Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters

Publication year range
1.
Cell ; 147(2): 293-305, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-22000010

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is one of the most common causes of death worldwide. We report in an emphysema model of mice chronically exposed to tobacco smoke that pulmonary vascular dysfunction, vascular remodeling, and pulmonary hypertension (PH) precede development of alveolar destruction. We provide evidence for a causative role of inducible nitric oxide synthase (iNOS) and peroxynitrite in this context. Mice lacking iNOS were protected against emphysema and PH. Treatment of wild-type mice with the iNOS inhibitor N(6)-(1-iminoethyl)-L-lysine (L-NIL) prevented structural and functional alterations of both the lung vasculature and alveoli and also reversed established disease. In chimeric mice lacking iNOS in bone marrow (BM)-derived cells, PH was dependent on iNOS from BM-derived cells, whereas emphysema development was dependent on iNOS from non-BM-derived cells. Similar regulatory and structural alterations as seen in mouse lungs were found in lung tissue from humans with end-stage COPD.


Subject(s)
Disease Models, Animal , Lung/pathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/pathology , Smoking/pathology , Animals , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Lung/physiopathology , Lysine/analogs & derivatives , Lysine/pharmacology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/pathology , Pulmonary Emphysema/physiopathology
2.
Mol Cell Biochem ; 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38381272

ABSTRACT

The recognition of microthrombi in the heart microcirculation has recently emerged from studies in COVID-19 decedents. The present study investigated the ultrastructure of coronary microthrombi in heart failure (HF) due to cardiomyopathies that are unrelated to COVID-19 infection. In addition, we have investigated the role of von Willebrand factor (VWF) and PECAM-1 in microthrombus formation. We used electron microscopy to investigate the occurrence of microthrombi in patients with HF due to dilated (DCM, n = 7), inflammatory (MYO, n = 6) and ischemic (ICM, n = 7) cardiomyopathy and 4 control patients. VWF and PECAM-1 was studied by quantitative immunohistochemistry and Western blot. In comparison to control, the number of microthrombi was increased 7-9 times in HF. This was associated with a 3.5-fold increase in the number of Weibel-Palade bodies (WPb) in DCM and MYO compared to control. A fivefold increase in WPb in ICM was significantly different from control, DCM and MYO. In Western blot, VWF was increased twofold in DCM and MYO, and more than threefold in ICM. The difference between ICM and DCM and MYO was statistically significant. These results were confirmed by quantitative immunohistochemistry. Compared to control, PECAM-1 was by approximatively threefold increased in all groups of patients. This is the first study to demonstrate the occurrence of microthrombi in the failing human heart. The occurrence of microthrombi is associated with increased expression of VWF and the number of WPb, being more pronounced in ICM. These changes are likely not compensated by increases in PECAM-1 expression.

3.
PLoS Genet ; 12(6): e1006099, 2016 06.
Article in English | MEDLINE | ID: mdl-27294373

ABSTRACT

Skeletal muscles provide metazoans with the ability to feed, reproduce and avoid predators. In humans, a heterogeneous group of genetic diseases, termed muscular dystrophies (MD), lead to skeletal muscle dysfunction. Mutations in the gene encoding Caveolin-3, a principal component of the membrane micro-domains known as caveolae, cause defects in muscle maintenance and function; however it remains unclear how caveolae dysfunction underlies MD pathology. The Cavin family of caveolar proteins can form membrane remodeling oligomers and thus may also impact skeletal muscle function. Changes in the distribution and function of Cavin4/Murc, which is predominantly expressed in striated muscles, have been reported to alter caveolae structure through interaction with Caveolin-3. Here, we report the generation and phenotypic analysis of murcb mutant zebrafish, which display impaired swimming capacity, skeletal muscle fibrosis and T-tubule abnormalities during development. To understand the mechanistic importance of Murc loss of function, we assessed Caveolin-1 and 3 localization and found it to be abnormal. We further identified an in vivo function for Murc in Erk signaling. These data link Murc with developmental defects in T-tubule formation and progressive muscle dysfunction, thereby providing a new candidate for the etiology of muscular dystrophy.


