen presentation by generating cd74a; cd74b mutants, which display a defective immune response. In these mutants, Cd4+ T cells and activated EdCs fail to efficiently populate the injured tissue and EdC proliferation is significantly decreased. cd74a; cd74b mutants exhibit additional defects in cardiac regeneration including reduced cardiomyocyte dedifferentiation and proliferation. Notably, Cd74 also becomes activated in neonatal mouse EdCs following cardiac injury. Altogether, these findings point to positive roles for antigen presentation during cardiac regeneration, potentially involving interactions between activated EdCs, classical antigen-presenting cells, and Cd4+ T cells.


Antigen Presentation , Heart Injuries , Histocompatibility Antigens Class II , Regeneration , Zebrafish , Animals , Regeneration/immunology , Antigen Presentation/immunology , Heart Injuries/immunology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/genetics , Mice , CD4-Positive T-Lymphocytes/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Cell Proliferation , Immunity, Innate , Heart/physiopathology , Heart/physiology , Mutation , Adaptive Immunity , Animals, Genetically Modified
2.
Mol Med Rep ; 24(5)2021 Nov.
Article En | MEDLINE | ID: mdl-34476499

A unique region of human parvovirus B19 virus­VP1 (B19V­VP1u) has been linked to a variety of cardiac disorders. However, the precise role of B19V­VP1u in inducing cardiac injury remains unknown. The present study investigated the effects of B19V­VP1u and different regions of B19V­VP1u, including B19V­VP1uA (residues 1­60), B19V­VP1uB (residues 61­129), B19V­VP1uC (residues 130­195) and B19V­VP1uD (residues 196­227), on inducing cardiac injury in naïve mice by zymography, immunoblotting, H&E staining and cytokine immunoassay. A significantly higher MMP­9/MMP­2 ratio and increased levels of inflammatory cytokines, including IL­6 and IL­1ß, were detected in the left ventricles of the mice injected with B19V­non­structural protein 1 (B19V­NS1) and B19V­VP1u, accompanied by increased expression levels of phosphorylated (p­)ERK and p­P38. Significantly upregulated expression levels of atrial natriuretic peptide (ANP), heart­type fatty acid­binding protein (H­FABP) and creatine kinase isoenzyme­MB (CK­MB), which are well­known cardiac injury markers, as well as increased infiltration of lymphocytes, were detected in the left ventricles of the mice injected with B19V­VP1, B19V­NS1 and B19V­VP1u. Moreover, a significantly higher MMP­9/MMP­2 ratio and increased levels of IL­6 and IL­1ß were observed in the left ventricles of the mice injected with B19V­VP1u, B19V­VP1u­A, B19V­VP1u­B and B19V­VP1u­C, accompanied by upregulated p­ERK and p­P38 expression. Notably, significantly lower levels of IL­6 and IL­1ß were observed in the left ventricles of the mice injected with B19V­VP1uD. Furthermore, significantly increased ANP, H­FABP and CK­MB expression levels were detected in the left ventricles of the mice injected with B19V­VP1u, B19V­VP1u­A and B19V­VP1u­B, along with enhanced infiltration of lymphocytes. Significantly higher serum IL­1ß, IL­6, TNF­α and IFN­Î³ levels were also detected in the mice injected with B19V­VP1u, B19V­VP1u­A and B19V­VP1u­B. To the best of our knowledge, the findings of the present study were the first to demonstrate that the N­terminal region (residues 1­129) of B19V­VP1u induces an increase in the levels of cardiac injury markers, thus providing evidence for understanding the possible functional regions within B19V­VP1u.


Capsid Proteins/immunology , Heart Injuries/immunology , Parvoviridae Infections/complications , Parvovirus B19, Human/immunology , Animals , Capsid Proteins/genetics , Capsid Proteins/isolation & purification , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Heart Injuries/blood , Heart Injuries/pathology , Heart Injuries/virology , Host-Pathogen Interactions/immunology , Humans , Mice , Parvoviridae Infections/blood , Parvoviridae Infections/immunology , Parvoviridae Infections/virology , Parvovirus B19, Human/genetics , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Signal Transduction/immunology
3.
Circ Res ; 128(11): 1766-1779, 2021 05 28.
Article En | MEDLINE | ID: mdl-34043424

