Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circ Res ; 133(12): 966-988, 2023 12 08.
Article in English | MEDLINE | ID: mdl-37955182

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a chronic vascular disease characterized, among other abnormalities, by hyperproliferative smooth muscle cells and a perturbed cellular redox and metabolic balance. Oxidants induce cell cycle arrest to halt proliferation; however, little is known about the redox-regulated effector proteins that mediate these processes. Here, we report a novel kinase-inhibitory disulfide bond in cyclin D-CDK4 (cyclin-dependent kinase 4) and investigate its role in cell proliferation and PH. METHODS: Oxidative modifications of cyclin D-CDK4 were detected in human pulmonary arterial smooth muscle cells and human pulmonary arterial endothelial cells. Site-directed mutagenesis, tandem mass-spectrometry, cell-based experiments, in vitro kinase activity assays, in silico structural modeling, and a novel redox-dead constitutive knock-in mouse were utilized to investigate the nature and definitively establish the importance of CDK4 cysteine modification in pulmonary vascular cell proliferation. Furthermore, the cyclin D-CDK4 oxidation was assessed in vivo in the pulmonary arteries and isolated human pulmonary arterial smooth muscle cells of patients with pulmonary arterial hypertension and in 3 preclinical models of PH. RESULTS: Cyclin D-CDK4 forms a reversible oxidant-induced heterodimeric disulfide dimer between C7/8 and C135, respectively, in cells in vitro and in pulmonary arteries in vivo to inhibit cyclin D-CDK4 kinase activity, decrease Rb (retinoblastoma) protein phosphorylation, and induce cell cycle arrest. Mutation of CDK4 C135 causes a kinase-impaired phenotype, which decreases cell proliferation rate and alleviates disease phenotype in an experimental mouse PH model, suggesting this cysteine is indispensable for cyclin D-CDK4 kinase activity. Pulmonary arteries and human pulmonary arterial smooth muscle cells from patients with pulmonary arterial hypertension display a decreased level of CDK4 disulfide, consistent with CDK4 being hyperactive in human pulmonary arterial hypertension. Furthermore, auranofin treatment, which induces the cyclin D-CDK4 disulfide, attenuates disease severity in experimental PH models by mitigating pulmonary vascular remodeling. CONCLUSIONS: A novel disulfide bond in cyclin D-CDK4 acts as a rapid switch to inhibit kinase activity and halt cell proliferation. This oxidative modification forms at a critical cysteine residue, which is unique to CDK4, offering the potential for the design of a selective covalent inhibitor predicted to be beneficial in PH.


Subject(s)
Cyclins , Pulmonary Arterial Hypertension , Humans , Mice , Animals , Cyclins/metabolism , Pulmonary Arterial Hypertension/metabolism , Cysteine/metabolism , Endothelial Cells/metabolism , Cell Proliferation , Pulmonary Artery/metabolism , Phosphorylation , Cell Cycle Checkpoints , Cyclin D/metabolism , Cells, Cultured , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism
2.
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
3.
Am J Respir Crit Care Med ; 207(3): 283-299, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36047984

ABSTRACT

Rationale: Although type II alveolar epithelial cells (AEC2s) are chronically injured in idiopathic pulmonary fibrosis (IPF), they contribute to epithelial regeneration in IPF. Objectives: We hypothesized that Notch signaling may contribute to AEC2 proliferation, dedifferentiation characterized by loss of surfactant processing machinery, and lung fibrosis in IPF. Methods: We applied microarray analysis, kinome profiling, flow cytometry, immunofluorescence analysis, western blotting, quantitative PCR, and proliferation and surface activity analysis to study epithelial differentiation, proliferation, and matrix deposition in vitro (AEC2 lines, primary murine/human AEC2s), ex vivo (human IPF-derived precision-cut lung slices), and in vivo (bleomycin and pepstatin application, Notch1 [Notch receptor 1] intracellular domain overexpression). Measurements and Main Results: We document here extensive SP-B and -C (surfactant protein-B and -C) processing defects in IPF AEC2s, due to loss of Napsin A, resulting in increased intra-alveolar surface tension and alveolar collapse and induction of endoplasmic reticulum stress in AEC2s. In vivo pharmacological inhibition of Napsin A results in the development of AEC2 injury and overt lung fibrosis. We also demonstrate that Notch1 signaling is already activated early in IPF and determines AEC2 fate by inhibiting differentiation (reduced lamellar body compartment, reduced capacity to process hydrophobic SP) and by causing increased epithelial proliferation and development of lung fibrosis, putatively via altered JAK (Janus kinase)/Stat (signal transducer and activator of transcription) signaling in AEC2s. Conversely, inhibition of Notch signaling in IPF-derived precision-cut lung slices improved the surfactant processing capacity of AEC2s and reversed fibrosis. Conclusions: Notch1 is a central regulator of AEC2 fate in IPF. It induces alveolar epithelial proliferation and loss of Napsin A and of surfactant proprotein processing, and it contributes to fibroproliferation.


