Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Mol Med (Berl) ; 102(5): 667-677, 2024 05.
Article in English | MEDLINE | ID: mdl-38436713

ABSTRACT

C1q/TNF-related protein 3 (CTRP3) represents an adipokine with various metabolic and immune-regulatory functions. While circulating CTRP3 has been proposed as a potential biomarker for cardiovascular disease (CVD), current data on CTRP3 regarding coronary artery disease (CAD) remains partially contradictory. This study aimed to investigate CTRP3 levels in chronic and acute settings such as chronic coronary syndrome (CCS) and acute coronary syndrome (ACS). A total of 206 patients were classified into three groups: CCS (n = 64), ACS having a first acute event (ACS-1, n = 75), and ACS having a recurrent acute event (ACS-2, n = 67). The control group consisted of 49 healthy individuals. ELISA measurement in peripheral blood revealed decreased CTRP3 levels in all patient groups (p < 0.001) without significant differences between the groups. This effect was exclusively observed in male patients. Females generally exhibited significantly higher CTRP3 plasma levels than males. ROC curve analysis in male patients revealed a valuable predictive potency of plasma CTRP3 in order to identify CAD patients, with a proposed cut-off value of 51.25 ng/mL. The sensitivity and specificity of prediction by CTRP3 were congruent for the subgroups of CCS, ACS-1, and ACS-2 patients. Regulation of circulating CTRP3 levels in murine models of cardiovascular pathophysiology was found to be partly opposite to the clinical findings, with male mice exhibiting higher circulating CTRP3 levels than females. We conclude that circulating CTRP3 levels are decreased in both male CCS and ACS patients. Therefore, CTRP3 might be useful as a biomarker for CAD but not for distinguishing an acute from a chronic setting. KEY MESSAGES: CTRP3 levels were found to be decreased in both male CCS and ACS patients compared to healthy controls. Plasma CTRP3 has a valuable predictive potency in order to identify CAD patients among men and is therefore proposed as a biomarker for CAD but not for distinguishing between acute and chronic settings. Regulation of circulating CTRP3 levels in murine models of cardiovascular pathophysiology was found to be partly opposite to the clinical findings in men.


Subject(s)
Biomarkers , Humans , Male , Female , Middle Aged , Aged , Biomarkers/blood , Animals , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Mice , Adipokines/blood , Chronic Disease , ROC Curve , Tumor Necrosis Factors/blood , Case-Control Studies
2.
Circ Res ; 133(7): 592-610, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37641931

ABSTRACT

BACKGROUND: Activation of immune-inflammatory pathways involving TNFα (tumor necrosis factor alpha) signaling is critical for revascularization and peripheral muscle tissue repair after ischemic injury. However, mechanisms of TNFα-driven inflammatory cascades directing recruitment of proangiogenic immune cells to sites of ischemia are unknown. METHODS: Muscle tissue revascularization after permanent femoral artery ligation was monitored in mutant mice by laser Doppler imaging and light sheet fluorescence microscopy. TNFα-mediated signaling and the role of the CCL20 (C-C motif chemokine ligand 20)-CCR6 (C-C chemokine receptor 6) axis for formation of new vessels was studied in vitro and in vivo using bone marrow transplantation, flow cytometry, as well as biochemical and molecular biological techniques. RESULTS: TNFα-mediated activation of TNFR (tumor necrosis factor receptor) 1 but not TNFR2 was found to be required for postischemic muscle tissue revascularization. Bone marrow-derived CCR6+ neutrophil granulocytes were identified as a previously undescribed TNFα-induced population of proangiogenic neutrophils, characterized by increased expression of VEGFA (vascular endothelial growth factor A). Mechanistically, postischemic activation of TNFR1 induced expression of the CCL20 in vascular cells and promoted translocation of the CCL20 receptor CCR6 to the cell surface of neutrophils, essentially conditioning VEGFA-expressing proangiogenic neutrophils for CCL20-dependent recruitment to sites of ischemia. Moreover, impaired revascularization of ischemic peripheral muscle tissue in diabetic mice was associated with reduced numbers of proangiogenic neutrophils and diminished CCL20 expression. Administration of recombinant CCL20 enhanced recruitment of proangiogenic neutrophils and improved revascularization of diabetic ischemic skeletal muscles, which was sustained by sequential treatment with fluvastatin. CONCLUSIONS: We demonstrate that site-specific activation of the CCL20-CCR6 axis via TNFα recruits proangiogenic VEGFA-expressing neutrophils to sites of ischemic injury for initiation of muscle tissue revascularization. The findings provide an attractive option for tissue revascularization, particularly under diabetic conditions.


