Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Mol Cell Cardiol ; 193: 53-66, 2024 Aug.
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.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Autophagy , Cardiomyopathies , Myocytes, Cardiac , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/deficiency , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/deficiency , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Mice , Myocardium/metabolism , Myocardium/pathology , Chloroquine/pharmacology , Mice, Knockout
2.
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
4.
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
5.
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
6.
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
7.
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
8.
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
10.
Cardiovasc Res ; 118(12): 2665-2687, 2022 09 20.
Article in English | MEDLINE | ID: mdl-34609500

ABSTRACT

AIMS: Mammalian models have been instrumental in investigating adult heart function and human disease. However, electrophysiological differences with human hearts and high costs motivate the need for non-mammalian models. The zebrafish is a well-established genetic model to study cardiovascular development and function; however, analysis of cardiovascular phenotypes in adult specimens is particularly challenging as they are opaque. METHODS AND RESULTS: Here, we optimized and combined multiple imaging techniques including echocardiography, magnetic resonance imaging, and micro-computed tomography to identify and analyse cardiovascular phenotypes in adult zebrafish. Using alk5a/tgfbr1a mutants as a case study, we observed morphological and functional cardiovascular defects that were undetected with conventional approaches. Correlation analysis of multiple parameters revealed an association between haemodynamic defects and structural alterations of the heart, as observed clinically. CONCLUSION: We report a new, comprehensive, and sensitive platform to identify otherwise indiscernible cardiovascular phenotypes in adult zebrafish.


Subject(s)
Cardiovascular System , Zebrafish , Animals , Echocardiography , Heart , Humans , Mammals , X-Ray Microtomography , Zebrafish/genetics
11.
Ann Transl Med ; 9(13): 1061, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34422973

ABSTRACT

BACKGROUND: Early recanalization of an occluded vessel is associated with a better clinical outcome in acute ischemic stroke. Intravenous thrombolysis using recombinant tissue plasminogen activator (rt-PA) is only available in a minority of patients and often fails to reopen the occluded vessel. Mechanical recanalization is more effective in this matter but only available for selected patients when a thrombectomy centre can be reached. Therefore, sonothrombolysis might represent an alternative or complementary approach. Here, we tested microbubble-mediated sonothrombolysis (mmSTL) in a thromboembolic stroke model for middle cerebral artery occlusion (MCAO) in rats. METHODS: Sixty-seven male Wistar rats underwent MCAO using an autologous full blood thrombus and were randomly assigned to four groups receiving rt-PA, mmSTL, a combination of both, or a placebo. Diagnostic workup included neurological examination, assessment of infarct size, and presence of intracerebral haemorrhage by magnetic resonance imaging (MRI) and presence of microbleedings in histological staining. RESULTS: Neurological examination revealed no differences between the treatment groups. In all treatment groups, there was a reduction in infarct size 24 hours after MCAO as compared to the placebo (P≤0.05), but there were no differences between the active treatment groups (P>0.05) (placebo 0.75±0.10 cm3; mmSTL 0.43±0.07 cm3; rt-PA 0.4±0.07 cm3; mmSTL + rt-PA 0.27±0.08 cm3). Histological staining displayed intracerebral microbleedings in all animals. The frequency of gross bleeding detected by MRI did not differ between the groups (placebo 3; mmSTL 4; rt-PA 2; mmSTL + rt-PA 2; P>0.05) and was not associated with worse performance in clinical testing (P>0.05). There were no statistical differences in the mortality between the groups (P>0.05). CONCLUSIONS: Our study showed the efficacy and safety of mmSTL with or without rt-PA in an embolic rat stroke model using a continuous full blood thrombus. Sonothrombolysis might be useful for patients who need to be transported to a thrombectomy centre or for those with distal vessel occlusion.

12.
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
13.
Cancer Res ; 80(19): 4199-4211, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32816854

ABSTRACT

Although NF-κB is known to play a pivotal role in lung cancer, contributing to tumor growth, microenvironmental changes, and metastasis, the epigenetic regulation of NF-κB in tumor context is largely unknown. Here we report that the IKK2/NF-κB signaling pathway modulates metastasis-associated protein 2 (MTA2), a component of the nucleosome remodeling and deacetylase complex (NuRD). In triple transgenic mice, downregulation of IKK2 (Sftpc-cRaf-IKK2DN) in cRaf-induced tumors in alveolar epithelial type II cells restricted tumor formation, whereas activation of IKK2 (Sftpc-cRaf-IKK2CA) supported tumor growth; both effects were accompanied by altered expression of MTA2. Further studies employing genetic inhibition of MTA2 suggested that in primary tumor growth, independent of IKK2, MTA2/NuRD corepressor complex negatively regulates NF-κB signaling and tumor growth, whereas later dissociation of MTA2/NuRD complex from the promoter of NF-κB target genes and IKK2-dependent positive regulation of MTA2 leads to activation of NF-κB signaling, epithelial-mesenchymal transition, and lung tumor metastasis. These findings reveal a previously unrecognized biphasic role of MTA2 in IKK2/NF-κB-driven primary-to-metastatic lung tumor progression. Addressing the interaction between MTA2 and NF-κB would provide potential targets for intervention of tumor growth and metastasis. SIGNIFICANCE: These findings strongly suggest a prominent role of MTA2 in primary tumor growth, lung metastasis, and NF-κB signaling modulatory functions.


Subject(s)
Histone Deacetylases/metabolism , Lung Neoplasms/pathology , NF-kappa B/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone Deacetylases/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inflammation/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , Repressor Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Tumor Microenvironment
14.
Nat Commun ; 11(1): 2039, 2020 04 27.
Article in English | MEDLINE | ID: mdl-32341350

ABSTRACT

Long non-coding RNAs (lncRNAs) contribute to cardiac (patho)physiology. Aging is the major risk factor for cardiovascular disease with cardiomyocyte apoptosis as one underlying cause. Here, we report the identification of the aging-regulated lncRNA Sarrah (ENSMUST00000140003) that is anti-apoptotic in cardiomyocytes. Importantly, loss of SARRAH (OXCT1-AS1) in human engineered heart tissue results in impaired contractile force development. SARRAH directly binds to the promoters of genes downregulated after SARRAH silencing via RNA-DNA triple helix formation and cardiomyocytes lacking the triple helix forming domain of Sarrah show an increase in apoptosis. One of the direct SARRAH targets is NRF2, and restoration of NRF2 levels after SARRAH silencing partially rescues the reduction in cell viability. Overexpression of Sarrah in mice shows better recovery of cardiac contractile function after AMI compared to control mice. In summary, we identified the anti-apoptotic evolutionary conserved lncRNA Sarrah, which is downregulated by aging, as a regulator of cardiomyocyte survival.


Subject(s)
Apoptosis , Myocardial Infarction/genetics , Myocytes, Cardiac/cytology , RNA, Long Noncoding/genetics , Aging , Animals , Carrier Proteins/genetics , Cell Survival , Coenzyme A-Transferases/genetics , Disease Models, Animal , Gene Silencing , Humans , LIM Domain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , RNA, Antisense/genetics , RNA, Small Interfering/genetics , p300-CBP Transcription Factors/genetics
15.
Nat Commun ; 11(1): 1343, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165640

ABSTRACT

Enlarged vestibular aqueduct (EVA) is one of the most commonly identified inner ear malformations in hearing loss patients including Pendred syndrome. While biallelic mutations of the SLC26A4 gene, encoding pendrin, causes non-syndromic hearing loss with EVA or Pendred syndrome, a considerable number of patients appear to carry mono-allelic mutation. This suggests faulty pendrin regulatory machinery results in hearing loss. Here we identify EPHA2 as another causative gene of Pendred syndrome with SLC26A4. EphA2 forms a protein complex with pendrin controlling pendrin localization, which is disrupted in some pathogenic forms of pendrin. Moreover, point mutations leading to amino acid substitution in the EPHA2 gene are identified from patients bearing mono-allelic mutation of SLC26A4. Ephrin-B2 binds to EphA2 triggering internalization with pendrin inducing EphA2 autophosphorylation weakly. The identified EphA2 mutants attenuate ephrin-B2- but not ephrin-A1-induced EphA2 internalization with pendrin. Our results uncover an unexpected role of the Eph/ephrin system in epithelial function.


Subject(s)
Ephrin-A2/genetics , Goiter, Nodular/genetics , Hearing Loss, Sensorineural/genetics , Sulfate Transporters/genetics , Amino Acid Sequence , Animals , Ephrin-A1/genetics , Ephrin-A1/metabolism , Ephrin-A2/chemistry , Ephrin-A2/metabolism , Ephrin-B2/genetics , Ephrin-B2/metabolism , Goiter, Nodular/metabolism , Hearing Loss, Sensorineural/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Point Mutation , Protein Binding , Receptor, EphA2 , Sulfate Transporters/chemistry , Sulfate Transporters/metabolism
16.
Elife ; 82019 09 24.
Article in English | MEDLINE | ID: mdl-31549965

ABSTRACT

Myogenic vasoconstriction is an autoregulatory function of small arteries. Recently, G-protein-coupled receptors have been involved in myogenic vasoconstriction, but the downstream signalling mechanisms and the in-vivo-function of this myogenic autoregulation are poorly understood. Here, we show that small arteries from mice with smooth muscle-specific loss of G12/G13 or the Rho guanine nucleotide exchange factor ARHGEF12 have lost myogenic vasoconstriction. This defect was accompanied by loss of RhoA activation, while vessels showed normal increases in intracellular [Ca2+]. In the absence of myogenic vasoconstriction, perfusion of peripheral organs was increased, systemic vascular resistance was reduced and cardiac output and left ventricular mass were increased. In addition, animals with defective myogenic vasoconstriction showed aggravated hypotension in response to endotoxin. We conclude that G12/G13- and Rho-mediated signaling plays a key role in myogenic vasoconstriction and that myogenic tone is required to maintain local and systemic vascular resistance under physiological and pathological condition.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Vascular Resistance , Vasoconstriction , Animals , GTP-Binding Protein alpha Subunits, G12-G13/deficiency , Mice, Inbred C57BL , Rho Guanine Nucleotide Exchange Factors/deficiency
17.
Sci Rep ; 9(1): 12741, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31484989

ABSTRACT

Constitutive expression of the chemokine Mcp1 in mouse cardiomyocytes creates a model of inflammatory cardiomyopathy, with death from heart failure at age 7-8 months. A critical pathogenic role has previously been proposed for induced oxidative stress, involving NADPH oxidase activation. To test this idea, we exposed the mice to elevated oxygen levels. Against expectation, this prevented, rather than accelerated, the ultrastructural and functional signs of heart failure. This result suggests that the immune signaling initiated by Mcp1 leads instead to the inhibition of cellular oxygen usage, for which mitochondrial respiration is an obvious target. To address this hypothesis, we combined the Mcp1 model with xenotopic expression of the alternative oxidase (AOX), which provides a sink for electrons blocked from passage to oxygen via respiratory complexes III and IV. Ubiquitous AOX expression provided only a minor delay to cardiac functional deterioration and did not prevent the induction of markers of cardiac and metabolic remodeling considered a hallmark of the model. Moreover, cardiomyocyte-specific AOX expression resulted in exacerbation of Mcp1-induced heart failure, and failed to rescue a second cardiomyopathy model directly involving loss of cIV. Our findings imply that mitochondrial involvement in the pathology of inflammatory cardiomyopathy is multifaceted and complex.


Subject(s)
Cardiomyopathies/metabolism , Hyperoxia/metabolism , Mitochondrial Proteins/metabolism , Oxidoreductases/metabolism , Plant Proteins/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Ciona/enzymology , Disease Models, Animal , Humans , Hyperoxia/genetics , Hyperoxia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Oxidative Phosphorylation , Oxidoreductases/genetics , Oxygen/metabolism , Plant Proteins/genetics , Reactive Oxygen Species/metabolism , Ventricular Remodeling
18.
Nat Commun ; 10(1): 2130, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31086178

ABSTRACT

Hypoxia signaling plays a major role in non-malignant and malignant hyperproliferative diseases. Pulmonary hypertension (PH), a hypoxia-driven vascular disease, is characterized by a glycolytic switch similar to the Warburg effect in cancer. Ras association domain family 1A (RASSF1A) is a scaffold protein that acts as a tumour suppressor. Here we show that hypoxia promotes stabilization of RASSF1A through NOX-1- and protein kinase C- dependent phosphorylation. In parallel, hypoxia inducible factor-1 α (HIF-1α) activates RASSF1A transcription via HIF-binding sites in the RASSF1A promoter region. Vice versa, RASSF1A binds to HIF-1α, blocks its prolyl-hydroxylation and proteasomal degradation, and thus enhances the activation of the glycolytic switch. We find that this mechanism operates in experimental hypoxia-induced PH, which is blocked in RASSF1A knockout mice, in human primary PH vascular cells, and in a subset of human lung cancer cells. We conclude that RASSF1A-HIF-1α forms a feedforward loop driving hypoxia signaling in PH and cancer.


Subject(s)
Cell Hypoxia , Hypertension, Pulmonary/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Disease Models, Animal , Glycolysis , HEK293 Cells , HeLa Cells , Humans , Hypertension, Pulmonary/surgery , Lung/blood supply , Lung/pathology , Lung/surgery , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle , NADPH Oxidase 1/metabolism , Primary Cell Culture , Promoter Regions, Genetic/genetics , Protein Binding , Protein Kinase C/metabolism , Proteolysis , Pulmonary Artery/cytology , Signal Transduction , Tumor Suppressor Proteins/genetics
19.
Biomed Res Int ; 2018: 3293584, 2018.
Article in English | MEDLINE | ID: mdl-29511676

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and a rise in right ventricular (RV) afterload. The increased RV afterload leads to right ventricular failure (RVF) which is the reason for the high morbidity and mortality in PAH patients. The objective was to evaluate the therapeutic efficacy and antiremodeling potential of the phosphodiesterase type 5 (PDE5) inhibitor sildenafil and the soluble guanylate cyclase stimulator riociguat in a model of pressure overload RV hypertrophy induced by pulmonary artery banding (PAB). Mice subjected to PAB, one week after surgery, were treated with either sildenafil (100 mg/kg/d, n = 5), riociguat (30 mg/kg/d, n = 5), or vehicle (n = 5) for 14 days. RV function and remodeling were assessed by right heart catheterization, magnetic resonance imaging (MRI), and histomorphometry. Both sildenafil and riociguat prevented the deterioration of RV function, as determined by a decrease in RV dilation and restoration of the RV ejection fraction (EF). Although both compounds did not decrease right heart mass and cellular hypertrophy, riociguat prevented RV fibrosis induced by PAB. Both compounds diminished TGF-beta1 induced collagen synthesis of RV cardiac fibroblasts in vitro. Treatment with either riociguat or sildenafil prevented the progression of pressure overload-induced RVF, representing a novel therapeutic approach.


Subject(s)
Heart Failure/drug therapy , Hypertension, Pulmonary/drug therapy , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Sildenafil Citrate/administration & dosage , Ventricular Dysfunction, Right/drug therapy , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Disease Models, Animal , Heart/drug effects , Heart/physiopathology , Heart Failure/genetics , Heart Failure/pathology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Male , Mice , Phosphodiesterase 5 Inhibitors/administration & dosage , Pressure , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Vascular Remodeling/drug effects , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/pathology
SELECTION OF CITATIONS
SEARCH DETAIL