Subject(s)
Idiopathic Pulmonary Fibrosis , Pulmonary Surfactants , Humans , Mice , Animals , Surface-Active Agents , Lung , Alveolar Epithelial Cells , Bleomycin , Receptor, Notch1
4.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892401

ABSTRACT

Increased mitochondrial reactive oxygen species (ROS) formation is important for the development of right ventricular (RV) hypertrophy (RVH) and failure (RVF) during pulmonary hypertension (PH). ROS molecules are produced in different compartments within the cell, with mitochondria known to produce the strongest ROS signal. Among ROS-forming mitochondrial proteins, outer-mitochondrial-membrane-located monoamine oxidases (MAOs, type A or B) are capable of degrading neurotransmitters, thereby producing large amounts of ROS. In mice, MAO-B is the dominant isoform, which is present in almost all cell types within the heart. We analyzed the effect of an inducible cardiomyocyte-specific knockout of MAO-B (cmMAO-B KO) for the development of RVH and RVF in mice. Right ventricular hypertrophy was induced by pulmonary artery banding (PAB). RV dimensions and function were measured through echocardiography. ROS production (dihydroethidium staining), protein kinase activity (PamStation device), and systemic hemodynamics (in vivo catheterization) were assessed. A significant decrease in ROS formation was measured in cmMAO-B KO mice during PAB compared to Cre-negative littermates, which was associated with reduced activity of protein kinases involved in hypertrophic growth. In contrast to littermates in which the RV was dilated and hypertrophied following PAB, RV dimensions were unaffected in response to PAB in cmMAO-B KO mice, and no decline in RV systolic function otherwise seen in littermates during PAB was measured in cmMAO-B KO mice. In conclusion, cmMAO-B KO mice are protected against RV dilatation, hypertrophy, and dysfunction following RV pressure overload compared to littermates. These results support the hypothesis that cmMAO-B is a key player in causing RV hypertrophy and failure during PH.


Subject(s)
Hypertension, Pulmonary , Hypertrophy, Right Ventricular , Mice, Knockout , Monoamine Oxidase , Reactive Oxygen Species , Animals , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/pathology , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Monoamine Oxidase/deficiency , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Mice , Reactive Oxygen Species/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Heart Failure/metabolism , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/pathology , Male , Disease Models, Animal , Heart Ventricles/pathology , Heart Ventricles/metabolism , Ventricular Dysfunction, Right/metabolism , Ventricular Dysfunction, Right/genetics , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/pathology
5.
Herz ; 48(4): 285-290, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37079028

ABSTRACT

Right ventricular (RV) function is a critical determinant of the prognosis of patients with pulmonary hypertension (PH). Upon establishment of PH, RV dysfunction develops, leading to a gradual worsening of the condition over time, culminating in RV failure and premature mortality. Despite this understanding, the underlying mechanisms of RV failure remain obscure. As a result, there are currently no approved therapies specifically targeting the right ventricle. One contributing factor to the lack of RV-directed therapies is the complexity of the pathogenesis of RV failure as observed in animal models and clinical studies. In recent years, various research groups have begun utilizing multiple models, including both afterload-dependent and afterload-independent models, to investigate specific targets and pharmacological agents in RV failure. In this review, we examine various animal models of RV failure and the recent advancements made utilizing these models to study the mechanisms of RV failure and the potential efficacy of therapeutic interventions, with the ultimate goal of translating these findings into clinical practice to enhance the management of individuals with PH.