Cardiac injury remains a major cause of morbidity and mortality worldwide. Despite significant advances, a full understanding of why the heart fails to fully recover function after acute injury, and why progressive heart failure frequently ensues, remains elusive. No therapeutics, short of heart transplantation, have emerged to reliably halt or reverse the inexorable progression of heart failure in the majority of patients once it has become clinically evident. To date, most pharmacological interventions have focused on modifying hemodynamics (reducing afterload, controlling blood pressure and blood volume) or on modifying cardiac myocyte function. However, important contributions of the immune system to normal cardiac function and the response to injury have recently emerged as exciting areas of investigation. Therapeutic interventions aimed at harnessing the power of immune cells hold promise for new treatment avenues for cardiac disease. Here, we review the immune response to heart injury, its contribution to cardiac fibrosis, and the potential of immune modifying therapies to affect cardiac repair.


Heart Failure/therapy , Heart Injuries/therapy , Immunotherapy/methods , Adaptive Immunity , B-Lymphocytes/physiology , Bioengineering , Cytokines/metabolism , Disease Progression , Eosinophils/physiology , Fibroblasts/physiology , Fibrosis , Heart Failure/etiology , Heart Failure/immunology , Heart Injuries/immunology , Humans , Immunotherapy, Adoptive , Macrophages/physiology , Mast Cells/physiology , Monocytes/physiology , Myocardium/pathology , Myocytes, Cardiac/physiology , Neutrophils/physiology , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , T-Lymphocytes/transplantation
4.
Signal Transduct Target Ther ; 6(1): 79, 2021 02 22.
Article En | MEDLINE | ID: mdl-33612829

The response of immune cells in cardiac injury is divided into three continuous phases: inflammation, proliferation and maturation. The kinetics of the inflammatory and proliferation phases directly influence the tissue repair. In cardiac homeostasis, cardiac tissue resident macrophages (cTMs) phagocytose bacteria and apoptotic cells. Meanwhile, NK cells prevent the maturation and transport of inflammatory cells. After cardiac injury, cTMs phagocytose the dead cardiomyocytes (CMs), regulate the proliferation and angiogenesis of cardiac progenitor cells. NK cells prevent the cardiac fibrosis, and promote vascularization and angiogenesis. Type 1 macrophages trigger the cardioprotective responses and promote tissue fibrosis in the early stage. Reversely, type 2 macrophages promote cardiac remodeling and angiogenesis in the late stage. Circulating macrophages and neutrophils firstly lead to chronic inflammation by secreting proinflammatory cytokines, and then release anti-inflammatory cytokines and growth factors, which regulate cardiac remodeling. In this process, dendritic cells (DCs) mediate the regulation of monocyte and macrophage recruitment. Recruited eosinophils and Mast cells (MCs) release some mediators which contribute to coronary vasoconstriction, leukocyte recruitment, formation of new blood vessels, scar formation. In adaptive immunity, effector T cells, especially Th17 cells, lead to the pathogenesis of cardiac fibrosis, including the distal fibrosis and scar formation. CMs protectors, Treg cells, inhibit reduce the inflammatory response, then directly trigger the regeneration of local progenitor cell via IL-10. B cells reduce myocardial injury by preserving cardiac function during the resolution of inflammation.


Heart Injuries/immunology , Myocardial Infarction/immunology , Myocardial Reperfusion Injury/immunology , Myocardium/immunology , Heart Injuries/pathology , Heart Injuries/therapy , Homeostasis/immunology , Humans , Killer Cells, Natural/immunology , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/therapy , Myocytes, Cardiac/immunology , Neutrophils/immunology , Regeneration/immunology
5.
Front Immunol ; 11: 802, 2020.
Article En | MEDLINE | ID: mdl-32431711