Subject(s)
Diabetes Mellitus, Experimental , Neutrophils , Animals , Mice , Receptors, CCR6/genetics , Tumor Necrosis Factor-alpha , Vascular Endothelial Growth Factor A , Vascular Surgical Procedures
3.
Int J Mol Sci ; 23(3)2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35163735

ABSTRACT

Oncostatin M (OSM), a member of the interleukin-6 family, functions as a major mediator of cardiomyocyte remodeling under pathological conditions. Its involvement in a variety of human cardiac diseases such as aortic stenosis, myocardial infarction, myocarditis, cardiac sarcoidosis, and various cardiomyopathies make the OSM receptor (OSMR) signaling cascades a promising therapeutic target. However, the development of pharmacological treatment strategies is highly challenging for many reasons. In mouse models of heart disease, OSM elicits opposing effects via activation of the type II receptor complex (OSMR/gp130). Short-term activation of OSMR/gp130 protects the heart after acute injury, whereas chronic activation promotes the development of heart failure. Furthermore, OSM has the ability to integrate signals from unrelated receptors that enhance fetal remodeling (dedifferentiation) of adult cardiomyocytes. Because OSM strongly stimulates the production and secretion of extracellular proteins, it is likely to exert systemic effects, which in turn, could influence cardiac remodeling. Compared with the mouse, the complexity of OSM signaling is even greater in humans because this cytokine also activates the type I leukemia inhibitory factor receptor complex (LIFR/gp130). In this article, we provide an overview of OSM-induced cardiomyocyte remodeling and discuss the consequences of OSMR/gp130 and LIFR/gp130 activation under acute and chronic conditions.


Subject(s)
Heart Failure , Interleukin-6 , Myocytes, Cardiac , Oncostatin M , Receptors, Oncostatin M , Animals , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/metabolism , Mice , Myocytes, Cardiac/metabolism , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
4.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923774

ABSTRACT

Cardiac sarcoidosis (CS) is a poorly understood disease and is characterized by the focal accumulation of immune cells, thus leading to the formation of granulomata (GL). To identify the developmental principles of fatal GL, fluorescence microscopy and Western blot analysis of CS and control patients is presented here. CS is visualized macroscopically by positron emission tomography (PET)/ computed tomography (CT). A battery of antibodies is used to determine structural, cell cycle and inflammatory markers. GL consist of CD68+, CD163+ and CD206+ macrophages surrounded by T-cells within fibrotic areas. Cell cycle markers such as phospho-histone H3, phospho-Aurora and Ki67 were moderately present; however, the phosphorylated ERM (ezrin, radixin and moesin) and Erk1/2 proteins, strong expression of the myosin motor protein and the macrophage transcription factor PU.1 indicate highly active GL. Mild apoptosis is consistent with PI3 kinase and Akt activation. Massive amounts of the IL-1R antagonist reflect a mild activation of stress and inflammatory pathways in GL. High levels of oncostatin M and the Reg3A and Reg3γ chemokines are in accordance with macrophage accumulation in areas of remodeling cardiomyocytes. We conclude that the formation of GL occurs mainly through chemoattraction and less by proliferation of macrophages. Furthermore, activation of the oncostatin/Reg3 axis might help at first to wall-off substances but might initiate the chronic development of heart failure.


Subject(s)
Cardiomyopathies/metabolism , Granuloma/metabolism , Myocardium/metabolism , Oncostatin M/metabolism , Pancreatitis-Associated Proteins/metabolism , Sarcoidosis/metabolism , Adult , Apoptosis , Aurora Kinases/metabolism , Cardiomyopathies/pathology , Cell Proliferation , Cytoskeletal Proteins/metabolism , Female , Granuloma/pathology , Histones/metabolism , Humans , Ki-67 Antigen/metabolism , Macrophages/metabolism , Macrophages/physiology , Male , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Middle Aged , Sarcoidosis/pathology
5.
Int J Mol Sci ; 21(17)2020 09 01.
Article in English | MEDLINE | ID: mdl-32882982

ABSTRACT

Fetal and hypertrophic remodeling are hallmarks of cardiac restructuring leading chronically to heart failure. Since the Ras/Raf/MEK/ERK cascade (MAPK) is involved in the development of heart failure, we hypothesized, first, that fetal remodeling is different from hypertrophy and, second, that remodeling of the MAPK occurs. To test our hypothesis, we analyzed models of cultured adult rat cardiomyocytes as well as investigated myocytes in the failing human myocardium by western blot and confocal microscopy. Fetal remodeling was induced through endothelial morphogens and monitored by the reexpression of Acta2, Actn1, and Actb. Serum-induced hypertrophy was determined by increased surface size and protein content of cardiomyocytes. Serum and morphogens caused reprogramming of Ras/Raf/MEK/ERK. In both models H-Ras, N-Ras, Rap2, B- and C-Raf, MEK1/2 as well as ERK1/2 increased while K-Ras was downregulated. Atrophy, MAPK-dependent ischemic resistance, loss of A-Raf, and reexpression of Rap1 and Erk3 highlighted fetal remodeling, while A-Raf accumulation marked hypertrophy. The knock-down of B-Raf by siRNA reduced MAPK activation and fetal reprogramming. In conclusion, we demonstrate that fetal and hypertrophic remodeling are independent processes and involve reprogramming of the MAPK.


Subject(s)
Cellular Reprogramming , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Cardiac/cytology , Vascular Remodeling , Animals , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/genetics , Male , Mitogen-Activated Protein Kinases/genetics , Myocytes, Cardiac/metabolism , Rats , Rats, Wistar , Signal Transduction
6.
Dis Markers ; 2020: 9356738, 2020.
Article in English | MEDLINE | ID: mdl-32774516

ABSTRACT

BACKGROUND: Pediatric patients show an impressive capacity of cardiac regeneration. In contrast, severely deteriorated adult hearts do usually not recover. Since cardiac remodeling-involving the expression of fetal genes-is regarded as an adaptation to stress, we compared hearts of adult patients suffering from dilated cardiomyopathy (DCM) with remodeling of cultured neonatal (NRC) as well as adult (ARC) rat cardiomyocytes and the developing postnatal myocardium. METHODS: NRC and ARC were stimulated with serum and cardiac morphogens derived from DCM hearts. Protein synthesis (PS) as well as protein accumulation (PA) was measured, and cell survival was determined under ischemic conditions. Fetal markers were investigated by Western blot. Biomarkers of remodeling were analyzed in controls, DCM, and 2- to 6-month-old children with tetralogy of Fallot as well as in neonatal and adult rats by immunofluorescence. RESULTS: In NRC, serum and morphogens strongly stimulated PS and PA and the reestablishment of cell-cell contacts (CCC). In ARC, both stimulants increased PS and CCC, but PA was only elevated after serum stimulation. In contrast to serum, morphogen treatment resulted in the expression of fetal genes in ARC as determined by nonmuscle α-actinin-1 and α-actinin-4 expression (NM-actinins) and was associated with increased survival under ischemia. NM-actinins were present in cardiomyocytes of DCM in a cross-striated pattern reminiscent of sarcomeres as well as in extensions of the area of the intercalated disc (ID). NM-actinins are expressed in NRC and in the developing heart. Radixin staining revealed remodeling of the area of the ID in DCM almost identical to stimulated cultured ARC. CONCLUSIONS: Remodeling was similar in ARC and in cardiomyocytes of DCM suggesting evolutionary conserved mechanisms of regeneration. Despite activation of fetal genes, the atrophy of ARC indicates differences in their regenerative capacity from NRC. Cardiac-derived factors induced NM-actinin expression and increased survival of ischemic ARC while circulating molecules were less effective. Identification of these cardiac-derived factors and determination of their individual capacity to heal or damage are of particular importance for a biomarker-guided therapy in adult patients.


Subject(s)
Actinin/metabolism , Cardiomyopathy, Dilated/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Myocytes, Cardiac/cytology , Tetralogy of Fallot/metabolism , Aged , Animals , Animals, Newborn , Cardiomyopathy, Dilated/blood , Cell Survival , Cells, Cultured , Female , Humans , Infant , Male , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Tetralogy of Fallot/blood
7.
J Biol Chem ; 293(52): 20181-20199, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30373773

ABSTRACT

The pleiotropic interleukin-6 (IL-6)-type cytokine oncostatin M (OSM) signals in multiple cell types, affecting processes such as cell differentiation, hematopoiesis, and inflammation. In humans, OSM exerts its effects through activation of either of two different heterodimeric receptor complexes, formed by glycoprotein 130 (gp130) and either OSM receptor (OSMR) or leukemia inhibitory factor receptor (LIFR). In contrast, the mouse OSM orthologue acts mainly through dimers containing OSMR and gp130 and shows limited activity through mouse LIFR. Despite their structural similarity, neither human nor mouse OSM signal through the other species' OSMR. The molecular basis for such species-specific signaling, however, remains poorly understood. To identify key molecular features of OSM that determine receptor activation in humans and mice, we generated chimeric mouse-human cytokines. Replacing regions within binding site III of murine OSM with the human equivalents showed that the cytokine's AB loop was critical for receptor selection. Substitutions of individual amino acids within this region demonstrated that residues Asn-37, Thr-40, and Asp-42 of the murine cytokine were responsible for limited LIFR activation and absence of human OSMR/LIFR signaling. In human OSM, Lys-44 appeared to be the main residue preventing mouse OSMR activation. Our data reveal that individual amino acids within the AB loop of OSM determine species-specific activities. These mutations might reflect a key step in the evolutionary process of this cytokine, in which receptor promiscuity gives way to ligand-receptor specialization.


Subject(s)
Oncostatin M/metabolism , Signal Transduction , Animals , Cell Line , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mice , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , Protein Multimerization/genetics , Protein Structure, Secondary , Species Specificity
8.
Cardiovasc Res ; 114(12): 1667-1679, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29850784

ABSTRACT

Aims: Myocardial infarction (MI) causes a massive increase of macrophages in the heart, which serve various non-redundant functions for cardiac repair. The identities of signals controlling recruitment of functionally distinct cardiac macrophages to sites of injury are only partially known. Previous work identified Regenerating islet-derived protein 3 beta (Reg3ß) as a novel factor directing macrophages to sites of myocardial injury. Herein, we aim to characterize functionally distinct macrophage subsets and understand the impact of different members of the Reg protein family including Reg3ß, Reg3γ, and Reg4 on their accumulation in the infarcted heart. Methods and results: We have determined dynamic changes of three phenotypically distinct tissue macrophage subpopulations in the mouse heart after MI by flow cytometry. RNA sequencing and bioinformatics analysis identified inflammatory gene expression patterns in MHC-IIhi/Ly6Clo and MHC-IIlo/Ly6Clo cardiac tissue macrophages while Ly6Chi cardiac tissue macrophages are characterized by gene activities associated with healing and revascularization of damaged tissue. Loss- and gain-of-function experiments revealed specific roles of Reg proteins for recruitment of cardiac tissue macrophage subpopulations to the site of myocardial injury. We found that expression of Reg3ß, Reg3γ, and Reg4 is strongly increased after MI in mouse and human hearts with Reg3ß providing the lead, followed by Reg3γ and Reg4. Inactivation of the Reg3ß gene prevented the increase of all types of cardiac tissue macrophages shortly after MI whereas local delivery of Reg3ß, Reg3γ, and Reg4 selectively stimulated recruitment of MHC-IIhi/Ly6Clo and MHC-IIlo/Ly6Clo but repressed accumulation of Ly6Chi cardiac tissue macrophages. Conclusion: We conclude that distinct cardiac macrophage subpopulations are characterized by substantially different gene expression patterns reflecting their pathophysiological role after MI. We argue that sequential, local production of Reg proteins orchestrates accumulation of macrophage subsets, which seem to act in a parallel or partially overlapping rather than in a successive manner.


Subject(s)
Chemotaxis , Macrophages/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Neoplasm Proteins/metabolism , Pancreatitis-Associated Proteins/metabolism , Animals , Antigens, Ly/metabolism , Cell Line , Disease Models, Animal , Gene Expression Regulation , Histocompatibility Antigens Class II/metabolism , Humans , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neoplasm Proteins/genetics , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , Pancreatitis-Associated Proteins/deficiency , Pancreatitis-Associated Proteins/genetics , Phenotype , Rats, Sprague-Dawley , Signal Transduction
9.
J Biol Chem ; 293(18): 7017-7029, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29511087

ABSTRACT

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are closely related members of the interleukin-6 (IL-6) cytokine family. Both cytokines share a common origin and structure, and both interact through a specific region, termed binding site III, to activate a dimeric receptor complex formed by glycoprotein 130 (gp130) and LIF receptor (LIFR) in humans. However, only OSM activates the OSM receptor (OSMR)-gp130 complex. The molecular features that enable OSM to specifically activate the OSMR are currently unknown. To define specific sequence motifs within OSM that are critical for initiating signaling via OSMR, here we generated chimeric OSM-LIF cytokines and performed alanine-scanning experiments. Replacement of the OSM AB loop within OSM's binding site III with that of LIF abrogated OSMR activation, measured as STAT3 phosphorylation at Tyr-705, but did not compromise LIFR activation. Correspondingly, substitution of the AB loop and D-helix in LIF with their OSM counterparts was sufficient for OSMR activation. The alanine-scanning experiments revealed that residues Tyr-34, Gln-38, Gly-39, and Leu-45 (in the AB loop) and Pro-153 (in the D-helix) had specific roles in activating OSMR but not LIFR signaling, whereas Leu-40 and Cys-49 (in the AB loop), and Phe-160 and Lys-163 (in the D-helix) were required for activation of both receptors. Because most of the key amino acid residues identified here are conserved between LIF and OSM, we concluded that comparatively minor differences in a few amino acid residues within binding site III account for the differential biological effects of OSM and LIF.


Subject(s)
Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Binding Sites , Cytokine Receptor gp130/metabolism , Cytokines/metabolism , Humans , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mutagenesis, Site-Directed , Oncostatin M/chemistry , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/chemistry , Oncostatin M Receptor beta Subunit/genetics , Phosphorylation , Protein Binding , Receptors, OSM-LIF/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
10.
Nat Med ; 21(4): 353-62, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751817

ABSTRACT

Cardiac healing after myocardial ischemia depends on the recruitment and local expansion of myeloid cells, particularly macrophages. Here we identify Reg3ß as an essential regulator of macrophage trafficking to the damaged heart. Using mass spectrometry-based secretome analysis, we found that dedifferentiating cardiomyocytes release Reg3ß in response to the cytokine OSM, which signals through Jak1 and Stat3. Loss of Reg3ß led to a large decrease in the number of macrophages in the ischemic heart, accompanied by increased ventricular dilatation and insufficient removal of neutrophils. This defect in neutrophil removal in turn caused enhanced matrix degradation, delayed collagen deposition and increased susceptibility to cardiac rupture. Our data indicate that OSM, acting through distinct intracellular pathways, regulates both cardiomyocyte dedifferentiation and cardiomyocyte-dependent regulation of macrophage trafficking. Release of OSM from infiltrating neutrophils and macrophages initiates a positive feedback loop in which OSM-induced production of Reg3ß in cardiomyocytes attracts additional OSM-secreting macrophages. The activity of the feedback loop controls the degree of macrophage accumulation in the heart, which is instrumental in myocardial healing.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Lectins, C-Type/metabolism , Macrophages/metabolism , Myocardial Ischemia/pathology , Myocardium/pathology , Oncostatin M/metabolism , Proteins/metabolism , Animals , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Collagen/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Heart/physiology , Heart Ventricles/metabolism , Inflammation , Interleukin-6/metabolism , Lectins, C-Type/genetics , Macrophages/cytology , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Pancreatitis-Associated Proteins , Proteins/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction
11.
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
12.
Cell Cycle ; 11(3): 439-45, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22262173

ABSTRACT

Dedifferentiation is a common phenomenon among plants but has only been found rarely in vertebrates where it is mostly associated with regenerative responses such as formation of blastemae in amphibians to initiate replacement of lost body parts. Relatively little attention has been paid to dedifferentiation processes in mammals although a decline of differentiated functions and acquisition of immature, "embryonic" properties is seen in various disease processes. Dedifferentiation of parenchymal cells in mammals might serve multiple purposes including (1) facilitation of tissue regeneration by generation of progenitor-like cells and (2) protection of cells from hypoxia by reduction of ATP consumption due to changes in energy metabolism and/or inactivation of energy-intensive "specialized" functions. We recently found that an inflammatory cytokine of the interleukin 6 family, oncostatin M (OSM), initiates dedifferentiation of cardiomyocytes both in vitro and in vivo. Interestingly, activation of the OSM signaling pathway protects the heart from acute myocardial ischemia but has a negative impact when continuously activated thereby promoting dilative cardiomyopathy. The strong presence of the OSM receptor on cardiomyocytes and the unique features of the OSM signaling circuit suggest a major role of OSM for cardiac protection and repair. We propose that continuous activation or malfunctions of the cellular dedifferentiation machinery might contribute to different disease conditions.


Subject(s)
Myocytes, Cardiac/cytology , Receptors, Oncostatin M/metabolism , Animals , Antineoplastic Agents/pharmacology , Cardiomyopathies/etiology , Cell Differentiation/drug effects , Humans , Myocardial Ischemia/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oncostatin M/pharmacology , Receptors, Oncostatin M/chemistry , Regeneration/physiology , Signal Transduction/drug effects
13.
Cell Stem Cell ; 9(5): 420-32, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22056139

ABSTRACT

Cardiomyocyte remodeling, which includes partial dedifferentiation of cardiomyocytes, is a process that occurs during both acute and chronic disease processes. Here, we demonstrate that oncostatin M (OSM) is a major mediator of cardiomyocyte dedifferentiation and remodeling during acute myocardial infarction (MI) and in chronic dilated cardiomyopathy (DCM). Patients suffering from DCM show a strong and lasting increase of OSM expression and signaling. OSM treatment induces dedifferentiation of cardiomyocytes and upregulation of stem cell markers and improves cardiac function after MI. Conversely, inhibition of OSM signaling suppresses cardiomyocyte remodeling after MI and in a mouse model of DCM, resulting in deterioration of heart function after MI but improvement of cardiac performance in DCM. We postulate that dedifferentiation of cardiomyocytes initially protects stressed hearts but fails to support cardiac structure and function upon continued activation. Manipulation of OSM signaling provides a means to control the differentiation state of cardiomyocytes and cellular plasticity.


Subject(s)
Cell Dedifferentiation , Myocytes, Cardiac/pathology , Oncostatin M/metabolism , Ventricular Remodeling/physiology , Animals , Biomarkers/metabolism , Blotting, Western , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/physiopathology , Cardiotonic Agents/metabolism , Cell Cycle/drug effects , Cell Dedifferentiation/drug effects , DNA/biosynthesis , Fluorescent Antibody Technique , Gene Deletion , Gene Expression Regulation/drug effects , Heart Function Tests/drug effects , Humans , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Ventricular Remodeling/drug effects
14.
Arterioscler Thromb Vasc Biol ; 31(10): 2297-305, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21737786

ABSTRACT

OBJECTIVE: Collateral artery growth or arteriogenesis is the primary means of the circulatory system to maintain blood flow in the face of major arterial occlusions. Arteriogenesis depends on activation of fibroblast growth factor (FGF) receptors, but relatively little is known about downstream mediators of FGF signaling. METHODS AND RESULTS: We screened for signaling components that are activated in response to administration of FGF-2 to cultured vascular smooth muscle cells (VSMCs) and detected a significant increase of Rap2 but not of other Ras family members, which corresponded to a strong upregulation of Rap2 and C-Raf in growing collaterals from rabbits with femoral artery occlusion. Small interfering RNAs directed against Rap2 did not affect FGF-2 induced proliferation of VSMC but strongly inhibited their migration. Inhibition of FGF receptor-1 (FGFR1) signaling by infusion of a sulfonic acid polymer or infection with a dominant-negative FGFR1 adenovirus inhibited Rap2 upregulation and collateral vessel growth. Similarly, expression of dominant-negative Rap2 blocked arteriogenesis, whereas constitutive active Rap2 enhanced collateral vessel growth. CONCLUSIONS: Rap2 is part of the arteriogenic program and acts downstream of the FGFR1 to stimulate VSMC migration. Specific modulation of Rap2 might be an attractive target to manipulate VSMC migration, which plays a role in numerous pathological processes.


Subject(s)
Arterial Occlusive Diseases/metabolism , Cell Movement , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , rap GTP-Binding Proteins/metabolism , Animals , Arterial Occlusive Diseases/genetics , Arterial Occlusive Diseases/pathology , Arterial Occlusive Diseases/physiopathology , Cell Proliferation , Collateral Circulation , Disease Models, Animal , Femoral Artery/metabolism , Femoral Artery/pathology , Femoral Artery/physiopathology , Femoral Artery/surgery , Fibroblast Growth Factor 2/metabolism , HEK293 Cells , Humans , Ligation , Mice , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , RNA Interference , Rabbits , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regional Blood Flow , Time Factors , Transfection , rap GTP-Binding Proteins/genetics
15.
J Mol Cell Cardiol ; 45(3): 411-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18634800

ABSTRACT

Previous studies have shown that p53 plays an important role in maintaining cell cycle arrest of cardiomyocytes, which might account for the inability of human hearts to regenerate adequately after injury. Therefore, inhibition of p53 represents an attractive strategy to restore cell cycle progression in cardiomyocytes although such an approach is hampered by the potential danger of concomitant tumor induction. During normal development, N-terminal truncated isoforms of the p53-related protein p73 are naturally occurring antagonists of p53 and p73, which are not related to tumor induction. We have generated recombinant adenoviruses encoding dominant-interfering p73 (Ad-p73DD) to inhibit p53/p73 in murine hearts at different developmental stages. We found that the expression of p73DD(wt) in newborn mice led to the increase of the relative heart weights after 14 days which is paralleled by a significant increase of proliferating cardiomyocytes as seen by ICC (BrdU-incorporation, phosphorylation of histone3, expression of AuroraB) without induction of apoptosis. Stimulation of cell cycle progression in cardiomyocytes went along with a significant down-regulation of the p53-dependent cdk-inhibitor p21WAF both on mRNA and protein level. Furthermore, mRNA levels and protein expression of D-type cyclins and cyclins A, B2, and E were selectively increased after expression of p73DD. We further show that the cell cycle entry of cardiomyocytes is not restricted to neonatal hearts but is also found in adult mouse hearts 5 days after intramyocardial injection of Ad-p73DD. Taken together we reason that directed expression of dominant-negative p73 might be utilized to stimulate proliferation of cardiomyocytes to improve cardiac regeneration.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Genes, Dominant/physiology , Myocytes, Cardiac/cytology , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Up-Regulation/physiology , Adenoviridae/genetics , Animals , Animals, Newborn , Cell Line , DNA-Binding Proteins/biosynthesis , Directed Molecular Evolution , Genetic Vectors , Humans , Mice , Mice, Transgenic , Myocytes, Cardiac/metabolism , Nuclear Proteins/biosynthesis , Point Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/biosynthesis
16.
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
17.
Circ Res ; 96(5): 509-17, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15718499

ABSTRACT

Proliferation of mammalian cardiomyocytes ceases around birth when a transition from hyperplastic to hypertrophic myocardial growth occurs. Previous studies demonstrated that directed expression of the transcription factor E2F1 induces S-phase entry in cardiomyocytes along with stimulation of programmed cell death. Here, we show that directed expression of E2F2 and E2F4 by adenovirus mediated gene transfer in neonatal cardiomyocytes induced S-phase entry but did not result in an onset of apoptosis whereas directed expression of E2F1 and E2F3 strongly evoked programmed cell death concomitant with cell cycle progression. Although both E2F2 and E2F4 induced S-phase entry only directed expression of E2F2 resulted in mitotic cell division of cardiomyocytes. Expression of E2F5 or a control LacZ-Adenovirus had no effects on cell cycle progression. Quantitative real time PCR revealed that E2F1, E2F2, E2F3, and E2F4 alleviate G0 arrest by induction of cyclinA and E cyclins. Furthermore, directed expression of E2F1, E2F3, and E2F5 led to a transcriptional activation of several proapoptotic genes, which were mitigated by E2F2 and E2F4. Our finding that expression of E2F2 induces cell division of cardiomyocytes along with a suppression of proapoptotic genes might open a new access to improve the regenerative capacity of cardiomyocytes.


Subject(s)
Apoptosis/physiology , DNA Replication/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Myocytes, Cardiac/metabolism , Transcription Factors/physiology , Adenoviridae/genetics , Animals , Apoptosis/genetics , Binding Sites , Carrier Proteins/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Cells, Cultured/cytology , Cells, Cultured/metabolism , Culture Media, Serum-Free , Cyclins/physiology , DNA Replication/genetics , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , E2F2 Transcription Factor , E2F3 Transcription Factor , E2F4 Transcription Factor , E2F5 Transcription Factor , Gene Expression Profiling , Gene Expression Regulation/genetics , Genetic Vectors/genetics , In Situ Nick-End Labeling , Mice , Myocytes, Cardiac/cytology , Nuclear Proteins/physiology , Polymerase Chain Reaction , Protein Binding , Protein Interaction Mapping , Proteins/physiology , Rats , Recombinant Fusion Proteins/physiology , Retinoblastoma Protein/physiology , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , S Phase/physiology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...