Subject(s)
Muscle Development/genetics , Muscle Proteins/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/pathology , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Animals, Genetically Modified , Caveolae/metabolism , Caveolin 1/metabolism , Caveolin 3/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Ion Channels/genetics , Muscle Proteins/metabolism , Muscular Dystrophies/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism
4.
Semin Cell Dev Biol ; 55: 22-30, 2016 07.
Article in English | MEDLINE | ID: mdl-26912117

ABSTRACT

Our previous studies suggested that an important variable of the progression of contractile dysfunction to terminal heart failure is the imbalance between myocyte cell death and myocyte renewal. For this reason, preventing myocyte cell death and an increasing generation of new myocytes may represent attractive targets in the treatment of human heart failure. Prospective clues to enhance myocardial regeneration are the newly discovered cells termed telocytes, formerly called interstitial Cajal-like cells, which are believed to nurse or guide the endogenous and exogenous stem cells for activation and commitment, but they also act as supporting cells for progenitor cells migration toward injured myocardium. We have recently found that telocytes are reduced in the diseased and failing myocardium. Importantly, the imbalance between telocyte proliferation and telocyte death is responsible for the telocytes depletion in cardiac diseases leading to heart failure. We have also demonstrated that telocytes are influenced by the extracellular matrix protein composition such that the telocytes are almost absent in areas of severe fibrosis. It is plausible that the reduction in telocytes in diseased human hearts could participate in the abnormal three-dimensional spatial organization and disturbed intercellular signalling of the myocardium. Decreased telocytes in diseased hearts would also be predicted to alter the property of telocytes to maintain cardiac stem cell niche by decreasing the pool of cardiac stem cells and thereby impairing cardiac regeneration.


Subject(s)
Heart Diseases/pathology , Myocardium/pathology , Telocytes/pathology , Animals , Humans , Models, Biological , Myocardium/ultrastructure , Regeneration , Stem Cell Niche
5.
Mol Cell Biochem ; 433(1-2): 27-40, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28337705

ABSTRACT

Cardiac surgery with extracorporeal circulation is characterized by different degrees of myocardial ischemia/reperfusion, which is often associated with postoperative atrial fibrillation (POAF). We have previously shown that a novel preventive therapy based on the reinforcement of the antioxidant system using omega-3 fatty acids plus antioxidant vitamin supplementation applied to patients undergoing cardiac surgery reduces POAF occurrence. We hypothesized that oxidative stress and nitrosative stress are involved in the development of an arrhythmogenic substrate by their effect on connexins (Cx40, Cx43 and Cx45) abundance and distribution pattern. Therefore, we have assessed the effect of redox status on atrial tissue in patients undergoing cardiac surgery. Placebo/POAF and supplemented/POAF patients showed 276 and 170% higher reactive oxygen species (ROS) levels and 223 and 96% higher nitrotyrosine residues levels, respectively, compared to sinus rhythm (SR). In POAF tissue, antioxidant supplementation prevented Cx40 and Cx43 lateralization on cardiomyocyte sarcolemma, keeping them at the intercalated disks. POAF samples showed Cx40 heterogeneous distribution pattern, presenting tissue areas lacking this protein (49 and 55% lower levels in placebo/POAF and supplemented/POAF groups, respectively, compared to SR). Of note, Cx45 overexpression occurred in POAF, being 211 and 167% higher in placebo/POAF and supplemented/POAF groups, respectively, compared to SR. It is concluded that treatment with omega-3 fatty acids and antioxidant vitamins reduces oxidative and nitrosative stress and prevents Cx40/Cx43 lateralization in atrial tissue likely contributing to POAF prevention. However, it failed to fully prevent POAF occurrence because these compounds have no effects on the normalization of Cx40 down-regulation and Cx45 up-regulation, which may promote POAF.


Subject(s)
Antioxidants/administration & dosage , Cardiac Surgical Procedures , Connexins/biosynthesis , Extracorporeal Circulation , Fatty Acids, Unsaturated/administration & dosage , Oxidative Stress/drug effects , Tyrosine/analogs & derivatives , Vitamins/administration & dosage , Female , Heart Atria/metabolism , Heart Atria/surgery , Humans , Male , Tyrosine/metabolism
6.
J Cell Mol Med ; 19(11): 2597-606, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311501

ABSTRACT

Telocytes (TCs) are a novel type of interstitial cells only recently described. This study aimed at characterizing and quantifying TCs and telopodes (Tps) in normal and diseased hearts. We have been suggested that TCs are influenced by the extracellular matrix (ECM) composition. We used transmission electron microscopy and c-kit immunolabelling to identify and quantify TCs in explanted human hearts with heart failure (HF) because of dilated, ischemic or inflammatory cardiomyopathy. LV myectomy samples from patients with aortic stenosis with preserved ejection fraction and samples from donor hearts which could not be used for transplantation served as controls. Quantitative immunoconfocal analysis revealed that 1 mm(2) of the normal myocardium contains 14.9 ± 3.4 TCs and 41.6 ± 5.9 Tps. As compared with the control group, the number of TCs and Tps in HF decreased more than twofold. There were no differences between HF and control in the number of Ki67-positive TCs. In contrast, terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling-positive TCs increased threefold in diseased hearts as compared to control. Significant inverse correlations were found between the amount of mature fibrillar collagen type I and the number of TCs (r = -0.84; P < 0.01) and Tps (r = -0.85; P < 0.01). The levels of degraded collagens showed a significant positive relationship with the TCs numbers. It is concluded that in HF the number of TCs are decreased because of higher rates of TCs apoptosis. Moreover, our results indicate that a close relationship exists between TCs and the ECM protein composition such that the number of TCs and Tps correlates negatively with the amount of mature fibrillar collagens and correlates positively with degraded collagens.


Subject(s)
Apoptosis , Fibrillar Collagens/analysis , Heart Failure/pathology , Telocytes/pathology , Cell Count , Extracellular Matrix Proteins/analysis , Heart Failure/metabolism , Humans , Immunologic Techniques , Microscopy, Electron, Transmission
7.
Circulation ; 129(5): 598-606, 2014 Feb 04.
Article in English | MEDLINE | ID: mdl-24201302

ABSTRACT

BACKGROUND: Atherosclerosis and vascular remodeling after injury are driven by inflammation and mononuclear cell infiltration. Extracellular RNA (eRNA) has recently been implicated to become enriched at sites of tissue damage and to act as a proinflammatory mediator. Here, we addressed the role of eRNA in high-fat diet-induced atherosclerosis and neointima formation after injury in atherosclerosis-prone mice. METHODS AND RESULTS: The presence of eRNA was revealed in atherosclerotic lesions from high-fat diet-fed low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice in a time-progressive fashion. RNase activity in plasma increased within the first 2 weeks (44±9 versus 70±7 mU/mg protein; P=0.0012), followed by a decrease to levels below baseline after 4 weeks of high-fat diet (44±9 versus 12±2 mU/mg protein; P<0.0001). Exposure of bone marrow-derived macrophages to eRNA resulted in a concentration-dependent upregulation of the proinflammatory mediators tumor necrosis factor-α, arginase-2, interleukin-1ß, interleukin-6, and interferon-γ. In a model of accelerated atherosclerosis after arterial injury in apolipoprotein E-deficient (ApoE(-/-)) mice, treatment with RNase1 diminished the increased plasma level of eRNA evidenced after injury. Likewise, RNase1 administration reduced neointima formation in comparison with vehicle-treated ApoE(-/-) controls (25.0±6.2 versus 46.9±6.9×10(3) µm(2), P=0.0339) and was associated with a significant decrease in plaque macrophage content. Functionally, RNase1 treatment impaired monocyte arrest on activated smooth muscle cells under flow conditions in vitro and inhibited leukocyte recruitment to injured carotid arteries in vivo. CONCLUSIONS: Because eRNA is associated with atherosclerotic lesions and contributes to inflammation-dependent plaque progression in atherosclerosis-prone mice, its targeting with RNase1 may serve as a new treatment option against atherosclerosis.


Subject(s)
Extracellular Fluid/physiology , Plaque, Atherosclerotic/blood , RNA/physiology , Ribonucleases/physiology , Animals , Atherosclerosis/blood , Atherosclerosis/chemically induced , Atherosclerosis/drug therapy , Diet, High-Fat/adverse effects , Extracellular Fluid/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/chemically induced , Plaque, Atherosclerotic/drug therapy , RNA/blood , Ribonucleases/therapeutic use
8.
Cell Tissue Res ; 361(3): 779-87, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25725788

ABSTRACT

Myostatin, a member of the TGF-ß superfamily of secreted growth factors, is a negative regulator of skeletal muscle growth. In the heart, it is expressed at lower levels compared to skeletal muscle but up-regulated under disease conditions. Cre recombinase-mediated inactivation of myostatin in adult cardiomyocytes leads to heart failure and increased mortality but cardiac function of surviving mice is restored after several weeks probably due to compensatory expression in non-cardiomyocytes. To study long-term effects of increased myostatin expression in the heart and to analyze the putative crosstalk between cardiomyocytes and fibroblasts, we overexpressed myostatin in cardiomyocytes. Increased expression of myostatin in heart muscle cells caused interstitial fibrosis via activation of the TAK-1-MKK3/6-p38 signaling pathway, compromising cardiac function in older mice. Our results uncover a novel role of myostatin in the heart and highlight the necessity for tight regulation of myostatin to maintain normal heart function.


Subject(s)
Cardiomyopathies/metabolism , Cardiomyopathies/pathology , MAP Kinase Kinase Kinases/metabolism , Myocytes, Cardiac/drug effects , Myostatin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cardiomyopathies/genetics , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression/genetics , Mice, Inbred C57BL , Muscle Development/genetics , Muscle, Skeletal/metabolism , Myostatin/genetics , Signal Transduction/genetics
9.
J Cell Mol Med ; 18(7): 1321-33, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24889158

ABSTRACT

Octamer binding trascription factor 4 (Oct4) is a transcription factor of POU family specifically expressed in embryonic stem cells (ESCs). A role for maintaining pluripotency and self-renewal of ESCs is assigned to Oct4 as a pluripotency marker. Oct4 can also be detected in adult stem cells such as bone marrow-derived mesenchymal stem cells. Several studies suggest a role for Oct4 in sustaining self-renewal capacity of adult stem cells. However, Oct4 gene ablation in adult stem cells revealed no abnormalities in tissue turnover or regenerative capacity. In the present study we have conspicuously found pulmonary Oct4-positive cells closely resembling the morphology of telocytes (TCs). These cells were found in the perivascular and peribronchial areas and their presence and location were confirmed by electron microscopy. Moreover, we have used Oct4-GFP transgenic mice which revealed a similar localization of the Oct4-GFP signal. We also found that Oct4 co-localized with several described TC markers such as vimentin, Sca-1, platelet-derived growth factor receptor-beta C-kit and VEGF. By flow cytometry analyses carried out with Oct4-GFP reporter mice, we described a population of EpCAM(neg) /CD45(neg) /Oct4-GFP(pos) that in culture displayed TC features. These results were supported by qRT-PCR with mRNA isolated from lungs by using laser capture microdissection. In addition, Oct4-positive cells were found to express Nanog and Klf4 mRNA. It is concluded for the first time that TCs in adult lung mouse tissue comprise Oct4-positive cells, which express pluripotency-related genes and represent therefore a population of adult stem cells which might contribute to lung regeneration.


Subject(s)
Green Fluorescent Proteins/metabolism , Lung/metabolism , Lung/ultrastructure , Octamer Transcription Factor-3/physiology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blotting, Western , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epithelial Cell Adhesion Molecule , Flow Cytometry , Fluorescent Antibody Technique , Green Fluorescent Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoenzyme Techniques , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Laser Capture Microdissection , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Nanog Homeobox Protein , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Basic Res Cardiol ; 109(1): 396, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24292852

ABSTRACT

Heart failure (HF) is a common and potentially deadly condition, which frequently develops as a consequence of various diseases of the heart. The incidence of heart failure continuously increases in aging societies illustrating the need for new therapeutic approaches. We recently discovered that continuous activation of oncostatin M (OSM), a cytokine of the interleukin-6 family that induces dedifferentiation of cardiomyocytes, promotes progression of heart failure in dilative cardiomyopathy. To evaluate whether inhibition of OSM signaling represents a meaningful therapeutic approach to prevent heart failure we attenuated OSM-receptor (Oß) signaling in a mouse model of inflammatory dilative cardiomyopathy. We found that administration of an antibody directed against the extracellular domain of Oß or genetic inactivation of a single allele of the Oß gene reduced cardiomyocyte remodeling and dedifferentiation resulting in improved cardiac performance and increased survival. We conclude that pharmacological attenuation of long-lasting Oß signaling is a promising strategy to treat different types and stages of HF that go along with infiltration by OSM-releasing inflammatory cells.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cardiomyopathy, Dilated/metabolism , Oncostatin M Receptor beta Subunit/antagonists & inhibitors , Signal Transduction/physiology , Animals , Blotting, Western , Cell Dedifferentiation , Disease Models, Animal , Heart Failure/metabolism , Humans , Inflammation/metabolism , Insulin-Like Growth Factor I , Magnetic Resonance Imaging , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects
11.
Int J Cardiol Heart Vasc ; 50: 101327, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38419608

ABSTRACT

Background and aim: Atrial fibrosis is an important factor in initiating and maintaining atrial fibrillation (AF). Collagen V belongs to fibrillar collagens. There are, however no data on collagen V in AF. The aim of this work was to study the quantity of collagen V and its relationship with the number of fibroblasts and TGF- b 1 expression in patients in sinus rhythm (SR) and in patients with atrial fibrillation (AF). Methods: We used quantitative immuhistochemistry to study collagen V in right and left atrial biopsies obtained from 35 patients in SR, 35 patients with paroxysmal AF (pAF) and 27 patients with chronic, long-standing persistent AF (cAF). In addition, we have quantified the number of vimentin-positive fibroblasts and expression levels of TGF-ß1. Results: Compared to patients in SR, collagen V was increased 1.8- and 3.1-fold in patients with pAF and cAF, respectively. In comparison with SR patients, the number of vimentin-positive cells increased significantly 1.46- and 1.8-fold in pAF and cAF patients, respectively.Compared to SR patients, expression levels of TGF-ß1, expressed as fluorescence units per tissue area, was significantly increased by 77 % and 300 % in patients with pAF and cAF, respectively. Similar to intensity measurements, the number of TGFß1-positive cells per 1 mm2 atrial tissue increased significantly from 35.5 ± 5.5 cells in SR patients to 61.9 ± 12.4 cells in pAF and 131.5 ± 23.5 cells in cAF. In both types of measurements, there was a statistically significant difference between pAF and cAF groups. Conclusions: This is the first study to show that AF is associated with increased expression levels of collagen V and TGF-ß1indicating its role in the pathogenesis of atrial fibrosis. In addition, increases in collagen V correlate with increased number of fibroblasts and TGF-ß1 and are more pronounced in cAF patients than those in pAF patients.

12.
Circ Res ; 109(7): 758-69, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21799151

ABSTRACT

RATIONALE: Telethonin (also known as titin-cap or t-cap) is a 19-kDa Z-disk protein with a unique ß-sheet structure, hypothesized to assemble in a palindromic way with the N-terminal portion of titin and to constitute a signalosome participating in the process of cardiomechanosensing. In addition, a variety of telethonin mutations are associated with the development of several different diseases; however, little is known about the underlying molecular mechanisms and telethonin's in vivo function. OBJECTIVE: Here we aim to investigate the role of telethonin in vivo and to identify molecular mechanisms underlying disease as a result of its mutation. METHODS AND RESULTS: By using a variety of different genetically altered animal models and biophysical experiments we show that contrary to previous views, telethonin is not an indispensable component of the titin-anchoring system, nor is deletion of the gene or cardiac specific overexpression associated with a spontaneous cardiac phenotype. Rather, additional titin-anchorage sites, such as actin-titin cross-links via α-actinin, are sufficient to maintain Z-disk stability despite the loss of telethonin. We demonstrate that a main novel function of telethonin is to modulate the turnover of the proapoptotic tumor suppressor p53 after biomechanical stress in the nuclear compartment, thus linking telethonin, a protein well known to be present at the Z-disk, directly to apoptosis ("mechanoptosis"). In addition, loss of telethonin mRNA and nuclear accumulation of this protein is associated with human heart failure, an effect that may contribute to enhanced rates of apoptosis found in these hearts. CONCLUSIONS: Telethonin knockout mice do not reveal defective heart development or heart function under basal conditions, but develop heart failure following biomechanical stress, owing at least in part to apoptosis of cardiomyocytes, an effect that may also play a role in human heart failure.


Subject(s)
Heart Failure/metabolism , Heart/physiopathology , Mechanotransduction, Cellular , Muscle Proteins/deficiency , Myocardium/metabolism , Adaptation, Physiological , Animals , Animals, Genetically Modified , Apoptosis , Biomechanical Phenomena , Cell Line, Tumor , Connectin , Disease Models, Animal , Echocardiography , Fibrosis , Genotype , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocardium/pathology , Phenotype , RNA Interference , Rats , Sarcomeres/metabolism , Stress, Mechanical , Transfection , Tumor Suppressor Protein p53/metabolism
13.
Eur Heart J ; 33(5): 595-605, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22106340

ABSTRACT

AIMS: Aortic stenosis causes cardiac hypertrophy and fibrosis, which often persists despite pressure unloading after aortic valve replacement. The persistence of myocardial fibrosis in particular leads to impaired cardiac function and increased mortality. We investigated whether granulocyte colony-stimulating factor (G-CSF) beneficially influences cardiac remodelling after pressure unloading. METHODS AND RESULTS: Left ventricular hypertrophy was induced by transverse aortic constriction in C57bl6 mice followed by debanding after 8 weeks. This model closely mimics aortic stenosis and subsequent aortic valve replacement. After debanding, mice were treated with either G-CSF or saline injection. Granulocyte colony-stimulating factor treatment significantly improved systolic (ejection fraction 70.48 ± 1.17 vs. 58.41 ± 1.56%, P < 0.001) and diastolic (E/E' 26.0 ± 1.0 vs. 32.6 ± 0.8, P < 0.05) function. Furthermore, cardiac fibrosis was significantly reduced in G-CSF-treated mice (collagen-I area fraction 7.96 ± 0.47 vs. 11.64 ± 1.22%, P < 0.05; collagen-III area fraction 10.73 ± 0.99 vs. 18.46 ± 0.71%, P < 0.001). Direct effects of G-CSF on cardiac fibroblasts or a relevant transdifferentiation of mobilized bone marrow cells could be excluded. However, a considerable infiltration of neutrophils was observed in G-CSF-treated mice. This sterile inflammation was accompanied by a selective release of interleukin-1 ß (IL-1ß) in the absence of other proinflammatory cytokines. In vitro experiments confirmed an increased expression of IL-1ß in neutrophils after G-CSF treatment. Interleukin-1ß directly induced the expression of the gelatinases matrix metalloproteinase-2 (MMP-2) and MMP-9 in cardiac fibroblasts thereby providing the regression of cardiac fibrosis. CONCLUSION: Granulocyte colony-stimulating factor treatment improves the cardiac function and leads to the regression of myocardial fibrosis after pressure unloading. These findings reveal a previously unknown mechanism of fibrosis regression. Granulocyte colony-stimulating factor might be a potential pharmacological treatment approach for patients suffering from congestive heart failure after aortic valve replacement, although further basic research and clinical trials are required in order to prove beneficial effects of G-CSF in the human organism.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hypertrophy, Left Ventricular/drug therapy , Interleukin-1beta/biosynthesis , Myocardium/pathology , Animals , Aorta , Aortic Valve Stenosis/complications , Aortic Valve Stenosis/physiopathology , Bone Marrow Cells/cytology , Cell Transdifferentiation , Constriction , Female , Fibrosis/drug therapy , Fibrosis/physiopathology , Hypertrophy, Left Ventricular/physiopathology , Mice , Mice, Inbred C57BL , Stroke Volume/physiology , Up-Regulation , Ventricular Remodeling/drug effects
14.
J Cell Mol Med ; 16(4): 701-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22188481

ABSTRACT

Telocytes (TCs) represent a new cell type recently described in mammalian skeletal muscle interstitium as well as in other organs. These have a specific morphology and phenotype, both in situ and in vitro. Telocytes are cells with long and slender cell prolongations, in contact with other interstitial cells, nerve fibres, blood capillaries and resident stem cells in niches. Our aim was to investigate the potential contribution of TCs to micro-vascular networks by immunofluorescent labelling of specific angiogenic growth factors and receptors. We found that in human skeletal muscle TCs were constantly located around intermediate and small blood vessels and endomysial capillaries. Epi-fluorescence and laser confocal microscopy showed that TCs express c-kit, platelet-derived growth factor receptor (PDGFR)-ß and VEGF, both in situ and in vitro. Telocytes were constantly located in the perivascular or pericapillary space, as confirmed by double staining of c-kit/CD31, PDGFR-ß/CD31 and PDGFR-ß/α-smooth muscle actin, respectively. Electron microscopy (EM) differentiated between pericytes and other cell types. Laminin labelling showed that TCs are not enclosed or surrounded by a basal lamina in contrast to mural cells. In conclusion, a) PDGFR-ß could be used as a marker for TCs and b) TCs are presumably a transitional population in the complex process of mural cell recruitment during angiogenesis and vascular remodelling.


Subject(s)
Muscle, Skeletal/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology
15.
Circ Res ; 106(4): 695-704, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20044516

ABSTRACT

RATIONALE: We previously discovered the human 10T-->C (Trp4Arg) missense mutation in exon 2 of the muscle LIM protein (MLP, CSRP3) gene. OBJECTIVE: We sought to study the effects of this single-nucleotide polymorphism in the in vivo situation. METHODS AND RESULTS: We now report the generation and detailed analysis of the corresponding Mlp(W4R/+) and Mlp(W4R/W4R) knock-in animals, which develop an age- and gene dosage-dependent hypertrophic cardiomyopathy and heart failure phenotype, characterized by almost complete loss of contractile reserve under catecholamine induced stress. In addition, evidence for skeletal muscle pathology, which might have implications for human mutation carriers, was observed. Importantly, we found significantly reduced MLP mRNA and MLP protein expression levels in hearts of heterozygous and homozygous W4R-MLP knock-in animals. We also detected a weaker in vitro interaction of telethonin with W4R-MLP than with wild-type MLP. These alterations may contribute to an increased nuclear localization of W4R-MLP, which was observed by immunohistochemistry. CONCLUSIONS: Given the well-known high frequency of this mutation in Caucasians of up to 1%, our data suggest that (W4R-MLP) might contribute significantly to human cardiovascular disease.


Subject(s)
Cardiomyopathy, Hypertrophic/metabolism , Heart Failure/metabolism , Muscle Proteins/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Ventricular Function, Left , Age Factors , Aging , Animals , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Cells, Cultured , Connectin , Disease Models, Animal , Fibrosis , Gene Knock-In Techniques , Genotype , Heart Failure/genetics , Heart Failure/physiopathology , Heterozygote , Homozygote , LIM Domain Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation, Missense , Myocytes, Cardiac/pathology , Phenotype , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Transfection
16.
J Biol Chem ; 285(19): 14467-74, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20185830

ABSTRACT

Maintenance of ion concentration gradients is essential for the function of many organs, including the kidney, the cornea, and the inner ear. Ion concentrations and fluid content in the cornea are regulated by endothelial cells that separate the collagenous avascular corneal stroma from the anterior eye chamber. Failure to maintain correct ion concentrations leads to swelling and destruction of the cornea. In the inner ear, the stria vascularis is responsible for generating proper ion concentrations in the endolymph, which is essential for hearing. Mutations of SLC4A11 in humans lead to syndromes associated with corneal dystrophy and perceptive deafness. The molecular mechanisms underlying these symptoms are poorly understood, impeding therapeutic interventions. The ion transporter SLC4A11 mediates sodium-dependent transport of borate as well as flux of sodium and hydroxyl ions in vitro. Here, we show that SLC4A11 is expressed in the endothelial cells of the cornea where it prevents severe morphological changes of the cornea caused by increased sodium chloride concentrations in the stroma. In the inner ear, SLC4A11 is located in fibrocytes underlying the stria vascularis. Loss of SLC4A11 leads to morphological changes in the fibrocytes and deafness. We demonstrate that SLC4A11 is essential for the generation of the endocochlear potential but not for regulation of potassium concentrations in the endolymph. In the kidney, SLC4A11 is expressed in the thin descending limb of Henle loop. SLC4A11 is essential for urinary concentration, suggesting that SLC4A11 participates in the countercurrent multiplication that concentrates urine in the kidney medulla.


Subject(s)
Anion Transport Proteins/physiology , Deafness/prevention & control , Fuchs' Endothelial Dystrophy/prevention & control , Osmosis , Polyuria/prevention & control , Sodium Chloride/metabolism , Symporters/physiology , Animals , Deafness/metabolism , Ear, Inner/metabolism , Ear, Inner/pathology , Endolymph/metabolism , Female , Fuchs' Endothelial Dystrophy/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyuria/metabolism , Potassium/metabolism , Subcellular Fractions
17.
J Cell Mol Med ; 14(7): 1917-21, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20604817

ABSTRACT

The existence of a new type of interstitial cells in the heart namely, interstitial Cajal-like cells (ICLC), has been described for the first time by Hinescu and Popescu in 2005. This study was then followed by an ascending trend of publications regarding the morphology, phenotype and distribution of myocardial ICLC in diverse species including human patients. Recently the new term 'telocytes' has been proposed for cells formerly known as ICLC, and the term 'telopodes' has been proposed for the prolongations of these cells. The identification of these cells is based on ultrastructural criteria. In addition, telocyters/telyopodes can be identified by several complementary approaches including methylene blue vital staining, silver impregnation and immunoreactivity against CD117/c-kit, vimentin, etc. This point of view presents critical data existing in literature, as well as own results, which unequivocally provide compelling evidence that telocytes are a new distinct cellular entity of myocardial interstitium. Several presumable functions of the myocardial telocytes are discussed: (i) intercellular signalling, (ii) cardiac repair/remodelling and (iii) stem cell nursing in cardiac renewal.


Subject(s)
Myocardium/pathology , Animals , Humans , Microscopy, Electron , Rats
19.
Circ Res ; 102(6): 703-10, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18239138

ABSTRACT

Sirt7 is a member of the mammalian sirtuin family consisting of 7 genes, Sirt1 to Sirt7, which all share a homology to the founding family member, the yeast Sir2 gene. Most sirtuins are supposed to act as histone/protein deacetylases, which use oxidized NAD in a sirtuin-specific, 2-step deacetylation reaction. To begin to decipher the biological role of Sirt7, we inactivated the Sirt7 gene in mice. Sirt7-deficient animals undergo a reduction in mean and maximum lifespans and develop heart hypertrophy and inflammatory cardiomyopathy. Sirt7 mutant hearts are also characterized by an extensive fibrosis, which leads to a 3-fold increase in collagen III accumulation. We found that Sirt7 interacts with p53 and efficiently deacetylates p53 in vitro, which corresponds to hyperacetylation of p53 in vivo and an increased rate of apoptosis in the myocardium of mutant mice. Sirt7-deficient primary cardiomyocytes show a approximately 200% increase in basal apoptosis and a significantly diminished resistance to oxidative and genotoxic stress suggesting a critical role of Sirt7 in the regulation of stress responses and cell death in the heart. We propose that enhanced activation of p53 by lack of Sirt7-mediated deacetylation contributes to the heart phenotype of Sirt7 mutant mice.


Subject(s)
Apoptosis , Cardiomegaly/enzymology , Cardiomyopathies/enzymology , DNA Damage , Myocarditis/enzymology , Myocytes, Cardiac/enzymology , Oxidative Stress , Sirtuins/metabolism , Acetylation , Aging/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/prevention & control , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/prevention & control , Cells, Cultured , Collagen Type III/metabolism , Fibrosis , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/genetics , Myocarditis/pathology , Myocarditis/prevention & control , Myocytes, Cardiac/pathology , Phenotype , Signal Transduction , Sirtuins/deficiency , Sirtuins/genetics , Tumor Suppressor Protein p53/metabolism
20.
Eur Heart J ; 30(17): 2155-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19525500

ABSTRACT

AIMS: The Tako-Tsubo cardiomyopathy (TTC) is characterized by a transient contractile dysfunction that has been assigned to excessive catecholamine levels after episodes of severe emotional or physical stress. Several studies have indicated that beta-adrenoceptor stimulation is associated with alteration in gene expression of Ca(2+)-regulatory proteins. Thus, the present study investigated the gene expression of crucial proteins [sarcoplasmic Ca(2+) ATPase (SERCA2a), sarcolipin (SLN), phospholamban (PLN), ryanodine receptor (RyR2), and sodium-calcium exchanger (NCX)] involved in the Ca(2+)-regulating system in TTC. METHODS AND RESULTS: In 10 consecutive patients, TTC was diagnosed by coronary angiography, ventriculography, and echocardiography. Endomyocardial biopsies were taken during the phase of severely impaired left ventricular (LV) function and after functional recovery. Non-diseased LV tissue from three donor hearts not used for transplantation served as healthy controls. Expression levels of Ca(2+)-regulatory proteins were analysed by means of real-time PCR, western blot, and immunohistochemistry. SLN, predominantly expressed in the atrial component, showed a remarkable ventricular expression in TTC patients. Gene expression of SERCA2a was significantly down-regulated. Conversely, PLN/SERCA2a ratio was increased. For PLN, dephosphorylation was documented using western blot and immunostaining of PLN-Ser(16) and PLN-Thr(17). No changes could be documented for NCX and RyR2. CONCLUSION: In TTC, ventricular expression of SLN and dephosphorylation of PLN potentially result in a reduced SERCA2a activity and its Ca(2+) affinity. Thus, the TTC is associated with specific alteration of Ca(2+)-handling proteins, which might be crucial for contractile dysfunction.


Subject(s)
Calcium-Binding Proteins/metabolism , Muscle Proteins/metabolism , Proteolipids/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism , Takotsubo Cardiomyopathy/etiology , Aged , Case-Control Studies , Female , Heart Ventricles/metabolism , Humans , Magnetic Resonance Angiography , Male , Middle Aged , Phosphorylation , Protein Phosphatase 1/metabolism , Takotsubo Cardiomyopathy/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL