Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Immunity ; 56(5): 979-997.e11, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37100060

ABSTRACT

Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.


Subject(s)
Interleukin-6 , RNA , Endothelial Cells/metabolism , Cytokine Receptor gp130 , Endothelium/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism
2.
Nature ; 622(7983): 619-626, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37758950

ABSTRACT

Postnatal maturation of cardiomyocytes is characterized by a metabolic switch from glycolysis to fatty acid oxidation, chromatin reconfiguration and exit from the cell cycle, instating a barrier for adult heart regeneration1,2. Here, to explore whether metabolic reprogramming can overcome this barrier and enable heart regeneration, we abrogate fatty acid oxidation in cardiomyocytes by inactivation of Cpt1b. We find that disablement of fatty acid oxidation in cardiomyocytes improves resistance to hypoxia and stimulates cardiomyocyte proliferation, allowing heart regeneration after ischaemia-reperfusion injury. Metabolic studies reveal profound changes in energy metabolism and accumulation of α-ketoglutarate in Cpt1b-mutant cardiomyocytes, leading to activation of the α-ketoglutarate-dependent lysine demethylase KDM5 (ref. 3). Activated KDM5 demethylates broad H3K4me3 domains in genes that drive cardiomyocyte maturation, lowering their transcription levels and shifting cardiomyocytes into a less mature state, thereby promoting proliferation. We conclude that metabolic maturation shapes the epigenetic landscape of cardiomyocytes, creating a roadblock for further cell divisions. Reversal of this process allows repair of damaged hearts.


Subject(s)
Cellular Reprogramming , Fatty Acids , Heart , Regeneration , Animals , Mice , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Cell Hypoxia , Cell Proliferation , Energy Metabolism , Enzyme Activation , Epigenesis, Genetic , Fatty Acids/metabolism , Heart/physiology , Histone Demethylases/metabolism , Ketoglutaric Acids/metabolism , Mutation , Myocardium , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Regeneration/physiology , Reperfusion Injury , Transcription, Genetic
3.
Circ Res ; 132(11): 1468-1485, 2023 05 26.
Article in English | MEDLINE | ID: mdl-37042252

ABSTRACT

BACKGROUND: The ability of the right ventricle (RV) to adapt to an increased pressure afterload determines survival in patients with pulmonary arterial hypertension. At present, there are no specific treatments available to prevent RV failure, except for heart/lung transplantation. The wingless/int-1 (Wnt) signaling pathway plays an important role in the development of the RV and may also be implicated in adult cardiac remodeling. METHODS: Molecular, biochemical, and pharmacological approaches were used both in vitro and in vivo to investigate the role of Wnt signaling in RV remodeling. RESULTS: Wnt/ß-catenin signaling molecules are upregulated in RV of patients with pulmonary arterial hypertension and animal models of RV overload (pulmonary artery banding-induced and monocrotaline rat models). Activation of Wnt/ß-catenin signaling leads to RV remodeling via transcriptional activation of FOSL1 and FOSL2 (FOS proto-oncogene [FOS] like 1/2, AP-1 [activator protein 1] transcription factor subunit). Immunohistochemical analysis of pulmonary artery banding -exposed BAT-Gal (ß-catenin-activated transgene driving expression of nuclear ß-galactosidase) reporter mice RVs exhibited an increase in ß-catenin expression compared with their respective controls. Genetic inhibition of ß-catenin, FOSL1/2, or WNT3A stimulation of RV fibroblasts significantly reduced collagen synthesis and other remodeling genes. Importantly, pharmacological inhibition of Wnt signaling using inhibitor of PORCN (porcupine O-acyltransferase), LGKK-974 attenuated fibrosis and cardiac hypertrophy leading to improvement in RV function in both, pulmonary artery banding - and monocrotaline-induced RV overload. CONCLUSIONS: Wnt- ß-Catenin-FOSL signaling is centrally involved in the hypertrophic RV response to increased afterload, offering novel targets for therapeutic interference with RV failure in pulmonary hypertension.


Subject(s)
Heart Failure , Pulmonary Arterial Hypertension , Rats , Mice , Animals , Ventricular Remodeling , beta Catenin , Catenins , Monocrotaline/toxicity , Signal Transduction , Disease Models, Animal , Ventricular Function, Right
4.
J Mol Cell Cardiol ; 193: 53-66, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38838815

ABSTRACT

The HSP70 co-chaperone BAG3 targets unfolded proteins to degradation via chaperone assisted selective autophagy (CASA), thereby playing pivotal roles in the proteostasis of adult cardiomyocytes (CMs). However, the complex functions of BAG3 for regulating autophagy in cardiac disease are not completely understood. Here, we demonstrate that conditional inactivation of Bag3 in murine CMs leads to age-dependent dysregulation of autophagy, associated with progressive cardiomyopathy. Surprisingly, Bag3-deficient CMs show increased canonical and non-canonical autophagic flux in the juvenile period when first signs of cardiac dysfunction appear, but reduced autophagy during later stages of the disease. Juvenile Bag3-deficient CMs are characterized by decreased levels of soluble proteins involved in synchronous contraction of the heart, including the gap junction protein Connexin 43 (CX43). Reiterative administration of chloroquine (CQ), an inhibitor of canonical and non-canonical autophagy, but not inactivation of Atg5, restores normal concentrations of soluble cardiac proteins in juvenile Bag3-deficient CMs without an increase of detergent-insoluble proteins, leading to complete recovery of early-stage cardiac dysfunction in Bag3-deficient mice. We conclude that loss of Bag3 in CMs leads to age-dependent differences in autophagy and cardiac dysfunction. Increased non-canonical autophagic flux in the juvenile period removes soluble proteins involved in cardiac contraction, leading to early-stage cardiomyopathy, which is prevented by reiterative CQ treatment.

5.
Circ Res ; 131(7): 580-597, 2022 09 16.
Article in English | MEDLINE | ID: mdl-36000401

ABSTRACT

BACKGROUND: ADAR1 (adenosine deaminase acting on RNA-1)-mediated adenosine to inosine (A-to-I) RNA editing plays an essential role for distinguishing endogenous from exogenous RNAs, preventing autoinflammatory ADAR1 also regulates cellular processes by recoding specific mRNAs, thereby altering protein functions, but may also act in an editing-independent manner. The specific role of ADAR1 in cardiomyocytes and its mode of action in the heart is not fully understood. To determine the role of ADAR1 in the heart, we used different mutant mouse strains, which allows to distinguish immunogenic, editing-dependent, and editing-independent functions of ADAR1. METHODS: Different Adar1-mutant mouse strains were employed for gene deletion or specific inactivation of ADAR1 enzymatic activity in cardiomyocytes, either alone or in combination with Ifih1 (interferon induced with helicase C domain 1) or Irf7 (interferon regulatory factor 7) gene inactivation. Mutant mice were investigated by immunofluorescence, Western blot, RNAseq, proteomics, and functional MRI analysis. RESULTS: Inactivation of Adar1 in cardiomyocytes resulted in late-onset autoinflammatory myocarditis progressing into dilated cardiomyopathy and heart failure at 6 months of age. Adar1 depletion activated interferon signaling genes but not NFκB (nuclear factor kappa B) signaling or apoptosis and reduced cardiac hypertrophy during pressure overload via induction of Irf7. Additional inactivation of the cytosolic RNA sensor MDA5 (melanoma differentiation-associated gene 5; encoded by the Ifih1 gene) in Adar1 mutant mice prevented activation of interferon signaling gene and delayed heart failure but did not prevent lethality after 8.5 months. In contrast, compound mutants only expressing catalytically inactive ADAR1 in an Ifih1-mutant background were completely normal. Inactivation of Irf7 attenuated the phenotype of Adar1-deficient cardiomyocytes to a similar extent as Ifih1 depletion, identifying IRF7 as the main mediator of autoinflammatory responses caused by the absence of ADAR1 in cardiomyocytes. CONCLUSIONS: Enzymatically active ADAR1 prevents IRF7-mediated autoinflammatory reactions in the heart triggered by endogenous nonedited RNAs. In addition to RNA editing, ADAR1 also serves editing-independent roles in the heart required for long-term cardiac function and survival.


Subject(s)
Adenosine Deaminase , Heart Failure , Adenosine/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Inosine/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/metabolism , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , RNA
7.
EMBO J ; 36(9): 1199-1214, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28314781

ABSTRACT

Control of energy homeostasis and metabolism is achieved by integrating numerous pathways, and miRNAs are involved in this process by regulating expression of multiple target genes. However, relatively little is known about the posttranscriptional processing of miRNAs and a potential role for the precursors they derive from. Here, we demonstrate that mature miRNA-22 is more abundant in muscle from male mice relative to females and that this enables sex-specific regulation of muscular lipid metabolism and body weight by repressing estrogen receptor alpha (ERα) expression. We found that the ERα adjusts its own activity by preventing processing of miR-22 via direct binding to a conserved ERα-binding element within the primary miR-22 precursor. Mutation of the ERα binding site within the pri-miR-22 in vivo eliminates sex-specific differences in miR-22 expression. We reason that the resulting tissue selective negative feedback regulation is essential to establish sex-specific differences in muscle metabolism and body weight development.


Subject(s)
Estrogen Receptor alpha/biosynthesis , Gene Expression Regulation , Lipid Metabolism , MicroRNAs/metabolism , Muscles/metabolism , Animals , Male , Mice , Sex Factors
8.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008777

ABSTRACT

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) signaling protects the heart after myocardial infarction (MI). In mice, oncostatin M receptor (OSMR) and leukemia inhibitory factor receptor (LIFR) are selectively activated by the respective cognate ligands while OSM activates both the OSMR and LIFR in humans, which prevents efficient translation of mouse data into potential clinical applications. We used an engineered human-like OSM (hlOSM) protein, capable to signal via both OSMR and LIFR, to evaluate beneficial effects on cardiomyocytes and hearts after MI in comparison to selective stimulation of either LIFR or OSMR. Cell viability assays, transcriptome and immunoblot analysis revealed increased survival of hypoxic cardiomyocytes by mLIF, mOSM and hlOSM stimulation, associated with increased activation of STAT3. Kinetic expression profiling of infarcted hearts further specified a transient increase of OSM and LIF during the early inflammatory phase of cardiac remodeling. A post-infarction delivery of hlOSM but not mOSM or mLIF within this time period combined with cardiac magnetic resonance imaging-based strain analysis uncovered a global cardioprotective effect on infarcted hearts. Our data conclusively suggest that a simultaneous and rapid activation of OSMR and LIFR after MI offers a therapeutic opportunity to preserve functional and structural integrity of the infarcted heart.


Subject(s)
Cardiotonic Agents/metabolism , Myocardial Infarction/prevention & control , Oncostatin M/metabolism , Receptors, OSM-LIF/metabolism , Animals , Cell Hypoxia/genetics , Cell Survival , Cells, Cultured , Humans , Kinetics , Leukemia Inhibitory Factor/metabolism , Mice , Myocardial Contraction , Myocardial Infarction/genetics , Myocytes, Cardiac/metabolism , Protein Engineering , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Oncostatin M/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Species Specificity , Transcriptome/genetics
9.
Proc Natl Acad Sci U S A ; 114(40): E8352-E8361, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28923965

ABSTRACT

Sirtuins (Sirt1-Sirt7) are NAD+-dependent protein deacetylases/ADP ribosyltransferases, which play decisive roles in chromatin silencing, cell cycle regulation, cellular differentiation, and metabolism. Different sirtuins control similar cellular processes, suggesting a coordinated mode of action but information about potential cross-regulatory interactions within the sirtuin family is still limited. Here, we demonstrate that Sirt1 requires autodeacetylation to efficiently deacetylate targets such as p53, H3K9, and H4K16. Sirt7 restricts Sirt1 activity by preventing Sirt1 autodeacetylation causing enhanced Sirt1 activity in Sirt7-/- mice. Increased Sirt1 activity in Sirt7-/- mice blocks PPARγ and adipocyte differentiation, thereby diminishing accumulation of white fat. Thus, reduction of Sirt1 activity restores adipogenesis in Sirt7-/- adipocytes in vitro and in vivo. We disclosed a principle controlling Sirt1 activity and uncovered an unexpected complexity in the crosstalk between two different sirtuins. We propose that antagonistic interactions between Sirt1 and Sirt7 are pivotal in controlling the signaling network required for maintenance of adipose tissue.


