Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Matrix Biol Plus ; 19-20: 100135, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38076279

ABSTRACT

Cardiac fibrosis is a central pathological feature in several cardiac diseases, but the underlying molecular players are insufficiently understood. The extracellular matrix proteoglycan versican is elevated in heart failure and suggested to be a target for treatment. However, the temporal expression and spatial distribution of versican and the versican cleavage fragment containing the neoepitope DPEAAE in cardiac fibrosis remains to be elucidated. In this study, we have examined versican during cardiac fibrosis development in a murine pressure overload model and in patients with cardiomyopathies. We found that versican, mainly the V1 isoform, was expressed immediately after induction of pressure overload, preceding collagen accumulation, and versican protein levels extended from the perivascular region into the cardiac interstitium. In addition, we found increased production of versican by collagen expressing fibroblasts, and that it was deposited extensively in the fibrotic extracellular matrix during pressure overload. In cardiac cell cultures, the expression of versican was induced by the pro-fibrotic transforming growth factor beta and mechanical stretch. Furthermore, we observed that the proteolytic cleavage of versican (DPEAAE fragment) increased in the late phase of fibrosis development during pressure overload. In patients with hypertrophic and dilated cardiomyopathies, we found elevated levels of versican and a positive correlation between versican and collagen mRNA in the heart, as well as increased cleavage of full-length protein. Taken together, the temporal expression profile and the spatial distribution of both the full-length versican and the DPEAAE fragment observed in this study indicates a role for versican in development of cardiac fibrosis.

2.
Cardiovasc Res ; 119(10): 1915-1927, 2023 08 19.
Article in English | MEDLINE | ID: mdl-37216909

ABSTRACT

AIMS: Heart failure is a condition with high mortality rates, and there is a lack of therapies that directly target maladaptive changes in the extracellular matrix (ECM), such as fibrosis. We investigated whether the ECM enzyme known as A disintegrin and metalloprotease with thrombospondin motif (ADAMTS) 4 might serve as a therapeutic target in treatment of heart failure and cardiac fibrosis. METHODS AND RESULTS: The effects of pharmacological ADAMTS4 inhibition on cardiac function and fibrosis were examined in rats exposed to cardiac pressure overload. Disease mechanisms affected by the treatment were identified based on changes in the myocardial transcriptome. Following aortic banding, rats receiving an ADAMTS inhibitor, with high inhibitory capacity for ADAMTS4, showed substantially better cardiac function than vehicle-treated rats, including ∼30% reduction in E/e' and left atrial diameter, indicating an improvement in diastolic function. ADAMTS inhibition also resulted in a marked reduction in myocardial collagen content and a down-regulation of transforming growth factor (TGF)-ß target genes. The mechanism for the beneficial effects of ADAMTS inhibition was further studied in cultured human cardiac fibroblasts producing mature ECM. ADAMTS4 caused a 50% increase in the TGF-ß levels in the medium. Simultaneously, ADAMTS4 elicited a not previously known cleavage of TGF-ß-binding proteins, i.e. latent-binding protein of TGF-ß and extra domain A-fibronectin. These effects were abolished by the ADAMTS inhibitor. In failing human hearts, we observed a marked increase in ADAMTS4 expression and cleavage activity. CONCLUSION: Inhibition of ADAMTS4 improves cardiac function and reduces collagen accumulation in rats with cardiac pressure overload, possibly through a not previously known cleavage of molecules that control TGF-ß availability. Targeting ADAMTS4 may serve as a novel strategy in heart failure treatment, in particular, in heart failure with fibrosis and diastolic dysfunction.


Subject(s)
Cardiomyopathies , Heart Failure , Rats , Humans , Animals , Disintegrins/metabolism , Disintegrins/pharmacology , Myocardium/metabolism , Heart Failure/metabolism , Cardiomyopathies/metabolism , Collagen/metabolism , Fibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Thrombospondins/metabolism , Metalloproteases/metabolism , Metalloproteases/pharmacology , Fibrosis
3.
ESC Heart Fail ; 10(2): 858-871, 2023 04.
Article in English | MEDLINE | ID: mdl-36444917

ABSTRACT

AIMS: Familial hypertrophic cardiomyopathy (HCM) is the most common form of inherited cardiac disease. It is characterized by myocardial hypertrophy and diastolic dysfunction, and can lead to severe heart failure, arrhythmias, and sudden cardiac death. Cardiac fibrosis, defined by excessive accumulation of extracellular matrix (ECM) components, is central to the pathophysiology of HCM. The ECM proteoglycan lumican is increased during heart failure and cardiac fibrosis, including HCM, yet its role in HCM remains unknown. We provide an in-depth assessment of lumican in clinical and experimental HCM. METHODS: Left ventricular (LV) myectomy specimens were collected from patients with hypertrophic obstructive cardiomyopathy (n = 15), and controls from hearts deemed unsuitable for transplantation (n = 8). Hearts were harvested from a mouse model of HCM; Myh6 R403Q mice administered cyclosporine A and wild-type littermates (n = 8-10). LV tissues were analysed for mRNA and protein expression. Patient myectomy or mouse mid-ventricular sections were imaged using confocal microscopy, direct stochastic optical reconstruction microscopy (dSTORM), or electron microscopy. Human foetal cardiac fibroblasts (hfCFBs) were treated with recombinant human lumican (n = 3) and examined using confocal microscopy. RESULTS: Lumican mRNA was increased threefold in HCM patients (P < 0.05) and correlated strongly with expression of collagen I (R2  = 0.60, P < 0.01) and III (R2  = 0.58, P < 0.01). Lumican protein was increased by 40% in patients with HCM (P < 0.01) and correlated with total (R2  = 0.28, P = 0.05) and interstitial (R2  = 0.30, P < 0.05) fibrosis. In mice with HCM, lumican mRNA increased fourfold (P < 0.001), and lumican protein increased 20-fold (P < 0.001) in insoluble ECM lysates. Lumican and fibrillar collagen were located together throughout fibrotic areas in HCM patient tissue, with increased co-localization measured in patients and mice with HCM (patients: +19%, P < 0.01; mice: +13%, P < 0.01). dSTORM super-resolution microscopy was utilized to image interstitial ECM which had yet to undergo overt fibrotic remodelling. In these interstitial areas, collagen I deposits located closer to (-15 nm, P < 0.05), overlapped more frequently with (+7.3%, P < 0.05) and to a larger degree with (+5.6%, P < 0.05) lumican in HCM. Collagen fibrils in such deposits were visualized using electron microscopy. The effect of lumican on collagen fibre formation was demonstrated by adding lumican to hfCFB cultures, resulting in thicker (+53.8 nm, P < 0.001), longer (+345.9 nm, P < 0.001), and fewer (-8.9%, P < 0.001) collagen fibres. CONCLUSIONS: The ECM proteoglycan lumican is increased in HCM and co-localizes with fibrillar collagen throughout areas of fibrosis in HCM. Our data suggest that lumican may promote formation of thicker collagen fibres in HCM.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Hypertrophic , Heart Failure , Humans , Animals , Mice , Lumican/physiology , Cardiomyopathy, Hypertrophic/genetics , Heart Failure/metabolism , Collagen Type I , Fibrosis , RNA, Messenger
4.
JACC Basic Transl Sci ; 5(12): 1210-1224, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33426377

ABSTRACT

An inflammatory response is required for tissue healing after a myocardial infarction (MI), but the process must be balanced to prevent maladaptive remodeling. This study shows that improved survival and cardiac function following MI, in mice deficient for the NLRP3 inflammasome, can be recapitulated in wild-type mice receiving bone marrow from Nlrp3 -/- mice. This suggests that NLRP3 activation in hematopoietic cells infiltrating in the myocardium increases mortality and late ventricular remodeling. Our data should encourage performing clinical trials directly targeting NLRP3 inflammasome and their inflammatory cytokines (interleukin-1ß and -18) in MI patients.

5.
Scand J Clin Lab Invest ; 77(5): 321-331, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28460577

ABSTRACT

We aimed to study the cardiac expression of bone morphogenetic protein 2, its receptor 1 b, and connective tissue growth factor, factors implicated in cardiac embryogenesis, following ischemia/hypoxia, heart failure, and in remodeling hearts from humans and mice. Biopsies from the left ventricle of patients with end-stage heart failure due to dilated cardiomyopathy or coronary artery disease were compared with donor hearts and biopsies from patients with normal heart function undergoing coronary artery bypass grafting. Mouse model of post-infarction remodeling was made by permanent ligation of the left coronary artery. Hearts were analyzed by real-time polymerase chain reaction and Western blotting after 24 hours and after 2 and 4 weeks. Patients with dilated cardiomyopathy and mice post-infarction had increased cardiac expression of connective tissue growth factor. Bone morphogenetic protein 2 was increased in human hearts failing due to coronary artery disease and in mice post-infarction. Gene expression of bone morphogenetic protein receptor 1 beta was reduced in hearts of patients with failure, but increased two weeks following permanent ligation of the left coronary artery in mice. In conclusion, connective tissue growth factor is upregulated in hearts of humans with dilated cardiomyopathy, bone morphogenetic protein 2 is upregulated in remodeling due to myocardial infarction while its receptor 1 b in human failing hearts is downregulated. A potential explanation might be an attempt to engage regenerative processes, which should be addressed by further, mechanistic studies.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Cardiomyopathy, Dilated/genetics , Connective Tissue Growth Factor/genetics , Coronary Artery Disease/genetics , Heart Failure/genetics , Adult , Aged , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Connective Tissue Growth Factor/metabolism , Coronary Artery Bypass , Coronary Artery Disease/complications , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Heart Function Tests , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Signal Transduction
6.
J Card Fail ; 22(10): 823-8, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27349571

ABSTRACT

BACKGROUND: Mitochondrial DNA (mtDNA) and possibly nuclear DNA (nDNA) are released as danger-associated molecular patterns during cardiac stress, and may activate several innate immune receptors. The purpose of this study was to investigate the regulation of these danger-associated molecular patterns during human heart failure (HF). METHODS AND RESULTS: Plasma levels of mtDNA and nDNA from HF patients (n = 84) were analyzed by reverse transcriptase-polymerase chain reaction and compared with controls (n = 72). Increased levels of mtDNA were found in New York Heart Association (NYHA) I-II and NYHA III-IV. There was evidence of increased nDNA in NYHA III-IV compared with controls and NYHA I-II. Kaplan-Meier analysis revealed higher mortality in patients with high nDNA levels, whereas high levels of mtDNA were associated with survival. CONCLUSIONS: Plasma levels of mtDNA and nDNA are elevated in human HF associated with increased and decreased mortality, respectively. This study may suggest a rationale for exploring interventions within inflammatory signaling pathways activated by nucleic acids as novel targets in treatment of HF.


Subject(s)
Co-Repressor Proteins/metabolism , Heart Failure/blood , Heart Failure/mortality , Mitochondria/metabolism , Aged , Biomarkers/blood , Case-Control Studies , Female , Heart Failure/physiopathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Predictive Value of Tests , Prognosis , Reference Values , Severity of Illness Index , Statistics, Nonparametric , Survival Analysis
7.
Life Sci ; 97(2): 145-54, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24398041

ABSTRACT

AIMS: Heart failure is associated with activation of fetal gene programs. Bone morphogenetic proteins (BMPs) regulate embryonic development through interaction with BMP receptors (BMPRs) on the cell surface. We investigated if the expression of BMP4 and its receptors BMPR1a and BMPR2 were activated in post-infarction remodeling and heart failure. MAIN METHODS: Left ventricular biopsies were taken from explanted hearts of patients with end-stage heart failure due to dilated cardiomyopathy (CMP; n=15) or ischemic heart disease (CAD; n=9), and compared with homograft control preparations from organ donors deceased due to non-cardiac causes (n=7). Other samples were taken from patients undergoing coronary artery bypass grafting (CABG; n=11). Mice were subjected to induced infarction by permanent coronary artery ligation or sham operation, and hearts were sampled serially thereafter (n=7 at each time point). KEY FINDINGS: Human and mouse hearts expressed BMP4 and both receptor subtypes. CABG and CMP patients had increased expression of mRNA encoding for BMP4, but unchanged protein. Mouse hearts had increased BMP4 precursor protein 24h after infarction. BMPR1a protein decreased in CAD patients and initially in postinfarcted mouse hearts, but increased again in the latter after two weeks. Human recombinant BMP4 promoted survival after H2O2 injury in HL-1 cells, and also protected adult mouse cardiomyocytes against hypoxia-reoxygenation injury. SIGNIFICANCE: Adult hearts express BMP4, the mRNA increasingly so in patients with coronary artery disease with good cardiac function. BMPRs are downregulated in cardiac remodeling and failure. Recombinant BMP4 has protective effects on cultured cardiomyocytes.


Subject(s)
Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Heart Failure/physiopathology , Myocardial Infarction/physiopathology , Adult , Aged , Animals , Cardiomyopathy, Dilated/physiopathology , Coronary Artery Disease/physiopathology , Disease Models, Animal , Down-Regulation , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/pathology , RNA, Messenger/metabolism , Time Factors , Ventricular Remodeling , Young Adult
8.
Int J Mol Med ; 33(3): 677-86, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24424466

ABSTRACT

Coronary atherosclerosis can lead to myocardial infarction, and secondarily to post-infarct remodelling and heart failure. Retinoic acid (RA) influences cell proliferation. We hypothesized that RA could influence gene expression and proliferation of cardiovascular cells. Left ventricular biopsies from patients with end-stage heart failure due to coronary artery disease (CAD) or dilated cardiomyopathy were investigated for the content of RA metabolites using liquid chromatography mass spectrometry (LC-MS/MS), and compared with healthy donors. All-trans retinoic acid (ATRA) was increased in the hearts of CAD patients. Gene expression (quantitative PCR) of RA target genes was not influenced in failing hearts, but was increased in the hearts of patients with CAD undergoing open heart surgery. The expression of RA target genes was increased in atherosclerotic lesions from carotid arteries compared to healthy arteries. Stimulation of cardiomyocytes, cardiofibroblasts, smooth muscle cells and endothelial cells with ATRA increased the gene expression of the key enzymes. Cardiofibroblast and smooth muscle cell proliferation were reduced by ATRA, which increased endothelial cell proliferation. Coronary artery disease leads to increased expression of RA target genes. ATRA accumulated in the failing human heart. All investigated cell types present in the heart had induced expression of RA target genes when stimulated with ATRA, which also influenced cell proliferation.


Subject(s)
Coronary Artery Disease/genetics , Myocardial Infarction/genetics , Receptors, Retinoic Acid/biosynthesis , Tretinoin/administration & dosage , Biopsy , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Heart Ventricles/metabolism , Humans , Myocardial Infarction/pathology , Myocardium/metabolism , Myocytes, Smooth Muscle/drug effects , Receptors, Retinoic Acid/genetics , Tandem Mass Spectrometry
10.
BMC Infect Dis ; 12: 144, 2012 Jun 25.
Article in English | MEDLINE | ID: mdl-22731696

ABSTRACT

BACKGROUND: The purpose of this study was to investigate mediators of inflammation and haemostasis in patients with chronic necrotizing pulmonary aspergillosis (CNPA), a locally, destructive process of the lung due to invasion by Aspergillus species. METHODS: Measurements of selected biomarkers in 10 patients with CNPA and 19 healthy, matched controls were performed with enzyme-linked immunosorbent assay (ELISA) and multiplex methodology. The gene expressions of relevant biomarkers were analyzed with real-time quantitative RT-PCR. RESULTS: Increased concentrations of circulating mediators of inflammation interleukin (IL)-6, IL-8, RANTES, TNF-α, ICAM-1 and mediators involved in endothelial activation and thrombosis (vWF, TF and PAI-1) were observed in patients with CNPA. The concentration of the anti-inflammatory cytokine IL-10 was increased both in plasma and in PBMC in the patient population. The gene expression of CD40L was decreased in PBMC from the patient group, accompanied by decreased concentrations of soluble (s) CD40L in the circulation. CONCLUSIONS: The proinflammatory response against Aspergillus may be counteracted by reduced CD40L and sCD40L, as well as increased IL-10, which may compromise the immune response against Aspergillus in patients with CNPA.


Subject(s)
Biomarkers/blood , Blood Coagulation Factors/analysis , Cytokines/blood , Invasive Pulmonary Aspergillosis/pathology , Aged , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Real-Time Polymerase Chain Reaction
11.
Circ Heart Fail ; 2(6): 624-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19919988

ABSTRACT

BACKGROUND: Inflammation has been implicated in the pathogenesis of heart failure (HF), but knowledge about the production and role of inflammatory actors remains incomplete. On the basis of its role in vascular inflammation, vascular proliferation, and matrix degradation, we hypothesized a role for the chemokine CXCL16 in the pathogenesis of myocardial remodeling and development of HF. METHODS AND RESULTS: Our main findings were (1) patients with chronic HF (n=188) had increased plasma levels of CXCL16, which correlated with disease severity. (2) Left ventricular tissue from patients with end-stage HF (n=8) showed enhanced CXCL16 levels compared with nonfailing left ventricular (n=6) as assessed by Western blotting. (3) In mice with postmyocardial infarction HF, expression of CXCL16, as assessed by real-time RT-PCR, was increased in the infarcted and the noninfarcted areas of left ventricular 3 and 7 days after coronary ligation, indicating early onset of CXCL16 production. Furthermore, mice exposed to aortic banding had enhanced CXCL16 expression in left ventricular, indicating that CXCL16 expression is not related to ischemia alone. (4) In vitro, CXCL16 promoted proliferation and impaired collagen synthesis in myocardial fibroblasts, and in cardiomyocytes and myocardial fibroblasts, CXCL16 increased matrix metalloproteinase activity, primarily reflecting increased matrix metalloproteinase-2 levels. (5) By using specific inhibitors, we showed that the effect of CXCL16 on fibroblasts involved activation of Jun N-terminal kinase. CONCLUSIONS: We show enhanced myocardial CXCL16 expression in experimental and clinical HF. The effect of CXCL16 on cardiomyocytes and fibroblasts suggests a role for CXCL16 in matrix remodeling and ultimately in the development of HF.


Subject(s)
Chemokine CXCL6/metabolism , Chemokines, CXC/metabolism , Extracellular Matrix/metabolism , Heart Failure/immunology , Heart Ventricles/immunology , Inflammation Mediators/metabolism , Myocytes, Cardiac/immunology , Receptors, Scavenger/metabolism , Ventricular Remodeling , Adult , Aged , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Chemokine CXCL16 , Chemokine CXCL6/genetics , Chemokines, CXC/blood , Chronic Disease , Collagen/biosynthesis , Disease Models, Animal , Enzyme Activation , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Heart Failure/metabolism , Heart Failure/pathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proline/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Scavenger/blood , Up-Regulation
12.
Eur J Heart Fail ; 10(4): 352-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18353719

ABSTRACT

BACKGROUND: Clinical and experimental studies suggest a pathogenic role for inflammation in chronic heart failure (HF). LIGHT is a member of the tumour necrosis factor superfamily involved in innate and adaptive immune responses. AIMS: We sought to investigate a potential pathogenic role of LIGHT in chronic HF. METHODS: We used various clinical and experimental approaches including studies in post-infarction HF rats and in vitro studies of endothelial cells and peripheral blood mononuclear cells (PBMC). RESULTS: Our main findings were: (i) LIGHT and its receptors (i.e., HVEM and lymphotoxin-beta receptor) were regulated during experimental HF, with strong expression in the infarcted area accompanied by up-regulation of HVEM in cardiomyocytes and endothelial cells also in the non-ischaemic part of the left ventricle. (ii) Patients with chronic HF had significantly increased expression of LIGHT on CD3(+) T-cells accompanied by increased expression of HVEM on monocytes and within the failing myocardium. (iii) LIGHT induced interleukin (IL)-6 expression in endothelial cells. In HF patients, but not in healthy controls, such an IL-6-inducing effect was also seen in LIGHT activated PBMC. CONCLUSION: Our findings in both clinical and experimental HF may suggest a role for LIGHT signalling pathways in the progression of chronic HF involving IL-6-related mechanisms.


Subject(s)
Disease Models, Animal , Heart Failure/pathology , Lymphotoxin beta Receptor/metabolism , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Adult , Aged , Animals , CD3 Complex/metabolism , Endothelium, Vascular/pathology , Female , Heart Ventricles/pathology , Humans , In Vitro Techniques , Interleukin-6/metabolism , Male , Middle Aged , Monocytes/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , T-Lymphocytes/pathology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...