Innate lymphoid cells (ILCs), including ILC1s, ILC2s, and ILC3s, play critical roles in regulating immunity, inflammation, and tissue homeostasis. However, limited attention is focused on the unique phenotype of ILCs in the heart tissue. In this study, we analyzed the ILC subsets in the heart by flow cytometry and found that ILC2s were the dominant population of ILCs, while a lower proportion of type 1 ILCs (including ILC1 and NK cells) and merely no ILC3s in the heart tissue of mice. Our results show that ILC2 development kinetically peaked in heart ILC2s at the age of 4 weeks after birth and later than lung ILC2s. By conducting parabiosis experiment, we show that heart ILC2s are tissue resident cells and minimally replaced by circulating cells. Notably, heart ILC2s have unique phenotypes, such as lower expression of ICOS, CD25 (IL-2Rα), and Ki-67, higher expression of Sca-1 and GATA3, and stronger ability to produce IL-4 and IL-13. In doxorubicin-induced myocardial necroptosis model of mouse heart tissue, IL-33 mRNA expression level and ILC2s were remarkably increased. In addition, IL-4 production by heart ILC2s, but not lung ILC2s, was also dramatically increased after doxorubicin treatment. Our results demonstrate that heart-resident ILC2s showed tissue-specific phenotypes and rapidly responded to heart injury. Thus, further studies are warranted to explore the potential for IL-33-elicited ILC2s response as therapeutics for attenuating heart damage.


Immunity, Innate , Lymphocytes/immunology , Myocardium/immunology , Phenotype , Animals , Disease Models, Animal , Doxorubicin/adverse effects , Female , Heart Injuries/chemically induced , Heart Injuries/immunology , Heart Injuries/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , Interleukin-4/metabolism , Lung/immunology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Necroptosis/drug effects , Parabiosis , Signal Transduction/drug effects
7.
Front Immunol ; 11: 92, 2020.
Article En | MEDLINE | ID: mdl-32117249

Cardiovascular disease is the leading cause of worldwide mortality. Intravital microscopy has provided unprecedented insight into leukocyte biology by enabling the visualization of dynamic responses within living organ systems at the cell-scale. The heart presents a uniquely dynamic microenvironment driven by periodic, synchronous electrical conduction leading to rhythmic contractions of cardiomyocytes, and phasic coronary blood flow. In addition to functions shared throughout the body, immune cells have specific functions in the heart including tissue-resident macrophage-facilitated electrical conduction and rapid monocyte infiltration upon injury. Leukocyte responses to cardiac pathologies, including myocardial infarction and heart failure, have been well-studied using standard techniques, however, certain questions related to spatiotemporal relationships remain unanswered. Intravital imaging techniques could greatly benefit our understanding of the complexities of in vivo leukocyte behavior within cardiac tissue, but these techniques have been challenging to apply. Different approaches have been developed including high frame rate imaging of the beating heart, explantation models, micro-endoscopy, and mechanical stabilization coupled with various acquisition schemes to overcome challenges specific to the heart. The field of cardiac science has only begun to benefit from intravital microscopy techniques. The current focused review presents an overview of leukocyte responses in the heart, recent developments in intravital microscopy for the murine heart, and a discussion of future developments and applications for cardiovascular immunology.


Heart Injuries/immunology , Intravital Microscopy/methods , Animals , Leukocytes , Mice , Microscopy, Fluorescence, Multiphoton/methods , Myocardial Infarction , Myocytes, Cardiac
8.
Biochem Biophys Res Commun ; 523(3): 580-587, 2020 03 12.
Article En | MEDLINE | ID: mdl-31941605

Sepsis induces critical myocardial dysfunction, resulting in an increased mortality. Gracillin (GRA) is a natural steroidal saponin, showing strong capacities of anti-inflammation, but its pharmacological effects on lipopolysaccharide (LPS)-induced acute cardiac injury still remain unclear. In this study, we attempted to explore if GRA was effective to attenuate cardiac injury in LPS-challenged mice and the underlying mechanisms. First, we found that GRA treatments markedly up-regulated the expression of miR-29a in cardiomyocytes. LPS-induced cytotoxicity in cardiomyocytes was significantly alleviated by GRA treatment, as evidenced by the improved cell viability and reduced lactate dehydrogenase (LDH) release. In addition, LPS-triggered apoptotic cell death was clearly ameliorated in cardiomyocytes co-treated with GRA. Notably, LPS-exposed cells showed significantly reduced expression of miR-29a, while being rescued by GRA treatment. In vivo, LPS apparently impaired cardiac function in mice, which was, however, alleviated by GRA administration. In addition, GRA markedly attenuated apoptosis in hearts of LPS-challenged mice by decreasing the expression of cleaved Caspase-3. LPS-triggered inflammatory response in cardiac tissues was also suppressed by GRA through blocking nuclear factor κB (NF-κB) signaling pathway. We also found that miR-29a expression was highly reduced in hearts of LPS-treated mice but was rescued by GRA pretreatment. Besides, miR-29a mimic alleviated LPS-induced apoptosis and inflammation in cardiomyocytes; however, LPS-caused effects were further accelerated by miR-29a. Of note, the protective effects of GRA on LPS-injured cardiac tissues were significantly abrogated by miR-29a suppression. In conclusion, our findings demonstrated that GRA exerted an effective role against LPS-induced acute cardiac injury through impeding apoptosis and inflammation regulated by miR-29a.


Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Heart Injuries/drug therapy , Inflammation/drug therapy , MicroRNAs/genetics , Spirostans/therapeutic use , Animals , Cell Line , Down-Regulation/drug effects , Heart Injuries/genetics , Heart Injuries/immunology , Inflammation/genetics , Inflammation/immunology , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/immunology , Rats
9.
Cells ; 10(1)2020 12 31.
Article En | MEDLINE | ID: mdl-33396359

The immune system plays a pivotal role in the initiation, development and resolution of inflammation following insult or damage to organs. The heart is a vital organ which supplies nutrients and oxygen to all parts of the body. Heart failure (HF) has been conventionally described as a disease associated with cardiac tissue damage caused by systemic inflammation, arrhythmia and conduction defects. Cardiac inflammation and subsequent tissue damage is orchestrated by the infiltration and activation of various immune cells including neutrophils, monocytes, macrophages, eosinophils, mast cells, natural killer cells, and T and B cells into the myocardium. After tissue injury, monocytes and tissue-resident macrophages undergo marked phenotypic and functional changes, and function as key regulators of tissue repair, regeneration and fibrosis. Disturbance in resident macrophage functions such as uncontrolled production of inflammatory cytokines, growth factors and inefficient generation of an anti-inflammatory response or unsuccessful communication between macrophages and epithelial and endothelial cells and fibroblasts can lead to aberrant repair, persistent injury, and HF. Therefore, in this review, we discuss the role of cardiac macrophages on cardiac inflammation, tissue repair, regeneration and fibrosis.


Fibrosis/metabolism , Heart Injuries/metabolism , Macrophages/metabolism , Regeneration , Animals , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/metabolism , Cytokines/metabolism , Diabetic Cardiomyopathies/immunology , Diabetic Cardiomyopathies/metabolism , Fibrosis/immunology , Heart/physiopathology , Heart Injuries/immunology , Homeostasis , Humans , Hypertension/immunology , Hypertension/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/cytology , Mitochondria/immunology , Myocardium/cytology , Myocardium/immunology
10.
Immunopharmacol Immunotoxicol ; 41(6): 630-643, 2019 Dec.
Article En | MEDLINE | ID: mdl-31724456

Aim: Renin-angiotensin system (RAS) is thought to have a noticeable effect in the pathophysiological injury in multiple organs by inducing different cellular and molecular reactions. The objective of the current study is to investigate the possible protective effects of perindopril against lipopolysaccharide (LPS)-induced cardiopulmonary oxidative and inflammatory damage in rats. Methods: To achieve this goal, animals were randomly divided into six groups: normal group, LPS group (3 mg/kg, i.p., single dose), perindopril-LPS treated group (1 mg/kg/day, i.p.), perindopril-LPS treated group (2 mg/kg/day, i.p.), and two perindopril negative groups (perindopril 1 or 2 mg/kg/day, i.p.) alone for seven days. Lungs and hearts tissue angiotensin II (Ang-II), angiotensin-1-7 (Ang-1-7), and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase were assessed using ELISA. Nuclear factor kappa-B-p65 (NF-κB-p65) was assessed using real time PCR, while protein kinase B (Akt) was evaluated by Western blot analysis. Furthermore, oxidative stress biomarkers and myeloperoxidase (MPO) enzyme were evaluated spectrophotometrically. Tissues inducible and endothelial nitric oxide synthases (iNOS and eNOS) were assessed immunohistochemically. Histopathological study was carried out to confirm our results. Results: LPS-intoxicated rats significantly elevated Ang-II, NF-κB-p65, Akt, and iNOS levels, coupled with significant down-regulation of Ang-1-7 and eNOS levels and corrected the oxidative stress biomarkers. Perindopril administration significantly attenuated the disturbances induced by LPS in a dose-dependent manner. Conclusion: Perindopril mitigates LPS-induced heart and lung damage through modulation of RAS, iNOS/eNOS, Akt, and NF-κB-p65 signaling pathways.


Heart Injuries/drug therapy , Lipopolysaccharides/toxicity , Lung Injury/drug therapy , Oxidative Stress/drug effects , Perindopril/pharmacology , Renin-Angiotensin System/drug effects , Animals , Heart Injuries/chemically induced , Heart Injuries/immunology , Heart Injuries/pathology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Lung Injury/chemically induced , Lung Injury/immunology , Lung Injury/pathology , Male , Oxidative Stress/immunology , Rats , Rats, Wistar , Renin-Angiotensin System/immunology , Signal Transduction/drug effects , Signal Transduction/immunology
11.
Proc Natl Acad Sci U S A ; 116(37): 18455-18465, 2019 09 10.
Article En | MEDLINE | ID: mdl-31451669

The adult mammalian heart has limited capacity for regeneration following injury, whereas the neonatal heart can readily regenerate within a short period after birth. To uncover the molecular mechanisms underlying neonatal heart regeneration, we compared the transcriptomes and epigenomes of regenerative and nonregenerative mouse hearts over a 7-d time period following myocardial infarction injury. By integrating gene expression profiles with histone marks associated with active or repressed chromatin, we identified transcriptional programs underlying neonatal heart regeneration, and the blockade to regeneration in later life. Our results reveal a unique immune response in regenerative hearts and a retained embryonic cardiogenic gene program that is active during neonatal heart regeneration. Among the unique immune factors and embryonic genes associated with cardiac regeneration, we identified Ccl24, which encodes a cytokine, and Igf2bp3, which encodes an RNA-binding protein, as previously unrecognized regulators of cardiomyocyte proliferation. Our data provide insights into the molecular basis of neonatal heart regeneration and identify genes that can be modulated to promote heart regeneration.


Animals, Newborn/physiology , Heart/physiology , Histone Code/physiology , Regeneration/physiology , Transcriptome/physiology , Animals , Animals, Newborn/growth & development , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Developmental , Heart Injuries/genetics , Heart Injuries/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Myocardial Infarction/genetics , Regeneration/genetics , Transcriptome/genetics
14.
Sci Rep ; 6: 26489, 2016 05 26.
Article En | MEDLINE | ID: mdl-27225830

NLRP3 inflammasomes recognize non-microbial danger signals and induce release of proinflammatory cytokine interleukin (IL)-1ß, leading to sterile inflammation in cardiovascular disease. Because sterile inflammation is involved in doxorubicin (Dox)-induced cardiotoxicity, we investigated the role of NLRP3 inflammasomes in Dox-induced cardiotoxicity. Cardiac dysfunction and injury were induced by low-dose Dox (15 mg/kg) administration in NLRP3-deficient (NLRP3(-/-)) mice but not in wild-type (WT) and IL-1ß(-/-) mice, indicating that NLRP3 deficiency enhanced the susceptibility to Dox-induced cardiotoxicity independent of IL-1ß. Although the hearts of WT and NLRP3(-/-) mice showed no significant difference in inflammatory cell infiltration, macrophages were the predominant inflammatory cells in the hearts, and cardiac IL-10 production was decreased in Dox-treated NLRP3(-/-) mice. Bone marrow transplantation experiments showed that bone marrow-derived cells contributed to the exacerbation of Dox-induced cardiotoxicity in NLRP3(-/-) mice. In vitro experiments revealed that NLRP3 deficiency decreased IL-10 production in macrophages. Furthermore, adeno-associated virus-mediated IL-10 overexpression restored the exacerbation of cardiotoxicity in the NLRP3(-/-) mice. These results demonstrated that NLRP3 regulates macrophage IL-10 production and contributes to the pathophysiology of Dox-induced cardiotoxicity, which is independent of IL-1ß. Our findings identify a novel role of NLRP3 and provided new insights into the mechanisms underlying Dox-induced cardiotoxicity.


Doxorubicin/toxicity , Heart Injuries/immunology , Interleukin-10/metabolism , Interleukin-1beta/genetics , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Animals , Bone Marrow Transplantation/adverse effects , Cardiotoxicity , Cells, Cultured , Disease Models, Animal , Heart Injuries/chemically induced , Heart Injuries/genetics , Humans , Interleukin-1beta/deficiency , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
15.
Cell Res ; 25(10): 1137-51, 2015 Oct.
Article En | MEDLINE | ID: mdl-26358185

Cardiac injury in neonatal 1-day-old mice stimulates a regenerative response characterized by reactive cardiomyocyte proliferation, which is distinguished from the fibrotic repair process in adults. Acute inflammation occurs immediately after heart injury and has generally been believed to exert a negative effect on heart regeneration by promoting scar formation in adults; however, little is known about the role of acute inflammation in the cardiac regenerative response in neonatal mice. Here, we show that acute inflammation induced cardiomyocyte proliferation after apical intramyocardial microinjection of immunogenic zymosan A particles into the neonatal mouse heart. We also found that cardiac injury-induced regenerative response was suspended after immunosuppression in neonatal mice, and that cardiomyocytes could not be reactivated to proliferate after neonatal heart injury in the absence of interleukin-6 (IL-6). Furthermore, cardiomyocyte-specific deletion of signal transducer and activator of transcription 3 (STAT3), the major downstream effector of IL-6 signaling, decreased reactive cardiomyocyte proliferation after apical resection. Our results indicate that acute inflammation stimulates the regenerative response in neonatal mouse heart, and suggest that modulation of inflammatory signals might have important implications in cardiac regenerative medicine.


Heart Injuries/immunology , Heart/physiology , Inflammation/immunology , Interleukin-6/immunology , Myocytes, Cardiac/cytology , Regeneration , Acute Disease , Animals , Cell Proliferation , Female , Heart/growth & development , Heart/physiopathology , Heart Injuries/physiopathology , Immunosuppression Therapy , Inflammation/physiopathology , Male , Mice , Myocytes, Cardiac/immunology , STAT3 Transcription Factor/immunology , Signal Transduction
16.
Chem Biol Interact ; 233: 46-55, 2015 May 25.
Article En | MEDLINE | ID: mdl-25557508

Sepsis is a cluster of heterogeneous syndromes associated with progressive endotoxemic developments, ultimately leading to damage of multiple organs, including the heart. This study is to investigate the effects of apigenin on heart injury in lipopolysaccharide-induced endotoxemic rat model. Normal Wistar rats were randomly divided into four groups: control group, LPS group (15 mg/kg), LPS plus apigenin groups with different apigenin doses (50 mg/kg, 100 mg/kg). Serum levels of creatine kinase (CK), lactate dehydrogenase (LDH), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß) were measured after the rats were sacrificed. SphK1/S1P signaling pathway proteins, cleaved caspase-3, cleaved caspase-9, Bax and Bcl-2 in heart were measured by Western blot. In vitro, we evaluated the protective effect of apigenin on rat embryonic heart-derived myogenic cell line H9c2 induced by LPS. Apigenin decreased serum levels of CK-MB, LDH, TNF-α, IL-6, IL-1ß. SphK1/S1P signaling pathway proteins, cleaved caspase-3, cleaved caspase-9, Bax in heart were found inhibited and Bcl-2 increased in the apigenin groups in vivo. In addition, apigenin inhibited intracellular calcium, the MAPK pathway and SphK1/S1P signaling pathway in vitro. Apigenin exerts pronounced cardioprotection in rats subjected to LPS likely through suppressing myocardial apoptosis and inflammation by inhibiting the SphK1/S1P signaling pathway.


Anti-Inflammatory Agents/therapeutic use , Apigenin/therapeutic use , Endotoxemia/complications , Heart Injuries/drug therapy , Heart Injuries/etiology , Lysophospholipids/immunology , Phosphotransferases (Alcohol Group Acceptor)/immunology , Sphingosine/analogs & derivatives , Animals , Cell Line , Endotoxemia/chemically induced , Endotoxemia/drug therapy , Endotoxemia/immunology , Heart/drug effects , Heart Injuries/immunology , Heart Injuries/pathology , Lipopolysaccharides , Male , Myocardium/immunology , Myocardium/pathology , Rats , Rats, Wistar , Signal Transduction/drug effects , Sphingosine/immunology
17.
Stem Cell Res ; 13(3 Pt B): 705-14, 2014 Nov.
Article En | MEDLINE | ID: mdl-25087895

Macrophages are an immune cell type found in every organ of the body. Classically, macrophages are recognised as housekeeping cells involved in the detection of foreign antigens and danger signatures, and the clearance of tissue debris. However, macrophages are increasingly recognised as a highly versatile cell type with a diverse range of functions that are important for tissue homeostasis and injury responses. Recent research findings suggest that macrophages contribute to tissue regeneration and may play a role in the activation and mobilisation of stem cells. This review describes recent advances in our understanding of the role played by macrophages in cardiac tissue maintenance and repair following injury. We examine the involvement of exogenous and resident tissue macrophages in cardiac inflammatory responses and their potential activity in regulating cardiac regeneration.


Heart Injuries/therapy , Hematopoietic Stem Cell Mobilization , Macrophages/immunology , Stem Cells/cytology , Animals , Heart Injuries/immunology , Heart Injuries/physiopathology , Humans , Regeneration
18.
Shock ; 39(5): 415-20, 2013 May.
Article En | MEDLINE | ID: mdl-23459112

INTRODUCTION: Clinical evidence supports the existence of a trauma-induced secondary cardiac injury. Experimental research suggests inflammation as a possible mechanism. The study aimed to determine if there was an early association between inflammation and secondary cardiac injury in trauma patients. METHODS: A cohort study of critically injured patients between January 2008 and January 2010 was undertaken. Levels of the cardiac biomarkers troponin I and heart-specific fatty acid-binding protein and the cytokines tumor necrosis factor α (TNF-α), interleukin (IL)-6, IL-1ß, and IL-8 were measured on admission to hospital, and again at 24 and 72 h. Participants were reviewed for adverse cardiac events (ACEs) and in-hospital mortality. RESULTS: Of 135 patients recruited, 18 (13%) had an ACE. Patients with ACEs had higher admission plasma levels of TNF-α (5.4 vs. 3.8 pg/mL; P = 0.03), IL-6 (140 vs. 58.9 pg/mL, P = 0.009), and IL-8 (19.3 vs. 9.1 pg/mL, P = 0.03) compared with those without events. Hour 24 cytokines were not associated with events, but IL-8 (14.5 vs. 5.8 pg/mL; P = 0.01) and IL-1ß (0.55 vs. 0.19 pg/mL; P = 0.04) were higher in patients with ACEs at 72 hours. Admission IL-6 was independently associated with heart-specific fatty acid-binding protein increase (P < 0.05). Patients who presented with an elevated troponin I combined with either an elevated TNF-α (relative risk [RR], 11.0; 95% confidence interval [CI], 1.8-66.9; P = 0.015), elevated IL-6 (RR, 17.3; 95% CI, 2.9-101.4; P = 0.001), or elevated IL-8 (RR, 15.0; 95% CI, 3.1-72.9; P = 0.008) were at the highest risk of in-hospital death when compared with individuals with normal biomarker and cytokine values. CONCLUSIONS: There is an association between hyperacute elevations in inflammatory cytokines with cardiac injury and ACEs in critically injured patients. Biomarker evidence of cardiac injury and inflammation on admission is associated with a higher risk of in-hospital death.


Biomarkers/blood , Cytokines/blood , Heart Injuries/etiology , Heart Injuries/immunology , Wounds and Injuries/complications , Wounds and Injuries/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Fatty Acid-Binding Proteins/blood , Female , Heart Injuries/blood , Humans , Interleukin-1/blood , Interleukin-1beta/blood , Interleukin-6/blood , Interleukin-8/blood , Male , Middle Aged , Troponin I/blood , Tumor Necrosis Factor-alpha/blood , Wounds and Injuries/blood , Young Adult
19.
PLoS One ; 7(4): e34177, 2012.
Article En | MEDLINE | ID: mdl-22509276

BACKGROUND: Experimental and clinical evidence has pinpointed a critical role for matrix metalloproteinase-2 (MMP-2) in ischemic ventricular remodeling and systolic heart failure. Prior studies have demonstrated that transgenic expression of the full-length, 68 kDa, secreted form of MMP-2 induces severe systolic failure. These mice also had unexpected and severe mitochondrial structural abnormalities and dysfunction. We hypothesized that an additional intracellular isoform of MMP-2, which affects mitochondrial function is induced under conditions of systolic failure-associated oxidative stress. METHODOLOGY AND PRINCIPAL FINDINGS: Western blots of cardiac mitochondria from the full length MMP-2 transgenics, ageing mice and a model of accelerated atherogenesis revealed a smaller 65 kDa MMP-2 isoform. Cultured cardiomyoblasts subjected to transient oxidative stress generated the 65 kDa MMP-2 isoform. The 65 kDa MMP-2 isoform was also induced by hypoxic culture of cardiomyoblasts. Genomic database analysis of the MMP-2 gene mapped transcriptional start sites and RNA transcripts induced by hypoxia or epigenetic modifiers within the first intron of the MMP-2 gene. Translation of these transcripts yields a 65 kDa N-terminal truncated isoform beginning at M(77), thereby deleting the signal sequence and inhibitory prodomain. Cellular trafficking studies demonstrated that the 65 kDa MMP-2 isoform is not secreted and is present in cytosolic and mitochondrial fractions, while the full length 68 kDa isoform was found only in the extracellular space. Expression of the 65 kDa MMP-2 isoform induced mitochondrial-nuclear stress signaling with activation of the pro-inflammatory NF-κB, NFAT and IRF transcriptional pathways. By microarray, the 65 kDa MMP-2 induces an innate immunity transcriptome, including viral stress response genes, innate immunity transcription factor IRF7, chemokines and pro-apoptosis genes. CONCLUSION: A novel N-terminal truncated intracellular isoform of MMP-2 is induced by oxidative stress. This isoform initiates a primary innate immune response that may contribute to progressive cardiac dysfunction in the setting of ischemia and systolic failure.


Immunity, Innate , Intracellular Space/enzymology , Matrix Metalloproteinase 2/metabolism , Oxidative Stress/immunology , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Animals , Cell Hypoxia/immunology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Enzyme Activation/drug effects , Enzyme Activation/immunology , Heart Injuries/enzymology , Heart Injuries/immunology , Heart Injuries/metabolism , Heart Injuries/pathology , Humans , I-kappa B Kinase/antagonists & inhibitors , Immunity, Innate/drug effects , Interferon Regulatory Factor-7/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/genetics , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/immunology , Mitochondria/pathology , Models, Molecular , Molecular Sequence Data , Molecular Weight , Oxidative Stress/drug effects , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Proteolysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Signal Transduction , Time Factors , Transcriptome/drug effects
20.
Bull Exp Biol Med ; 147(4): 517-20, 2009 Apr.
Article En, Ru | MEDLINE | ID: mdl-19704962

We studied the effects of bone marrow cell transplantation on myocardium of the prenecrotic zone in Wistar rats. Intramyocardial cell transplantation reduced the severity of hypertrophy of myocardium and the degree of its cicatricial degeneration on day 40 after cryodestruction. The morphology of the myocardium in the prenecrotic zone depended on the type of transplanted cells. The course of inflammation was swifter; vascularization of the myocardium was more intensive. The best effect, evaluated by the number of new vessels, was observed after MSC transplantation. Hence, the positive effect of bone marrow cell transplantation is realized at the expense of more rapid structural organization of the damaged site and stimulation of myocardial vascularization.


Bone Marrow Transplantation , Cold Temperature/adverse effects , Heart Injuries/pathology , Heart Injuries/surgery , Myocardium/pathology , Animals , Body Weight , Cardiac Surgical Procedures , Cardiomegaly/pathology , Cardiomegaly/surgery , Heart Injuries/immunology , Heart Ventricles/immunology , Heart Ventricles/pathology , Heart Ventricles/surgery , Inflammation/pathology , Inflammation/surgery , Male , Myocardium/immunology , Neovascularization, Physiologic , Organ Size , Rats , Rats, Wistar
...