Subject(s)
Adipogenesis/physiology , Adipose Tissue, White/cytology , Adipose Tissue/cytology , Sirtuin 1/physiology , Sirtuins/physiology , Acetylation , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Catalysis , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Conformation , Signal Transduction , Sirtuin 1/chemistry , Sirtuins/chemistry
10.
Circ Res ; 118(12): 1906-17, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27140435

ABSTRACT

RATIONALE: Activated cardiac fibroblasts (CF) are crucial players in the cardiac damage response; excess fibrosis, however, may result in myocardial stiffening and heart failure development. Inhibition of activated CF has been suggested as a therapeutic strategy in cardiac disease, but whether this truly improves cardiac function is unclear. OBJECTIVE: To study the effect of CF ablation on cardiac remodeling. METHODS AND RESULTS: We characterized subgroups of murine CF by single-cell expression analysis and identified periostin as the marker showing the highest correlation to an activated CF phenotype. We generated bacterial artificial chromosome-transgenic mice allowing tamoxifen-inducible Cre expression in periostin-positive cells as well as their diphtheria toxin-mediated ablation. In the healthy heart, periostin expression was restricted to valvular fibroblasts; ablation of this population did not affect cardiac function. After chronic angiotensin II exposure, ablation of activated CF resulted in significantly reduced cardiac fibrosis and improved cardiac function. After myocardial infarction, ablation of periostin-expressing CF resulted in reduced fibrosis without compromising scar stability, and cardiac function was significantly improved. Single-cell transcriptional analysis revealed reduced CF activation but increased expression of prohypertrophic factors in cardiac macrophages and cardiomyocytes, resulting in localized cardiomyocyte hypertrophy. CONCLUSIONS: Modulation of the activated CF population is a promising approach to prevent adverse cardiac remodeling in response to angiotensin II and after myocardial infarction.


Subject(s)
Cell Adhesion Molecules/metabolism , Fibroblasts/metabolism , Heart Ventricles/metabolism , Myocardial Infarction/metabolism , Ventricular Remodeling , Angiotensins/toxicity , Animals , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cells, Cultured , Fibroblasts/drug effects , Fibrosis , Heart Ventricles/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/etiology , Myocytes, Cardiac/metabolism
11.
Hum Mol Genet ; 24(3): 802-13, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25274778

ABSTRACT

Mutations in EMD, encoding emerin cause skeletal muscle and heart defects in patients with X-linked Emery-Dreifuss muscular dystrophy (X-EDMD) but the underlying mechanisms leading to cardiac defects are poorly understood. Here, we investigated the role of emerin in controlling cardiomyocyte proliferation and cardiac remodeling and explored its function in regulation of the Wnt/ß-catenin pathway. We observed a remarkable increase of cardiomyocytes in emerin-null adult mice accompanied with decreased numbers of multinucleated cells. Depletion of emerin in mouse ES cell-derived cardiomyocytes by shRNA caused hyperactivation of Wnt/ß-catenin signaling, increased proliferation and abrogated timely cardiac differentiation. Likewise, emerin-null mice exhibited increased Wnt/ß-catenin signaling, cardiac dysfunction and perturbed hypertrophic remodeling following pressure overload. Pharmacological inhibition of ß-catenin normalized proliferation and differentiation of ES cell-derived cardiomyocytes while inactivation of a single allele of ß-catenin efficiently rescued cardiac dysfunction in emerin-null mice. We conclude that emerin constrains ß-catenin signaling in the heart providing tight control of cardiomyocyte numbers. Enhanced Wnt/ß-catenin signaling seems to contribute to cardiac defects observed in X-EDMD. Hence, therapeutic inhibition of Wnt/ß-catenin signaling might be beneficial for X-EDMD patients.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/genetics , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/metabolism , Heart/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology
12.
Circ Res ; 115(2): 296-310, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24807786

ABSTRACT

RATIONALE: Myostatin is a major negative regulator of skeletal muscle mass and initiates multiple metabolic changes, including enhanced insulin sensitivity. However, the function of myostatin in the heart is barely understood, although it is upregulated in the myocardium under several pathological conditions. OBJECTIVE: Here, we aimed to decipher the role of myostatin and myostatin-dependent signaling pathways for cardiac function and cardiac metabolism in adult mice. To avoid potential counterregulatory mechanisms occurring in constitutive and germ-line-based myostatin mutants, we generated a mouse model that allows myostatin inactivation in adult cardiomyocytes. METHODS AND RESULTS: Cardiac MRI revealed that genetic inactivation of myostatin signaling in the adult murine heart caused cardiac hypertrophy and heart failure, partially recapitulating effects of the age-dependent decline of the myostatin paralog growth and differentiation factor 11. We found that myostatin represses AMP-activated kinase activation in the heart via transforming growth factor-ß-activated kinase 1, thereby preventing a metabolic switch toward glycolysis and glycogen accumulation. Furthermore, myostatin stimulated expression of regulator of G-protein signaling 2, a GTPase-activating protein that restricts Gaq and Gas signaling and thereby protects against cardiac failure. Inhibition of AMP-activated kinase in vivo rescued cardiac hypertrophy and prevented enhanced glycolytic flow and glycogen accumulation after inactivation of myostatin in cardiomyocytes. CONCLUSIONS: Our results uncover an important role of myostatin in the heart for maintaining cardiac energy homeostasis and preventing cardiac hypertrophy.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/genetics , Energy Metabolism/physiology , Heart Failure/prevention & control , Myocardium/metabolism , Myostatin/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Cardiomyopathy, Hypertrophic, Familial/complications , Cell Lineage , Gene Expression Regulation/physiology , Glycogen/metabolism , Glycolysis/physiology , Heart Failure/etiology , Homeostasis/physiology , MAP Kinase Kinase Kinases/physiology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myostatin/deficiency , RGS Proteins/physiology , Recombinant Fusion Proteins , Signal Transduction/physiology
13.
Am J Respir Crit Care Med ; 191(4): 437-47, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25536148

ABSTRACT

RATIONALE: Recent studies indicate that tumor-associated macrophages (MΦ) with an M2 phenotype can influence cancer progression and metastasis, but the regulatory pathways remain poorly characterized. OBJECTIVES: This study investigated the role of tumor-associated MΦ in lung cancer. METHODS: Coculturing of MΦ with mouse Lewis lung carcinoma (LLC1) and 10 different human lung cancer cell lines (adenocarcinoma, squamous cell carcinoma, and large cell carcinoma) caused up-regulation of CCR2/CCL2 and CX3CR1/CX3CL1 in both the cancer cells and the MΦ. MEASUREMENTS AND MAIN RESULTS: In the MΦ-tumor cell system, IL-10 drove CCR2 and CX3CR1 up-regulation, whereas CCL1, granulocyte colony-stimulating factor, and MIP1α were required for the up-regulation of CCL2 and CX3CL1. Downstream phenotypic effects included enhanced LLC1 proliferation and migration and MΦ M2 polarization. In vivo, MΦ depletion (clodronate, MΦ Fas-induced apoptosis mice) and genetic ablation of CCR2 and CX3CR1 all inhibited LLC1 tumor growth and metastasis, shifted tumor-associated MΦ toward M1 polarization, suppressed tumor vessel growth, and enhanced survival (metastasis model). Furthermore, mice treated with CCR2 antagonist mimicked genetic ablation of CCR2, showing reduced tumor growth and metastasis. In human lung cancer samples, tumor MΦ infiltration and CCR2 expression correlated with tumor stage and metastasis. CONCLUSIONS: Tumor-associated MΦ play a central role in lung cancer growth and metastasis, with bidirectional cross-talk between MΦ and cancer cells via CCR2 and CX3CR1 signaling as a central underlying mechanism. These findings suggest that the therapeutic strategy of blocking CCR2 and CX3CR1 may prove beneficial for halting lung cancer progression.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Large Cell/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Adenocarcinoma/pathology , Animals , CX3C Chemokine Receptor 1 , Carcinoma, Large Cell/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Chemokine CCL2/metabolism , Chemokine CX3CL1/metabolism , Humans , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Neoplasm Staging , Receptor Cross-Talk , Receptors, CCR2/metabolism , Receptors, Chemokine/metabolism , Up-Regulation
14.
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
15.
Proc Natl Acad Sci U S A ; 109(29): 11794-9, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22753500

ABSTRACT

Inflammation is a major factor in heart disease. IκB kinase (IKK) and its downstream target NF-κB are regulators of inflammation and are activated in cardiac disorders, but their precise contributions and targets are unclear. We analyzed IKK/NF-κB function in the heart by a gain-of-function approach, generating an inducible transgenic mouse model with cardiomyocyte-specific expression of constitutively active IKK2. In adult animals, IKK2 activation led to inflammatory dilated cardiomyopathy and heart failure. Transgenic hearts showed infiltration with CD11b(+) cells, fibrosis, fetal reprogramming, and atrophy of myocytes with strong constitutively active IKK2 expression. Upon transgene inactivation, the disease was reversible even at an advanced stage. IKK-induced cardiomyopathy was dependent on NF-κB activation, as in vivo expression of IκBα superrepressor, an inhibitor of NF-κB, prevented the development of disease. Gene expression and proteomic analyses revealed enhanced expression of inflammatory cytokines, and an IFN type I signature with activation of the IFN-stimulated gene 15 (ISG15) pathway. In that respect, IKK-induced cardiomyopathy resembled Coxsackievirus-induced myocarditis, during which the NF-κB and ISG15 pathways were also activated. Vice versa, in cardiomyocytes lacking the regulatory subunit of IKK (IKKγ/NEMO), the induction of ISG15 was attenuated. We conclude that IKK/NF-κB activation in cardiomyocytes is sufficient to cause cardiomyopathy and heart failure by inducing an excessive inflammatory response and myocyte atrophy.


Subject(s)
Cardiomyopathies/etiology , Enzyme Activation/physiology , Heart Failure/etiology , I-kappa B Kinase/metabolism , Myocytes, Cardiac/enzymology , NF-kappa B/metabolism , Analysis of Variance , Animals , Blotting, Western , CD11b Antigen/metabolism , Cardiomyopathies/enzymology , Cardiomyopathies/pathology , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Heart Failure/enzymology , Heart Failure/pathology , Histological Techniques , I-kappa B Proteins/metabolism , Luminescent Measurements , Mice , Mice, Transgenic , Microscopy, Fluorescence , NF-KappaB Inhibitor alpha
16.
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
17.
Sci Rep ; 14(1): 10998, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38745068

ABSTRACT

Using ferric chloride (FeCl3) to induce experimental superior sagittal sinus (SSS) thrombosis might interfere with magnetic resonance imaging (MRI)-assisted visualization and evaluation of the thrombus, the brain parenchyma, and the quality of the occlusion. The aim of this study was to investigate whether aluminum chloride (AlCl3)-induced thrombosis of the SSS has comparable properties to those of FeCl3 without causing artifacts in MRI. SSS thrombosis was induced in 14 male Wistar rats by exposure of the SSS and subsequent topical application of a filter paper strip soaked in AlCl3 (n = 7) or FeCl3 (n = 7) over a period of 15 min. The animals with AlCl3-induced SSS thrombosis showed a constant and complete occlusion with in histological analysis large thrombi. Blood flow measurements indicated a significant reduction on the first and seventh postoperative day compared to preoperative measurements. MRI enabled visualization and subsequent evaluation of the thrombus and the surrounding parenchyma. In comparison, FeCl3-induced SSS thrombosis could not be evaluated by MRI due to artifacts caused by the paramagnetic properties and increased susceptibility of FeCl3. The occluded sinus and the surrounding area appeared hypointense. The quality of SSS occlusion by AlCl3 was comparable to that of FeCl3. AlCl3 therefore represents a significant alternative substance in experimental SSS thrombosis ideally suited for studies using MRI.


Subject(s)
Aluminum Chloride , Artifacts , Chlorides , Disease Models, Animal , Ferric Compounds , Magnetic Resonance Imaging , Rats, Wistar , Animals , Magnetic Resonance Imaging/methods , Male , Rats , Chlorides/pharmacology , Chlorides/administration & dosage , Sagittal Sinus Thrombosis/diagnostic imaging , Sagittal Sinus Thrombosis/chemically induced , Aluminum Compounds , Superior Sagittal Sinus/diagnostic imaging , Superior Sagittal Sinus/drug effects
18.
Basic Res Cardiol ; 108(2): 325, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23325387

ABSTRACT

Pulmonary arterial hypertension (PAH) is a fatal disease for which no cure is yet available. The leading cause of death in PAH is right ventricular (RV) failure. Previously, the TNF receptor superfamily member fibroblast growth factor-inducible molecule 14 (Fn14) has been associated with different fibrotic diseases. However, so far there is no study demonstrating a causal role for endogenous Fn14 signaling in RV or LV heart disease. The purpose of this study was to determine whether global ablation of Fn14 prevents RV fibrosis and remodeling improving heart function. Here, we provide evidence for a causative role of Fn14 in pulmonary artery banding (PAB)-induced RV fibrosis and dysfunction in mice. Fn14 expression was increased in the RV after PAB. Mice lacking Fn14 (Fn14(-/-)) displayed substantially reduced RV fibrosis and dysfunction following PAB compared to wild-type littermates. Cell culture experiments demonstrated that activation of Fn14 induces collagen expression via RhoA-dependent nuclear translocation of myocardin-related transcription factor-A (MRTF-A)/MAL. Furthermore, activation of Fn14 in vitro caused fibroblast proliferation and myofibroblast differentiation, which corresponds to suppression of PAB-induced RV fibrosis in Fn14(-/-) mice. Moreover, our findings suggest that Fn14 expression is regulated by endothelin-1 (ET-1) in cardiac fibroblasts. We conclude that Fn14 is an endogenous key regulator in cardiac fibrosis and suggest this receptor as potential new target for therapeutic interventions in heart failure.