Subject(s)
Heart Failure , Hypertension, Pulmonary , Ventricular Dysfunction, Right , Animals , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/therapy , Heart Failure/therapy , Heart Failure/etiology , Heart Ventricles , Models, Theoretical , Ventricular Dysfunction, Right/diagnosis , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/therapy , Ventricular Function, Right
6.
Int J Mol Sci ; 24(16)2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37628831

ABSTRACT

Pulmonary arterial hypertension (PAH) is a complex disorder characterized by vascular remodeling and a consequent increase in pulmonary vascular resistance. The histologic hallmarks of PAH include plexiform and neointimal lesions of the pulmonary arterioles, which are composed of dysregulated, apoptosis-resistant endothelial cells and myofibroblasts. Platelet-derived growth factor receptors (PDGFR) α and ß, colony stimulating factor 1 receptor (CSF1R), and mast/stem cell growth factor receptor kit (c-KIT) are closely related kinases that have been implicated in PAH progression. In addition, emerging data indicate significant crosstalk between PDGF signaling and the bone morphogenetic protein receptor type 2 (BMPR2)/transforming growth factor ß (TGFß) receptor axis. This review will discuss the importance of the PDGFR-CSF1R-c-KIT signaling network in PAH pathogenesis, present evidence that the inhibition of all three nodes in this kinase network is a potential therapeutic approach for PAH, and highlight the therapeutic potential of seralutinib, currently in development for PAH, which targets these pathways.


Subject(s)
Pulmonary Arterial Hypertension , Humans , Endothelial Cells , Familial Primary Pulmonary Hypertension , Protein Kinase Inhibitors , Receptor Protein-Tyrosine Kinases , Proto-Oncogene Proteins c-kit
7.
J Gen Virol ; 103(6)2022 06.
Article in English | MEDLINE | ID: mdl-35771598

ABSTRACT

Constant evolution of influenza A viruses (IAVs) leads to the occurrence of new virus strains, which can cause epidemics and occasional pandemics. Here we compared two medically relevant IAVs, namely A/Hamburg/4/09 (H1N1pdm09) of the 2009 pandemic and the highly pathogenic avian IAV human isolate A/Thailand/1(KAN-1)/2004 (H5N1), for their ability to trigger intracellular phosphorylation patterns using a highly sensitive peptide-based kinase activity profiling approach. Virus-dependent tyrosine phosphorylations of substrate peptides largely overlap between the two viruses and are also strongly overrepresented in comparison to serine/threonine peptide phosphorylations. Both viruses trigger phosphorylations with distinct kinetics by overlapping and different kinases from which many form highly interconnected networks. As approximately half of the kinases forming a signalling hub have no known function for the IAV life cycle, we interrogated selected members of this group for their ability to interfere with IAV replication. These experiments revealed negative regulation of H1N1pdm09 and H5N1 replication by NUAK [novel (nua) kinase] kinases and by redundant ephrin A (EphA) receptor tyrosine kinases.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza, Human , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A virus/metabolism , Phosphorylation , Protein Kinases , Tyrosine , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
8.
Eur Respir J ; 59(4)2022 04.
Article in English | MEDLINE | ID: mdl-34475225

ABSTRACT

BACKGROUND: Pulmonary hypertension (PH) is a common complication of COPD, associated with increased mortality and morbidity. Intriguingly, pulmonary vascular alterations have been suggested to drive emphysema development. Previously, we identified inducible nitric oxide synthase (iNOS) as an essential enzyme for development and reversal of smoke-induced PH and emphysema, and showed that iNOS expression in bone-marrow-derived cells drives pulmonary vascular remodelling, but not parenchymal destruction. In this study, we aimed to identify the iNOS-expressing cell type driving smoke-induced PH and to decipher pro-proliferative pathways involved. METHODS: To address this question we used 1) myeloid-cell-specific iNOS knockout mice in chronic smoke exposure and 2) co-cultures of macrophages and pulmonary artery smooth muscle cells (PASMCs) to decipher underlying signalling pathways. RESULTS: Myeloid-cell-specific iNOS knockout prevented smoke-induced PH but not emphysema in mice. Moreover, iNOS deletion in myeloid cells ameliorated the increase in expression of CD206, a marker of M2 polarisation, on interstitial macrophages. Importantly, the observed effects on lung macrophages were hypoxia-independent, as these mice developed hypoxia-induced PH. In vitro, smoke-induced PASMC proliferation in co-cultures with M2-polarised macrophages could be abolished by iNOS deletion in phagocytic cells, as well as by extracellular signal-regulated kinase inhibition in PASMCs. Crucially, CD206-positive and iNOS-positive macrophages accumulated in proximity of remodelled vessels in the lungs of COPD patients, as shown by immunohistochemistry. CONCLUSION: In summary, our results demonstrate that iNOS deletion in myeloid cells confers protection against PH in smoke-exposed mice and provide evidence for an iNOS-dependent communication between M2-like macrophages and PASMCs in underlying pulmonary vascular remodelling.


Subject(s)
Emphysema , Hypertension, Pulmonary , Pulmonary Emphysema , Animals , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/prevention & control , Hypoxia , Macrophages/metabolism , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Smoke/adverse effects , Nicotiana/metabolism , Vascular Remodeling
9.
Proc Natl Acad Sci U S A ; 116(26): 13016-13025, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31186362

ABSTRACT

Chronic hypoxia causes pulmonary hypertension (PH), vascular remodeling, right ventricular (RV) hypertrophy, and cardiac failure. Protein kinase G Iα (PKGIα) is susceptible to oxidation, forming an interprotein disulfide homodimer associated with kinase targeting involved in vasodilation. Here we report increased disulfide PKGIα in pulmonary arteries from mice with hypoxic PH or lungs from patients with pulmonary arterial hypertension. This oxidation is likely caused by oxidants derived from NADPH oxidase-4, superoxide dismutase 3, and cystathionine γ-lyase, enzymes that were concomitantly increased in these samples. Indeed, products that may arise from these enzymes, including hydrogen peroxide, glutathione disulfide, and protein-bound persulfides, were increased in the plasma of hypoxic mice. Furthermore, low-molecular-weight hydropersulfides, which can serve as "superreductants" were attenuated in hypoxic tissues, consistent with systemic oxidative stress and the oxidation of PKGIα observed. Inhibiting cystathionine γ-lyase resulted in decreased hypoxia-induced disulfide PKGIα and more severe PH phenotype in wild-type mice, but not in Cys42Ser PKGIα knock-in (KI) mice that are resistant to oxidation. In addition, KI mice also developed potentiated PH during hypoxia alone. Thus, oxidation of PKGIα is an adaptive mechanism that limits PH, a concept further supported by polysulfide treatment abrogating hypoxia-induced RV hypertrophy in wild-type, but not in the KI, mice. Unbiased transcriptomic analysis of hypoxic lungs before structural remodeling identified up-regulation of endothelial-to-mesenchymal transition pathways in the KI compared with wild-type mice. Thus, disulfide PKGIα is an intrinsic adaptive mechanism that attenuates PH progression not only by promoting vasodilation but also by limiting maladaptive growth and fibrosis signaling.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Hypertension, Pulmonary/pathology , Hypoxia/complications , Pulmonary Artery/pathology , Adult , Animals , Cell Line , Cyclic GMP-Dependent Protein Kinase Type I/chemistry , Cystathionine gamma-Lyase/antagonists & inhibitors , Cystathionine gamma-Lyase/metabolism , Disease Models, Animal , Disease Progression , Disulfides/chemistry , Female , Fibrosis , Gene Knock-In Techniques , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/prevention & control , Hypoxia/blood , Hypoxia/drug therapy , Lung/blood supply , Lung/pathology , Male , Mice , Mice, Transgenic , Middle Aged , Oxidants/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Sulfides/administration & dosage , Sulfides/blood , Sulfides/metabolism , Up-Regulation , Vasoconstriction/drug effects , Vasodilation/drug effects
10.
Univers Access Inf Soc ; : 1-13, 2022 Jul 30.
Article in English | MEDLINE | ID: mdl-35966188

ABSTRACT

Technology use is a socially embedded process, especially when it comes to older adults and care. However, the restrictions associated with the COVID-19 pandemic have limited social contact to protect vulnerable groups in care homes, and even if technology use has increased in other areas, there is little known about the potential uptake of communication technology and changes in social interaction in the care context during a lasting crisis. This paper explores changes in communication technology use triggered by the pandemic at two care homes, using a qualitative diary study, online interviews and observations, and in-situ interviews within the care home with residents and workers. Our findings point to increasing use of tablets and video conference software triggered by COVID-related experiences, with implications for living and working in care homes. We also characterise the isolation experience of the residents, the workers' concerns about the residents and changes in social interaction. We observed new areas of technology usage, associated changing work practices, technical affinity issues and context-specific attitudes towards future technologies. While the pandemic has triggered the use of communication technology in care homes on a small scale, this has also caused increasing workload and in particular articulation work, which requires support structures and the re-definition of work roles.

11.
Am J Respir Cell Mol Biol ; 64(1): 100-114, 2021 01.
Article in English | MEDLINE | ID: mdl-33052714

ABSTRACT

In pulmonary arterial hypertension (PAH), progressive structural remodeling accounts for the pulmonary vasculopathy including the obliteration of the lung vasculature that causes an increase in vascular resistance and mean blood pressure in the pulmonary arteries ultimately leading to right heart failure-mediated death. Deciphering the molecular details of aberrant signaling of pulmonary vascular cells in PAH is fundamental for the development of new therapeutic strategies. We aimed to identify kinases as new potential drug targets that are dysregulated in PAH by means of a peptide-based kinase activity assay. We performed a tyrosine kinase-dependent phosphorylation assay using 144 selected microarrayed substrate peptides. The differential signature of phosphopeptides was used to predict alterations in tyrosine kinase activities in human pulmonary arterial smooth muscle cells (HPASMCs) from patients with idiopathic PAH (IPAH) compared with healthy control cells. Thereby, we observed an overactivation and an increased expression of Jak2 (Janus kinase 2) in HPASMCs from patients with IPAH as compared with controls. In vitro, IL-6-induced proliferation and migration of HPASMCs from healthy individuals as well as from patients with IPAH were reduced in a dose-dependent manner by the U.S. Food and Drug Administration-approved Jak1 and Jak2 inhibitor ruxolitinib. In vivo, ruxolitinib therapy in two experimental models of pulmonary arterial hypertension dose-dependently attenuated the elevation in pulmonary arterial pressure, partially reduced right ventricular hypertrophy, and almost completely restored cardiac index without signs of adverse events on cardiac function. Therefore, we propose that ruxolitinib may present a novel therapeutic option for patients with PAH by reducing pulmonary vascular remodeling through effectively blocking Jak2-Stat3 (signal transducer of activators of transcription)-mediated signaling pathways.


Subject(s)
Hypertension, Pulmonary/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Humans , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitriles , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pyrazoles/pharmacology , Pyrimidines , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Vascular Remodeling/drug effects , Vascular Remodeling/physiology , Vascular Resistance/drug effects , Vascular Resistance/physiology
12.
Int J Mol Sci ; 22(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540939

ABSTRACT

Pulmonary hypertension (PH) is characterized by a progressive elevation of mean arterial pressure followed by right ventricular failure and death. Previous studies have indicated that numerous inhibitors of receptor tyrosine kinase signaling could be either beneficial or detrimental for the treatment of PH. Here we investigated the therapeutic potential of the multi-kinase inhibitor regorafenib (BAY 73-4506) for the treatment of PH. A peptide-based kinase activity assay was performed using the PamStation®12 platform. The 5-bromo-2'-deoxyuridine proliferation and transwell migration assays were utilized in pulmonary arterial smooth muscle cells (PASMCs). Regorafenib was administered to monocrotaline- and hypoxia-induced PH in rats and mice, respectively. Functional parameters were analyzed by hemodynamic and echocardiographic measurements. The kinase activity assay revealed upregulation of twenty-nine kinases in PASMCs from patients with idiopathic PAH (IPAH), of which fifteen were established as potential targets of regorafenib. Regorafenib showed strong anti-proliferative and anti-migratory effects in IPAH-PASMCs compared to the control PASMCs. Both experimental models indicated improved cardiac function and reduced pulmonary vascular remodeling upon regorafenib treatment. In lungs from monocrotaline (MCT) rats, regorafenib reduced the phosphorylation of c-Jun N-terminal kinase and extracellular signal-regulated kinase 1/2. Overall, our data indicated that regorafenib plays a beneficial role in experimental PH.


Subject(s)
Hypertension, Pulmonary/drug therapy , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Animals , Cell Division/drug effects , Cell Movement/drug effects , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypoxia/complications , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Monocrotaline/toxicity , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phenylurea Compounds/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Pulmonary Artery/cytology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Vascular Remodeling/drug effects
13.
Brain Behav Immun ; 75: 251-257, 2019 01.
Article in English | MEDLINE | ID: mdl-30790541

ABSTRACT

Apoptosis is a genetically regulated form of programmed cell death which promotes the elimination of potentially detrimental immune cells. However, exercise-associated apoptosis is thought to induce a temporarily decline of the adaptive immune competence in the early post-exercise period. The purpose of the present study was to investigate if the aerobic endurance training status affects the sensitivity of human peripheral blood lymphocytes towards different types of apoptosis inducers and secondly, if this is mediated by the modulation of apoptosis-associated proteins and microRNAs. Collected at resting conditions, isolated lymphocytes of endurance trained athletes (ET) and healthy untrained subjects were either exposed to phytohemagglutinin-L (PHA-L), hydrogen peroxide (H2O2), or dexamethasone (DEX) as apoptosis inducer. Results revealed no significant differences between ET and UT in terms of lymphocyte apoptosis immediately following isolation as determined by flow cytometry using annexin V staining. After 24 h of ex vivo cultivation, lymphocytes of ET showed a reduced sensitivity to PHA-L-induced lymphocyte apoptosis which was accompanied by a noticeably up-regulation of the prominent apoptosis inhibitor genes X-linked inhibitor of apoptosis (XIAP) and Cyclin dependent kinase inhibitor 1B (CDKN1B) as analyzed by quantitative real-time PCR. Moreover, a trend was observed for the suppression of the corresponding pro-apoptotic miR-221. Lymphocyte apoptosis in control, H2O2 and DEX treated cells was not affected by aerobic endurance training status. However, distinct molecular signatures could be identified in un-treated control samples characterized by a counterbalanced modulation of pro- and anti-apoptotic mediators in ET. The results of the current study suggest that lymphocytes adapt to repetitive endurance exercise training by promoting lymphocyte homeostasis and increasing their resistance to apoptosis. This could be based on an up-regulation of anti-apoptotic proteins and a reduction in pro-apoptotic microRNAs which together tightly regulate the genetically defined apoptotic pathways governed by the type of apoptosis stimuli. Thus, the lymphocytes of endurance-trained athletes may be primed to counteract the transient immune suppression post-exercise.


Subject(s)
Apoptosis/physiology , Exercise/physiology , Lymphocytes/physiology , Adaptation, Physiological , Adult , Athletes , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dexamethasone/pharmacology , Endurance Training/methods , Gene Expression Regulation/physiology , Humans , Hydrogen Peroxide/pharmacology , Lymphocytes/metabolism , Male , MicroRNAs/metabolism , MicroRNAs/physiology , Phytohemagglutinins/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism
15.
Arterioscler Thromb Vasc Biol ; 33(1): 34-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23139294

ABSTRACT

OBJECTIVE: Mutations in the bone morphogenetic protein type II receptor (BMPR-II) are responsible for the majority of cases of heritable pulmonary arterial hypertension (PAH), and BMPR-II deficiency contributes to idiopathic and experimental forms of PAH. Sildenafil, a potent type-5 nucleotide-dependent phosphodiesterase inhibitor, is an established treatment for PAH, but whether sildenafil affects bone morphogenetic protein (BMP) signaling in the pulmonary circulation remains unknown. METHODS AND RESULTS: Studies were undertaken in human pulmonary arterial smooth muscle cells (PASMCs) and in vivo in the monocrotaline rat model of PAH. In PASMCs, sildenafil enhanced BMP4-induced phosphorylation of Smad1/5, Smad nuclear localization, and Inhibitor of DNA binding protein 1 gene and protein expression. This effect was mimicked by 8-bromo-cyclic GMP. Pharmacological inhibition or small interfering RNA knockdown of cyclic GMP-dependent protein kinase I inhibited the effect of sildenafil on BMP signaling. In functional studies, we observed that sildenafil potentiated the antiproliferative effects of BMP4 on PASMC proliferation. Furthermore, sildenafil restored the antiproliferative response to BMP4 in PASMCs harboring mutations in BMPR-II. In the monocrotaline rat model of PAH, which is characterized by BMPR-II deficiency, sildenafil prevented the development of pulmonary hypertension and vascular remodeling, and partly restored Smad1/5 phosphorylation and Inhibitor of DNA binding protein 1 gene expression in vivo in monocrotaline exposed rat lungs. CONCLUSIONS: Sildenafil enhances canonical BMP signaling via cyclic GMP and cyclic GMP-dependent protein kinase I in vitro and in vivo, and partly restores deficient BMP signaling in BMPR-II mutant PASMCs. Our findings demonstrate a novel mechanism of action of sildenafil in the treatment of PAH and suggest that targeting BMP signaling may be beneficial in this disease.


Subject(s)
Antihypertensive Agents/pharmacology , Bone Morphogenetic Proteins/metabolism , Hypertension, Pulmonary/prevention & control , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Piperazines/pharmacology , Pulmonary Artery/drug effects , Signal Transduction/drug effects , Sulfones/pharmacology , Vasodilator Agents/pharmacology , Animals , Binding Sites , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type II/drug effects , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Male , Monocrotaline , Muscle, Smooth, Vascular/metabolism , Mutation , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Promoter Regions, Genetic , Pulmonary Artery/metabolism , Purines/pharmacology , RNA Interference , Rats , Rats, Sprague-Dawley , Sildenafil Citrate , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Transfection
16.
Front Immunol ; 15: 1414195, 2024.
Article in English | MEDLINE | ID: mdl-38903521

ABSTRACT

Introduction: Protein kinases are indispensable reversible molecular switches that adapt and control protein functions during cellular processes requiring rapid responses to internal and external events. Bacterial infections can affect kinase-mediated phosphorylation events, with consequences for both innate and adaptive immunity, through regulation of antigen presentation, pathogen recognition, cell invasiveness and phagocytosis. Streptococcus pneumoniae (Spn), a human respiratory tract pathogen and a major cause of community-acquired pneumoniae, affects phosphorylation-based signalling of several kinases, but the pneumococcal mediator(s) involved in this process remain elusive. In this study, we investigated the influence of pneumococcal H2O2 on the protein kinase activity of the human lung epithelial H441 cell line, a generally accepted model of alveolar epithelial cells. Methods: We performed kinome analysis using PamGene microarray chips and protein analysis in Western blotting in H441 lung cells infected with Spn wild type (SpnWT) or with SpnΔlctOΔspxB -a deletion mutant strongly attenuated in H2O2 production- to assess the impact of pneumococcal hydrogen peroxide (H2O2) on global protein kinase activity profiles. Results: Our kinome analysis provides direct evidence that kinase activity profiles in infected H441 cells significantly vary according to the levels of pneumococcal H2O2. A large number of kinases in H441 cells infected with SpnWT are significantly downregulated, whereas this no longer occurs in cells infected with the mutant SpnΔlctOΔspxB strain, which lacks H2O2. In particular, we describe for the first time H2O2-mediated downregulation of Protein kinase B (Akt1) and activation of lymphocyte-specific tyrosine protein kinase (Lck) via H2O2-mediated phosphorylation.


Subject(s)
Hydrogen Peroxide , Streptococcus pneumoniae , Streptococcus pneumoniae/immunology , Hydrogen Peroxide/metabolism , Humans , Phosphorylation , Host-Pathogen Interactions/immunology , Cell Line , Protein Kinases/metabolism , Protein Kinases/genetics , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Signal Transduction
17.
Vascul Pharmacol ; 155: 107379, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762131

ABSTRACT

Pulmonary hypertension (PH) is a progressive, severe and to date not curable disease of the pulmonary vasculature. Alterations of the insulin-like growth factor 1 (IGF-1) system are known to play a role in vascular pathologies and IGF-binding proteins (IGFBPs) are important regulators of the bioavailability and function of IGFs. In this study, we show that circulating plasma levels of IGFBP-1, IGFBP-2 and IGFBP-3 are increased in idiopathic pulmonary arterial hypertension (IPAH) patients compared to healthy individuals. These binding proteins inhibit the IGF-1 induced IGF-1 receptor (IGF1R) phosphorylation and exhibit diverging effects on the IGF-1 induced signaling pathways in human pulmonary arterial cells (i.e. healthy as well as IPAH-hPASMCs, and healthy hPAECs). Furthermore, IGFBPs are differentially expressed in an experimental mouse model of PH. In hypoxic mouse lungs, IGFBP-1 mRNA expression is decreased whereas the mRNA for IGFBP-2 is increased. In contrast to IGFBP-1, IGFBP-2 shows vaso-constrictive properties in the murine pulmonary vasculature. Our analyses show that IGFBP-1 and IGFBP-2 exhibit diverging effects on IGF-1 signaling and display a unique IGF1R-independent kinase activation pattern in human pulmonary arterial smooth muscle cells (hPASMCs), which represent a major contributor of PAH pathobiology. Furthermore, we could show that IGFBP-2, in contrast to IGFBP-1, induces epidermal growth factor receptor (EGFR) signaling, Stat-3 activation and expression of Stat-3 target genes. Based on our results, we conclude that the IGFBP family, especially IGFBP-1, IGFBP-2 and IGFBP-3, are deregulated in PAH, that they affect IGF signaling and thereby regulate the cellular phenotype in PH.


Subject(s)
Disease Models, Animal , Insulin-Like Growth Factor Binding Protein 1 , Insulin-Like Growth Factor Binding Protein 2 , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor I , Myocytes, Smooth Muscle , Pulmonary Artery , Receptor, IGF Type 1 , Signal Transduction , Humans , Animals , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 1/genetics , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor I/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Cells, Cultured , Male , Insulin-Like Growth Factor Binding Protein 1/metabolism , Insulin-Like Growth Factor Binding Protein 1/genetics , Phosphorylation , STAT3 Transcription Factor/metabolism , Case-Control Studies , Mice, Inbred C57BL , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/physiopathology , Familial Primary Pulmonary Hypertension/pathology , Familial Primary Pulmonary Hypertension/genetics , Female , ErbB Receptors/metabolism , Middle Aged , Vascular Remodeling , Adult , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology
18.
Nat Commun ; 15(1): 87, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167746

ABSTRACT

Influenza A virus (IAV) infection mobilizes bone marrow-derived macrophages (BMDM) that gradually undergo transition to tissue-resident alveolar macrophages (TR-AM) in the inflamed lung. Combining high-dimensional single-cell transcriptomics with complex lung organoid modeling, in vivo adoptive cell transfer, and BMDM-specific gene targeting, we found that transitioning ("regenerative") BMDM and TR-AM highly express Placenta-expressed transcript 1 (Plet1). We reveal that Plet1 is released from alveolar macrophages, and acts as important mediator of macrophage-epithelial cross-talk during lung repair by inducing proliferation of alveolar epithelial cells and re-sealing of the epithelial barrier. Intratracheal administration of recombinant Plet1 early in the disease course attenuated viral lung injury and rescued mice from otherwise fatal disease, highlighting its therapeutic potential.


Subject(s)
Influenza A virus , Influenza, Human , Pneumonia, Viral , Animals , Female , Humans , Mice , Pregnancy , Lung , Macrophages, Alveolar , Placenta
19.
Front Robot AI ; 10: 1062714, 2023.
Article in English | MEDLINE | ID: mdl-37102131

ABSTRACT

Similar to human-human interaction (HHI), gaze is an important modality in conversational human-robot interaction (HRI) settings. Previously, human-inspired gaze parameters have been used to implement gaze behavior for humanoid robots in conversational settings and improve user experience (UX). Other robotic gaze implementations disregard social aspects of gaze behavior and pursue a technical goal (e.g., face tracking). However, it is unclear how deviating from human-inspired gaze parameters affects the UX. In this study, we use eye-tracking, interaction duration, and self-reported attitudinal measures to study the impact of non-human inspired gaze timings on the UX of the participants in a conversational setting. We show the results for systematically varying the gaze aversion ratio (GAR) of a humanoid robot over a broad parameter range from almost always gazing at the human conversation partner to almost always averting the gaze. The main results reveal that on a behavioral level, a low GAR leads to shorter interaction durations and that human participants change their GAR to mimic the robot. However, they do not copy the robotic gaze behavior strictly. Additionally, in the lowest gaze aversion setting, participants do not gaze back as much as expected, which indicates a user aversion to the robot gaze behavior. However, participants do not report different attitudes toward the robot for different GARs during the interaction. In summary, the urge of humans in conversational settings with a humanoid robot to adapt to the perceived GAR is stronger than the urge of intimacy regulation through gaze aversion, and a high mutual gaze is not always a sign of high comfort, as suggested earlier. This result can be used as a justification to deviate from human-inspired gaze parameters when necessary for specific robot behavior implementations.

20.
Metabolites ; 12(7)2022 Jul 16.
Article in English | MEDLINE | ID: mdl-35888781

ABSTRACT

Endurance training induces several adaptations in substrate metabolism, especially in relation to glycogen conservation. The study aimed to investigate differences in the metabolism of lipids, lipid-like substances, and amino acids between highly trained and untrained subjects using targeted metabolomics. Depending on their maximum relative oxygen uptake (VO2max), subjects were categorized as either endurance-trained (ET) or untrained (UT). Resting blood was taken and plasma isolated. It was screened for changes of 345 metabolites, including amino acids and biogenic amines, acylcarnitines, glycerophosphocholines (GPCs), sphingolipids, hexoses, bile acids, and polyunsaturated fatty acids (PUFAs) by using liquid chromatography coupled to tandem mass spectrometry. Acylcarnitine (C14:1, down in ET) and five GPCs (lysoPC a C18:2, up in ET; PC aa C42:0, up in ET; PC ae C38:2, up in ET; PC aa C38:5, down in ET; lysoPC a C26:0, down in ET) were differently regulated in ET compared to UT. TCDCA was down-regulated in athletes, while for three ratios of bile acids CA/CDCA, CA/(GCA+TCA), and DCA/(GDCA+TDCA) an up-regulation was found. TXB2 and 5,6-EET were down-regulated in the ET group and 18S-HEPE, a PUFA, showed higher levels in 18S-HEPE in endurance-trained subjects. For PC ae C38:2, TCDCA, and the ratio of cholic acid to chenodeoxycholic acid, an association with VO2max was found. Numerous phospholipids, acylcarnitines, glycerophosphocholines, bile acids, and PUFAs are present in varying concentrations at rest in ET. These results might represent an adaptation of lipid metabolism and account for the lowered cardiovascular risk profile of endurance athletes.

SELECTION OF CITATIONS
SEARCH DETAIL