Subject(s)
Hypertrophy, Right Ventricular/prevention & control , Myocardium/pathology , Receptors, Tumor Necrosis Factor/physiology , Ventricular Dysfunction, Right/prevention & control , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Differentiation , Cell Proliferation , Collagen/metabolism , Cytokine TWEAK , Endothelin-1/physiology , Familial Primary Pulmonary Hypertension , Fibrosis/prevention & control , Fluorescent Antibody Technique , Hypertension, Pulmonary/complications , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Immunohistochemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myofibroblasts , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction , TWEAK Receptor , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism , Up-Regulation , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/physiopathology
19.
Cytokine ; 64(1): 43-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23764551

ABSTRACT

Cardiac pressure overload-induced hypertrophy and pathological remodelling frequently leads to right ventricular dysfunction, which is the most frequent cause of death in patients with pulmonary arterial hypertension. Nowadays, accumulating reports support the concept that proinflammatory cytokines and growth factors play crucial roles in the failing heart. We recently identified Fn14 as an endogenous key regulator in cardiac fibrosis in the PAB (Pulmonary Artery Banding) pressure-overload model. Right ventricular overload after PAB is also characterized by hypertrophy. The aim of this study was to determine whether right ventricular (RV) cardiac hypertrophy induced by PAB is mediated by the TWEAK/Fn14 axis. After baseline MRI, Fn14(-/-) mice and wild-type (WT) littermates were randomly assigned to two groups: (1) SHAM-operated (n⩾4, per genotype) and (2) PAB (n⩾11, per genotype). The results of MRI and histological analysis demonstrated that Fn14(-/-) mice exhibit less PAB-induced cardiac hypertrophy compared to WT littermates. Moreover, Fn14 overexpression in cultured adult rat cardiomyocytes enhanced cardiomyocyte size. Collectively, our studies demonstrate that Fn14 ablation attenuates RV hypertrophy after PAB and that activation of TWEAK/Fn14 signaling promotes cardiomyocyte growth in vitro. These results nominate Fn14 as a potential novel target for the treatment of heart hypertrophy.


Subject(s)
Hypertrophy, Right Ventricular/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Animals , Cells, Cultured , Familial Primary Pulmonary Hypertension , Heart/growth & development , Hypertension, Pulmonary/surgery , Mice , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Pulmonary Artery/surgery , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction , TWEAK Receptor
20.
Nat Commun ; 14(1): 1980, 2023 04 08.
Article in English | MEDLINE | ID: mdl-37031213

ABSTRACT

Activation of endothelial YAP/TAZ signaling is crucial for physiological and pathological angiogenesis. The mechanisms of endothelial YAP/TAZ regulation are, however, incompletely understood. Here we report that the protocadherin FAT1 acts as a critical upstream regulator of endothelial YAP/TAZ which limits the activity of these transcriptional cofactors during developmental and tumor angiogenesis by promoting their degradation. We show that loss of endothelial FAT1 results in increased endothelial cell proliferation in vitro and in various angiogenesis models in vivo. This effect is due to perturbed YAP/TAZ protein degradation, leading to increased YAP/TAZ protein levels and expression of canonical YAP/TAZ target genes. We identify the E3 ubiquitin ligase Mind Bomb-2 (MIB2) as a FAT1-interacting protein mediating FAT1-induced YAP/TAZ ubiquitination and degradation. Loss of MIB2 expression in endothelial cells in vitro and in vivo recapitulates the effects of FAT1 depletion and causes decreased YAP/TAZ degradation and increased YAP/TAZ signaling. Our data identify a pivotal mechanism of YAP/TAZ regulation involving FAT1 and its associated E3 ligase MIB2, which is essential for YAP/TAZ-dependent angiogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Trans-Activators , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Proteolysis , Trans-Activators/metabolism , Endothelial Cells/metabolism , YAP-Signaling Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Neovascularization, Pathologic/metabolism , Phosphoproteins/metabolism , Cadherins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL