Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 172
Filtrer
1.
Int J Nanomedicine ; 19: 4411-4427, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774028

RÉSUMÉ

Background: Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease characterized by synovial inflammation and joint destruction. Despite progress in RA therapy, it remains difficult to achieve long-term remission in RA patients. Phosphodiesterase 3B (Pde3b) is a member of the phosphohydrolyase family that are involved in many signal transduction pathways. However, its role in RA is yet to be fully addressed. Methods: Studies were conducted in arthritic DBA/1 mice, a suitable mouse strain for collagen-induced rheumatoid arthritis (CIA), to dissect the role of Pde3b in RA pathogenesis. Next, RNAi-based therapy with Pde3b siRNA-loaded liposomes was assessed in a CIA model. To study the mechanism involved, we investigated the effect of Pde3b knockdown on macrophage polarization and related signaling pathway. Results: We demonstrated that mice with CIA exhibited upregulated Pde3b expression in macrophages. Notably, intravenous administration of liposomes loaded with Pde3b siRNA promoted the macrophage anti-inflammatory program and alleviated CIA in mice, as indicated by the reduced inflammatory response, synoviocyte infiltration, and bone and cartilage erosion. Mechanistic study revealed that depletion of Pde3b increased cAMP levels, by which it enhanced PKA-CREB-C/EBPß pathway to transcribe the expression of anti-inflammatory program-related genes. Conclusion: Our results support that Pde3b is involved in the pathogenesis of RA, and Pde3b siRNA-loaded liposomes might serve as a promising therapeutic approach against RA.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Cyclic Nucleotide Phosphodiesterases, Type 3 , Thérapie génétique , Liposomes , Macrophages , Souris de lignée DBA , Petit ARN interférent , Animaux , Liposomes/composition chimique , Liposomes/administration et posologie , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/induit chimiquement , Souris , Arthrite expérimentale/génétique , Arthrite expérimentale/prévention et contrôle , Arthrite expérimentale/thérapie , Macrophages/effets des médicaments et des substances chimiques , Petit ARN interférent/génétique , Petit ARN interférent/administration et posologie , Thérapie génétique/méthodes , Mâle , Transduction du signal/effets des médicaments et des substances chimiques
2.
Biochem Biophys Res Commun ; 696: 149489, 2024 Feb 12.
Article de Anglais | MEDLINE | ID: mdl-38244313

RÉSUMÉ

Lung cancer has a high incidence rate and requires more effective treatment strategies and drug options for clinical patients. EGFR is a common genetic alteration event in lung cancer that affects patient survival and drug strategy. Our study discovered aberrant aldolase A (ALDOA) expression and dysfunction in lung cancer patients with EGFR mutations. In addition to investigating relevant metabolic processes like glucose uptake, lactate production, and ATPase activity, we examined multi-omics profiles (transcriptomics, proteomics, and pull-down assays). It was observed that phosphodiesterase 3A (PDE3A) enzyme and ALDOA exhibit correlation, and furthermore, they impact M2 macrophage polarization through ß-catenin and downstream ID3. In addition to demonstrating the aforementioned mechanism of action, our experiments discovered that the PDE3 inhibitor trequinsin has a substantial impact on lung cancer cell lines with EGFR mutants. The trequinsin medication was found to decrease the M2 macrophage polarization status and several cancer phenotypes, in addition to transduction. These findings have potential prognostic and therapeutic applications for clinical patients with EGFR mutation and lung cancer.


Sujet(s)
Tumeurs du poumon , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Fructose bisphosphate aldolase/génétique , bêta-Caténine/génétique , bêta-Caténine/métabolisme , Transduction du signal/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Lignée cellulaire tumorale , Mutation , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Protéines tumorales/métabolisme , Protéines d'inhibition de la différenciation/génétique
3.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 4911-4925, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38165426

RÉSUMÉ

Cuproptosis is a new Cu-dependent programmed cell death manner that has shown regulatory functions in many tumor types, however, its mechanism in bladder cancer remains unclear. Here, we reveal that Phosphodiesterase 3B (PDE3B), a cuproptosis-associated gene, could reduce the invasion and migration of bladder cancer. PDE3B is downregulated in bladder cancer tissues, which is correlated with better prognosis. Conversely, overexpression of PDE3B in bladder cancer cell could significantly resist invasion and migration, which is consistent with the TCGA database results. Future study demonstrate the anti-cancer effect of PDE3B is mediated by Keratin 6B (KRT6B) which leads to the keratinization. Therefore, PDE3B can reduce KRT6B expression and inhibit the invasion and migration of bladder cancer. Meanwhile, increased expression of PDE3B was able to enhance the sensitivity of Cuproptosis drug thiram. This study show that PDE3B/KRT6B is a potential cancer therapeutic target and PDE3B activation is able to increase the sensitivity of bladder cancer cells to copper ionophores.


Sujet(s)
Mouvement cellulaire , Cuivre , Cyclic Nucleotide Phosphodiesterases, Type 3 , Kératine-6 , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Lignée cellulaire tumorale , Cuivre/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Kératine-6/métabolisme , Kératine-6/génétique , Invasion tumorale , Régulation de l'expression des gènes tumoraux
4.
Hypertension ; 80(6): 1171-1179, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37035914

RÉSUMÉ

Hypertension with brachydactyly (HTNB) represents an autosomal dominant form of hypertension. It is a rare syndrome, in which the blood pressure can rise by more than 50 mmHg. If untreated, the patients die of stroke by the age of 50 years. In HTNB, vascular smooth muscle cell proliferation is increased, vasodilation compromised, and the kidney not affected. Surprisingly, after decades of hypertension, HTNB is not associated with hypertension-induced cardiac damage. HTNB is caused by gain-of-function mutations in the PDE3A (phosphodiesterase 3A) gene. The mutant enzymes are hyperactive. PDE3A (phosphodiesterase 3A) hydrolyzes and thereby terminates cyclic adenosine monophosphate signaling in defined cellular compartments. The cardioprotective effect involves local changes of cyclic adenosine monophosphate signaling and inhibition of Ca2+ reuptake into the sarcoplasmic reticulum of cardiac myocytes. This review introduces HTNB and discusses how insight into the molecular mechanisms underlying HTNB could contribute to a better understanding of blood pressure control and lead to PDE3A-directed strategies for the treatment of essential hypertension and the prevention of hypertension-induced cardiac damage. A focus will be on cAMP (cyclic adenosine monophosphate) signaling compartments.


Sujet(s)
Hypertension artérielle , Humains , Adulte d'âge moyen , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Hypertension artérielle/génétique , Mutation , Myocytes cardiaques , AMP
5.
Circulation ; 147(16): 1221-1236, 2023 04 18.
Article de Anglais | MEDLINE | ID: mdl-36876489

RÉSUMÉ

BACKGROUND: Increasing SERCA2 (sarco[endo]-plasmic reticulum Ca2+ ATPase 2) activity is suggested to be beneficial in chronic heart failure, but no selective SERCA2-activating drugs are available. PDE3A (phosphodiesterase 3A) is proposed to be present in the SERCA2 interactome and limit SERCA2 activity. Disruption of PDE3A from SERCA2 might thus be a strategy to develop SERCA2 activators. METHODS: Confocal microscopy, 2-color direct stochastic optical reconstruction microscopy, proximity ligation assays, immunoprecipitations, peptide arrays, and surface plasmon resonance were used to investigate colocalization between SERCA2 and PDE3A in cardiomyocytes, map the SERCA2/PDE3A interaction sites, and optimize disruptor peptides that release PDE3A from SERCA2. Functional experiments assessing the effect of PDE3A-binding to SERCA2 were performed in cardiomyocytes and HEK293 vesicles. The effect of SERCA2/PDE3A disruption by the disruptor peptide OptF (optimized peptide F) on cardiac mortality and function was evaluated during 20 weeks in 2 consecutive randomized, blinded, and controlled preclinical trials in a total of 148 mice injected with recombinant adeno-associated virus 9 (rAAV9)-OptF, rAAV9-control (Ctrl), or PBS, before undergoing aortic banding (AB) or sham surgery and subsequent phenotyping with serial echocardiography, cardiac magnetic resonance imaging, histology, and functional and molecular assays. RESULTS: PDE3A colocalized with SERCA2 in human nonfailing, human failing, and rodent myocardium. Amino acids 277-402 of PDE3A bound directly to amino acids 169-216 within the actuator domain of SERCA2. Disruption of PDE3A from SERCA2 increased SERCA2 activity in normal and failing cardiomyocytes. SERCA2/PDE3A disruptor peptides increased SERCA2 activity also in the presence of protein kinase A inhibitors and in phospholamban-deficient mice, and had no effect in mice with cardiomyocyte-specific inactivation of SERCA2. Cotransfection of PDE3A reduced SERCA2 activity in HEK293 vesicles. Treatment with rAAV9-OptF reduced cardiac mortality compared with rAAV9-Ctrl (hazard ratio, 0.26 [95% CI, 0.11 to 0.63]) and PBS (hazard ratio, 0.28 [95% CI, 0.09 to 0.90]) 20 weeks after AB. Mice injected with rAAV9-OptF had improved contractility and no difference in cardiac remodeling compared with rAAV9-Ctrl after aortic banding. CONCLUSIONS: Our results suggest that PDE3A regulates SERCA2 activity through direct binding, independently of the catalytic activity of PDE3A. Targeting the SERCA2/PDE3A interaction prevented cardiac mortality after AB, most likely by improving cardiac contractility.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 3 , Défaillance cardiaque , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animaux , Humains , Souris , Calcium/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Défaillance cardiaque/métabolisme , Cellules HEK293 , Myocarde/métabolisme , Myocytes cardiaques/métabolisme , Réticulum sarcoplasmique/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme
6.
Circulation ; 146(23): 1758-1778, 2022 12 06.
Article de Anglais | MEDLINE | ID: mdl-36259389

RÉSUMÉ

BACKGROUND: Phosphodiesterase 3A (PDE3A) gain-of-function mutations cause hypertension with brachydactyly (HTNB) and lead to stroke. Increased peripheral vascular resistance, rather than salt retention, is responsible. It is surprising that the few patients with HTNB examined so far did not develop cardiac hypertrophy or heart failure. We hypothesized that, in the heart, PDE3A mutations could be protective. METHODS: We studied new patients. CRISPR-Cas9-engineered rat HTNB models were phenotyped by telemetric blood pressure measurements, echocardiography, microcomputed tomography, RNA-sequencing, and single nuclei RNA-sequencing. Human induced pluripotent stem cells carrying PDE3A mutations were established, differentiated to cardiomyocytes, and analyzed by Ca2+ imaging. We used Förster resonance energy transfer and biochemical assays. RESULTS: We identified a new PDE3A mutation in a family with HTNB. It maps to exon 13 encoding the enzyme's catalytic domain. All hitherto identified HTNB PDE3A mutations cluster in exon 4 encoding a region N-terminally from the catalytic domain of the enzyme. The mutations were recapitulated in rat models. Both exon 4 and 13 mutations led to aberrant phosphorylation, hyperactivity, and increased PDE3A enzyme self-assembly. The left ventricles of our patients with HTNB and the rat models were normal despite preexisting hypertension. A catecholamine challenge elicited cardiac hypertrophy in HTNB rats only to the level of wild-type rats and improved the contractility of the mutant hearts, compared with wild-type rats. The ß-adrenergic system, phosphodiesterase activity, and cAMP levels in the mutant hearts resembled wild-type hearts, whereas phospholamban phosphorylation was decreased in the mutants. In our induced pluripotent stem cell cardiomyocyte models, the PDE3A mutations caused adaptive changes of Ca2+ cycling. RNA-sequencing and single nuclei RNA-sequencing identified differences in mRNA expression between wild-type and mutants, affecting, among others, metabolism and protein folding. CONCLUSIONS: Although in vascular smooth muscle, PDE3A mutations cause hypertension, they confer protection against hypertension-induced cardiac damage in hearts. Nonselective PDE3A inhibition is a final, short-term option in heart failure treatment to increase cardiac cAMP and improve contractility. Our data argue that mimicking the effect of PDE3A mutations in the heart rather than nonselective PDE3 inhibition is cardioprotective in the long term. Our findings could facilitate the search for new treatments to prevent hypertension-induced cardiac damage.


Sujet(s)
Défaillance cardiaque , Hypertension artérielle , Cellules souches pluripotentes induites , Humains , Rats , Animaux , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Microtomographie aux rayons X , Cellules souches pluripotentes induites/métabolisme , Hypertension artérielle/complications , Hypertension artérielle/génétique , Myocytes cardiaques/métabolisme , Cardiomégalie , ARN
7.
Phytother Res ; 36(9): 3540-3554, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35703011

RÉSUMÉ

Protein signaling complexes play important roles in prevention of several cancer types and can be used for development of targeted therapy. The roles of signaling complexes of phosphodiesterase 3B (PDE3B) and Rap guanine nucleotide exchange factor 3 (RAPGEF3), which are two important enzymes of cyclic adenosine monophosphate (cAMP) metabolism, in cancer have not been fully explored. In the current study, a natural product Kaempferol-3-O-(3'',4''-di-E-p-coumaroyl)-α-L-rhamnopyranoside designated as KOLR was extracted from Cinnamomum pauciflorum Nees leaves. KOLR exhibited higher cytotoxic effects against BxCP-3 pancreatic cancer cell line. In BxPC-3 cells, the KOLR could enhance the formation of RAPGEF 3/ PDE3B protein complex to inhibit the activation of Rap-1 and PI3K-AKT pathway, thereby promoting cell apoptosis and inhibiting cell metastasis. Mutation of RAPGEF3 G557A or low expression of PDE3B inactivated the binding action of KOLR resulting in KOLR resistance. The findings of this study show that PDE3B/RAPGEF3 complex is a potential therapeutic cancer target.


Sujet(s)
Cinnamomum , Phosphatidylinositol 3-kinases , AMP cyclique/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Facteurs d'échange de nucléotides guanyliques/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Feuilles de plante/métabolisme
8.
Cell Signal ; 94: 110322, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35346821

RÉSUMÉ

Cyclic-AMP (cAMP), the first second messenger to be identified, is synthesized, and is universally utilized as a second messenger, and plays important roles in integrity, and function of organs, including heart. Through its coupling with other intracellular messengers, cAMP facilitates excitation-contraction coupling, increases heart rate and conduction velocity. It is degraded by a class of enzymes called cAMP-dependent phosphodiesterase (PDE), with PDE3 and PDE4 being the predominant isoforms in the heart. This highly diverse class of enzymes degrade cAMP and through anchoring proteins generates dynamic microdomains to target specific proteins and control specific cell functions in response to various stimuli. The impaired function of the anchoring protein either by inherited genetic mutations or acquired injuries results in altered intracellular targeting, and blunted responsiveness to stimulating pathways and contributes to pathological cardiac remodeling, cardiac arrhythmias and reduced cell survival. Recent genetic studies provide compelling evidence for an association between the variants in the anchoring protein PDE4DIP and atrial fibrillation, stroke, and heart failure.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Défaillance cardiaque , 3',5'-Cyclic-AMP Phosphodiesterases/métabolisme , AMP cyclique/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 4/génétique , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Coeur , Humains , Systèmes de seconds messagers
9.
Cell Chem Biol ; 29(6): 958-969.e5, 2022 06 16.
Article de Anglais | MEDLINE | ID: mdl-35104454

RÉSUMÉ

The canonical function of phosphodiesterase 3A (PDE3A) is to hydrolyze the phosphodiester bonds in second messenger molecules, such as cyclic AMP (cAMP) and cyclic guanosine monophosphate (cGMP). Recently, a phosphodiesterase-activity-independent role for PDE3A was reported. In this noncanonical function, PDE3A physically interacts with Schlafen 12 (SLFN12) upon treatment of cells with cytotoxic PDE3A modulators. Here, we confirmed that the cytotoxic PDE3A modulators act as molecular glues to initiate the association of PDE3A and SLFN12. The PDE3A-SLFN12 interaction increases the protein stability of SLFN12 located in the cytoplasm, while at the same time also inducing SLFN12 dephosphorylation (including serines 368 and 573). Mutational analysis demonstrates that dephosphorylation is required for cell death induced by cytotoxic PDE3A modulators. Finally, we found that dephosphorylation promoted the rRNA RNase activity of SLFN12 and show that this nucleolytic activity is essential for SLFN12's cell-death-inducing function. Thus, our study deepens the understanding of the biochemical mechanisms underlying SLFN12-mediated cell death.


Sujet(s)
Antinéoplasiques , AMP cyclique , Antinéoplasiques/pharmacologie , Mort cellulaire , AMP cyclique/métabolisme , GMP cyclique , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme
10.
Bioengineered ; 13(2): 2217-2225, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35030973

RÉSUMÉ

Diabetes mellitus is a metabolic disease caused by defective insulin secretion and/or insulin action. And insulin is the main hormone released by the pancreatic ß-cells. Diosgenin (DG) is a phytochemical with pharmacological activity that increases insulin secretion in streptozotocin (STZ)-induced pancreatic ß-cells of diabetic rats. In this paper, we investigated the effect and mechanism of DG on cell apoptosis and dysfunction in STZ-induced pancreatic ß-cells. Cell viability was detected by CCK-8, apoptosis by flow cytometry, and apoptosis-related protein expression by Western blot. Western blot and RT-qPCR were performed to detect the expression of related genes. The results showed that in STZ-induced INS-1 cells, DG could improve cell viability, inhibit apoptosis, attenuate oxidative stress levels and increase insulin secretion. Notably, PDE3B was highly expressed in STZ-induced INS-1 cells, while DG could significantly inhibit PDE3B expression in a dose-dependent manner. More importantly, overexpression PDE3B remarkably reversed the effect of DG on STZ-induced INS-1 cells. It is thus clear that DG might inhibit STZ-treated pancreatic ß-cell apoptosis and reduce dysfunction via downregulating PDE3B, which provided a more reliable theoretical basis for the treatment of diabetes mellitus with DG.


Sujet(s)
AMP-Activated Protein Kinases/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Cellules à insuline/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Streptozocine/toxicité , Sérine-thréonine kinases TOR/métabolisme , AMP-Activated Protein Kinases/génétique , Animaux , Apoptose/génétique , Lignée cellulaire tumorale , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Rats , Transduction du signal/génétique , Sérine-thréonine kinases TOR/génétique
11.
Sci Rep ; 11(1): 19365, 2021 09 29.
Article de Anglais | MEDLINE | ID: mdl-34588469

RÉSUMÉ

Genome-wide association studies have identified numerous common genetic variants associated with spirometric measures of pulmonary function, including forced expiratory volume in one second (FEV1), forced vital capacity, and their ratio. However, variants with lower minor allele frequencies are less explored. We conducted a large-scale gene-smoking interaction meta-analysis on exonic rare and low-frequency variants involving 44,429 individuals of European ancestry in the discovery stage and sought replication in the UK BiLEVE study with 45,133 European ancestry samples and UK Biobank study with 59,478 samples. We leveraged data on cigarette smoking, the major environmental risk factor for reduced lung function, by testing gene-by-smoking interaction effects only and simultaneously testing the genetic main effects and interaction effects. The most statistically significant signal that replicated was a previously reported low-frequency signal in GPR126, distinct from common variant associations in this gene. Although only nominal replication was obtained for a top rare variant signal rs142935352 in one of the two studies, interaction and joint tests for current smoking and PDE3B were significantly associated with FEV1. This study investigates the utility of assessing gene-by-smoking interactions and underscores their effects on potential pulmonary function.


Sujet(s)
Fumer des cigarettes/épidémiologie , Volume expiratoire maximal par seconde/génétique , Interaction entre gènes et environnement , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Fumer des cigarettes/effets indésirables , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Jeux de données comme sujet , Exons/génétique , Études de faisabilité , Femelle , Étude d'association pangénomique , Humains , Poumon/physiologie , Mâle , Adulte d'âge moyen , Polymorphisme de nucléotide simple , Récepteurs couplés aux protéines G/génétique , Facteurs de risque
12.
Cells ; 10(8)2021 08 05.
Article de Anglais | MEDLINE | ID: mdl-34440764

RÉSUMÉ

OBJECTIVE: platelets possess not only haemostatic but also inflammatory properties, which combined are thought to play a detrimental role in thromboinflammatory diseases such as acute coronary syndromes and stroke. Phosphodiesterase (PDE) 3 and -5 inhibitors have demonstrated efficacy in secondary prevention of arterial thrombosis, partially mediated by their antiplatelet action. Yet it is unclear whether such inhibitors also affect platelets' inflammatory functions. Here, we aimed to examine the effect of the PDE3A inhibitor cilostazol and the PDE5 inhibitor tadalafil on platelet function in various aspects of thromboinflammation. Approach and results: cilostazol, but not tadalafil, delayed ex vivo platelet-dependent fibrin formation under whole blood flow over type I collagen at 1000 s-1. Similar results were obtained with blood from Pde3a deficient mice, indicating that cilostazol effects are mediated via PDE3A. Interestingly, cilostazol specifically reduced the release of phosphatidylserine-positive extracellular vesicles (EVs) from human platelets while not affecting total EV release. Both cilostazol and tadalafil reduced the interaction of human platelets with inflamed endothelium under arterial flow and the release of the chemokines CCL5 and CXCL4 from platelets. Moreover, cilostazol, but not tadalafil, reduced monocyte recruitment and platelet-monocyte interaction in vitro. CONCLUSIONS: this study demonstrated yet unrecognised roles for platelet PDE3A and platelet PDE5 in platelet procoagulant and proinflammatory responses.


Sujet(s)
Anti-inflammatoires/pharmacologie , Plaquettes/effets des médicaments et des substances chimiques , Cilostazol/pharmacologie , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Fibrinolytiques/pharmacologie , Inhibiteurs de la phosphodiestérase-3/pharmacologie , Activation plaquettaire/effets des médicaments et des substances chimiques , Animaux , Coagulation sanguine/effets des médicaments et des substances chimiques , Plaquettes/enzymologie , Plaquettes/immunologie , Cellules cultivées , Chimiokines/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Fibrine/métabolisme , Humains , Médiateurs de l'inflammation/métabolisme , Souris de lignée C57BL , Souris knockout , Inhibiteurs de la phosphodiestérase-5/pharmacologie , Adhésivité plaquettaire/effets des médicaments et des substances chimiques , Transduction du signal , Tadalafil/pharmacologie
13.
Nat Commun ; 12(1): 4375, 2021 07 16.
Article de Anglais | MEDLINE | ID: mdl-34272366

RÉSUMÉ

DNMDP and related compounds, or velcrins, induce complex formation between the phosphodiesterase PDE3A and the SLFN12 protein, leading to a cytotoxic response in cancer cells that express elevated levels of both proteins. The mechanisms by which velcrins induce complex formation, and how the PDE3A-SLFN12 complex causes cancer cell death, are not fully understood. Here, we show that PDE3A and SLFN12 form a heterotetramer stabilized by binding of DNMDP. Interactions between the C-terminal alpha helix of SLFN12 and residues near the active site of PDE3A are required for complex formation, and are further stabilized by interactions between SLFN12 and DNMDP. Moreover, we demonstrate that SLFN12 is an RNase, that PDE3A binding increases SLFN12 RNase activity, and that SLFN12 RNase activity is required for DNMDP response. This new mechanistic understanding will facilitate development of velcrin compounds into new cancer therapies.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/composition chimique , Protéines et peptides de signalisation intracellulaire/composition chimique , Pyridazines/composition chimique , AMP/composition chimique , Calorimétrie différentielle à balayage , Domaine catalytique , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/génétique , Cryomicroscopie électronique , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Endoribonucleases/composition chimique , Cellules HEK293 , Cellules HeLa , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Cinétique , Spectrométrie de masse , Complexes multienzymatiques/ultrastructure , Mutation , Liaison aux protéines , Structure en hélice alpha , Multimérisation de protéines , Pyridazines/pharmacologie , Protéines recombinantes , Tétrahydroisoquinoléines/composition chimique
14.
J Cardiovasc Pharmacol ; 78(5): e749-e760, 2021 11 01.
Article de Anglais | MEDLINE | ID: mdl-34321402

RÉSUMÉ

ABSTRACT: Atherosclerosis (AS) is a major risk factor for cardiovascular disease, in which circular RNAs play important regulatory roles. This research aimed to explore the biological role of circular RNA Sterol Regulatory Element Binding Transcription Factor Chaperone (circSCAP) (hsa_circ_0001292) in AS development. Real-time PCR or Western blot assay was conducted to analyze RNA or protein expression. Cell proliferation and apoptosis were analyzed by CCK-8 assay and flow cytometry. The levels of lipid accumulation-associated indicators and oxidative stress factors were detected using commercial kits. The levels of inflammatory cytokines were examined using enzyme-linked immunosorbent assay. Intermolecular interaction was verified by dual-luciferase reporter analysis or RNA pull-down analysis. CircSCAP and phosphodiesterase 3B (PDE3B) levels were elevated, whereas the miR-221-5p level was decreased in patients with AS and oxidized low-density lipoprotein (ox-LDL)-induced THP-1 cells. CircSCAP absence suppressed lipid deposition, inflammation, and oxidative stress in ox-LDL-induced THP-1 cells. MiR-221-5p was a target of circSCAP, and anti-miR-221-5p largely reversed si-circSCAP-induced effects in ox-LDL-induced THP-1 cells. PDE3B was a target of miR-221-5p, and PDE3B overexpression largely counteracted miR-221-5p accumulation-mediated effects in ox-LDL-induced THP-1 cells. NF-κB signaling pathway was regulated by circSCAP/miR-221-5p/PDE3B axis in ox-LDL-induced THP-1 cells. In conclusion, circSCAP facilitated lipid accumulation, inflammation, and oxidative stress in ox-LDL-induced THP-1 macrophages by regulating miR-221-5p/PDE3B axis.


Sujet(s)
Athérosclérose/enzymologie , Cyclic Nucleotide Phosphodiesterases, Type 3/biosynthèse , Lipoprotéines LDL/toxicité , Macrophages/effets des médicaments et des substances chimiques , microARN/métabolisme , ARN circulaire/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Études cas-témoins , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cytokines/métabolisme , Induction enzymatique , Femelle , Humains , Médiateurs de l'inflammation/métabolisme , Macrophages/enzymologie , Macrophages/anatomopathologie , Mâle , microARN/génétique , Adulte d'âge moyen , Stress oxydatif/effets des médicaments et des substances chimiques , ARN circulaire/génétique , Transduction du signal , Cellules THP-1
15.
Elife ; 102021 05 27.
Article de Anglais | MEDLINE | ID: mdl-34042046

RÉSUMÉ

Sphingolipids are important structural components of cell membranes and prominent signaling molecules controlling cell growth, differentiation, and apoptosis. Sphingolipids are particularly abundant in the brain, and defects in sphingolipid degradation are associated with several human neurodegenerative diseases. However, molecular mechanisms governing sphingolipid metabolism remain unclear. Here, we report that sphingolipid degradation is under transcriptional control of SIRT1, a highly conserved mammalian NAD+-dependent protein deacetylase, in mouse embryonic stem cells (mESCs). Deletion of SIRT1 results in accumulation of sphingomyelin in mESCs, primarily due to reduction of SMPDL3B, a GPI-anchored plasma membrane bound sphingomyelin phosphodiesterase. Mechanistically, SIRT1 regulates transcription of Smpdl3b through c-Myc. Functionally, SIRT1 deficiency-induced accumulation of sphingomyelin increases membrane fluidity and impairs neural differentiation in vitro and in vivo. Our findings discover a key regulatory mechanism for sphingolipid homeostasis and neural differentiation, further imply that pharmacological manipulation of SIRT1-mediated sphingomyelin degradation might be beneficial for treatment of human neurological diseases.


All cells in the brain start life as stem cells which are yet to have a defined role in the body. A wide range of molecules and chemical signals guide stem cells towards a neuronal fate, including a group of molecules called sphingolipids. These molecules sit in the membrane surrounding the cell and play a pivotal role in a number of processes which help keep the neuronal cell healthy. Various enzymes work together to break down sphingolipids and remove them from the membrane. Defects in these enzymes can result in excess levels of sphingolipids, which can lead to neurodegenerative diseases, such as Alzheimer's, Parkinson's and Huntington's disease. But how these enzymes are used and controlled during neuronal development is still somewhat of a mystery. To help answer this question, Fan et al. studied an enzyme called SIRT1 which has been shown to alleviate symptoms in animal models of neurodegenerative diseases. Stem cells were extracted from a mouse embryo lacking the gene for SIRT1 and cultured in the laboratory. These faulty cells were found to have superfluous amounts of sphingolipids, which made their membranes more fluid and reduced their ability to develop into neuronal cells. Further investigation revealed that SIRT1 regulates the degradation of sphingolipids by promoting the production of another enzyme called SMPDL3B. Fan et al. also found that when female mice were fed a high-fat diet, this caused sphingolipids to accumulate in their embryos which lacked the gene for SIRT1; this, in turn, impaired the neural development of their offspring. These findings suggest that targeting SIRT1 may offer new strategies for treating neurological diseases. The discovery that embryos deficient in SIRT1 are sensitive to high-fat diets implies that activating this enzyme might attenuate some of the neonatal complications associated with maternal obesity.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Cellules souches embryonnaires de souris/enzymologie , Cellules souches neurales/enzymologie , Neurogenèse , Protéines proto-oncogènes c-myc/métabolisme , Sirtuine-1/métabolisme , Sphingolipides/métabolisme , Animaux , Lignée cellulaire , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Régulation de l'expression des gènes au cours du développement , Souris , Souris de lignée C57BL , Souris knockout , Protéines proto-oncogènes c-myc/génétique , Transduction du signal , Sirtuine-1/génétique , Transcription génétique
16.
Ann Hum Genet ; 85(2): 80-91, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33249558

RÉSUMÉ

Phosphodiesterase 3A (PDE3A) is an enzyme that plays an important role in the regulation of cyclic adenosine monophosphate (cAMP)-mediated intracellular signaling in cardiac myocytes and platelets. PDE3A hydrolyzes cAMP, which results in a decrease in intracellular cAMP levels and leads to platelet activation. Whole-exome sequencing of 50 DNA samples from a healthy Korean population revealed a total of 13 single nucleotide polymorphisms including five missense variants, D12N, Y497C, H504Q, C707R, and A980V. Recombinant proteins for the five variants of PDE3A (and wild-type protein) were expressed in a FreeStyle 293 expression system with site-directed mutagenesis. The expression of the recombinant PDE3A proteins was confirmed with Western blotting. Catalytic activity of the PDE3A missense variants and wild-type enzyme was measured with a PDE-based assay. Effects of the missense variants on the inhibition of PDE3A activity by cilostazol were also investigated. All variant proteins showed reduced activity (33-53%; p < .0001) compared to the wild-type protein. In addition, PDE3A activity was inhibited by cilostazol in a dose-dependent manner and was further suppressed in the missense variants. Specifically, the PDE3A Y497C showed significantly reduced activity, consistent with the predictions of in silico analyses. The present study provides evidence that individuals carrying the PDE3A Y497C variant may have lower enzyme activity for cAMP hydrolysis, which could cause interindividual variation in cAMP-mediated physiological functions.


Sujet(s)
Cilostazol/administration et posologie , AMP cyclique/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Activation plaquettaire/effets des médicaments et des substances chimiques , Adulte , Plaquettes/effets des médicaments et des substances chimiques , Cilostazol/effets indésirables , Relation dose-effet des médicaments , Femelle , Humains , Mâle , Mutation faux-sens/génétique , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Activation plaquettaire/génétique , Antiagrégants plaquettaires/administration et posologie , Antiagrégants plaquettaires/effets indésirables , Polymorphisme génétique/génétique , Transduction du signal/effets des médicaments et des substances chimiques ,
17.
Endocr Relat Cancer ; 28(1): 1-13, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-33112806

RÉSUMÉ

Familial primary aldosteronism (PA) is rare and mostly diagnosed in early-onset hypertension (HT). However, 'sporadic' bilateral adrenal hyperplasia (BAH) is the most frequent cause of PA and remains without genetic etiology in most cases. Our aim was to investigate new genetic defects associated with BAH and PA. We performed whole-exome sequencing (paired blood and adrenal tissue) in six patients with PA caused by BAH that underwent unilateral adrenalectomy. Additionally, we conducted functional studies in adrenal hyperplastic tissue and transfected cells to confirm the pathogenicity of the identified genetic variants. Rare germline variants in phosphodiesterase 2A (PDE2A) and 3B (PDE3B) genes were identified in three patients. The PDE2A heterozygous variant (p.Ile629Val) was identified in a patient with BAH and early-onset HT at 13 years of age. Two PDE3B heterozygous variants (p.Arg217Gln and p.Gly392Val) were identified in patients with BAH and HT diagnosed at 18 and 33 years of age, respectively. A strong PDE2A staining was found in all cases of BAH in zona glomerulosa and/or micronodules (that were also positive for CYP11B2). PKA activity in frozen tissue was significantly higher in BAH from patients harboring PDE2A and PDE3B variants. PDE2A and PDE3B variants significantly reduced protein expression in mutant transfected cells compared to WT. Interestingly, PDE2A and PDE3B variants increased SGK1 and SCNN1G/ENaCg at mRNA or protein levels. In conclusion, PDE2A and PDE3B variants were associated with PA caused by BAH. These novel genetic findings expand the spectrum of genetic etiologies of PA.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Hyperaldostéronisme/enzymologie , Adolescent , Adulte , Sujet âgé , Cyclic Nucleotide Phosphodiesterases, Type 2/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Femelle , Humains , Hyperaldostéronisme/génétique , Mâle , Adulte d'âge moyen
18.
Nutrients ; 12(11)2020 Oct 26.
Article de Anglais | MEDLINE | ID: mdl-33114701

RÉSUMÉ

Glaucoma, a leading cause of blindness, has multifactorial causes, including environmental and genetic factors. We evaluated genetic risk factors of glaucoma with gene-gene interaction and explored modifications of genetic risk with gene-lifestyles interaction in adults >40 years. The present study included 377 subjects with glaucoma and 47,820 subjects without glaucoma in a large-scale hospital-based cohort study from 2004 to 2013. The presence of glaucoma was evaluated by a diagnostic questionnaire evaluated by a doctor. The genome-wide association study was performed to identify genetic variants associated with glaucoma risk. Food intake was assessed using a semiquantitative food frequency questionnaire. We performed generalized multifactor dimensionality reduction analysis to construct polygenetic-risk score (PRS) and explored gene × nutrient interaction. PRS of the best model included LIM-domain binding protein-2 (LDB2) rs3763969, cyclin-dependent kinase inhibitor 2B (CDKN2B) rs523096, ABO rs2073823, phosphodiesterase-3A (PDE3A) rs12314390, and cadherin 13 (CDH13) rs12449180. Glaucoma risk in the high-PRS group was 3.02 times that in the low-PRS group after adjusting for confounding variables. For those with low serum glucose levels (<126 mg/dL), but not for those with high serum glucose levels, glaucoma risk in the high-PRS group was 3.16 times that in the low-PRS group. In those with high carbohydrate intakes (≥70%), but not in those with low carbohydrate intakes, glaucoma risk was 3.74 times higher in the high-PRS group than in the low-PRS group. The glaucoma risk was 3.87 times higher in the high-PRS group than in the low-PRS group only in a low balanced diet intake. In conclusion, glaucoma risk increased by three-fold in adults with a high PRS, and it can be reduced by good control of serum glucose concentrations and blood pressure (BP) with a balanced diet intake. These results can be applied to precision nutrition to reduce glaucoma risk.


Sujet(s)
Glycémie/analyse , Pression sanguine , Hydrates de carbone alimentaires/analyse , Prédisposition génétique à une maladie/étiologie , Glaucome/génétique , Système ABO de groupes sanguins/génétique , Adulte , Sujet âgé , Cadhérines/génétique , Études de cohortes , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Inhibiteur p15 de kinase cycline-dépendante/génétique , Régime alimentaire/effets indésirables , Enquêtes sur le régime alimentaire , Femelle , Galactosyltransferases/génétique , Étude d'association pangénomique , Régulation de la glycémie/statistiques et données numériques , Humains , Protéines à domaine LIM/génétique , Mâle , Adulte d'âge moyen , Phénomènes physiologiques nutritionnels/génétique , Polymorphisme de nucléotide simple , République de Corée , Appréciation des risques , Facteurs de risque , Facteurs de transcription/génétique
19.
BMC Med Genet ; 21(1): 144, 2020 07 06.
Article de Anglais | MEDLINE | ID: mdl-32631253

RÉSUMÉ

BACKGROUND: Autosomal dominant hypertension with brachydactyly type E syndrome caused by pathogenic variants in the PDE3A gene was first reported in 2015. To date, there are only a few reports of this kind of syndrome. Other patients still lack a genetic diagnosis. CASE PRESENTATION: Whole-exome sequencing was performed in an 18-year-old female proband with a clinical diagnosis of hypertension with brachydactyly syndrome. Quantitative real-time PCR was used to identify pathogenic copy number variations (CNVs). After bioinformatics analysis and healthy control database filtering, we revealed a heterozygous missense PDE3A variant (c.1346G > A, p.Gly449Asp). The variant was absent in the ExAC database and located in a highly evolutionarily conserved cluster of reported PDE3A pathogenic variants. Importantly, this variant was predicted to affect protein function by both SIFT (score = 0) and PolyPhen-2 (score = 1). After Sanger sequencing, the variant was determined to be absent in the healthy parents of the proband as well as 800 ethnically and geographically matched healthy controls. CONCLUSION: We present a report linking a de novo PDE3A variant to autosomal dominant hypertension with brachydactyly type E syndrome.


Sujet(s)
Brachydactylie/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , , Gènes dominants , Hypertension artérielle/congénital , Mutation/génétique , Adolescent , Brachydactylie/imagerie diagnostique , Exons/génétique , Femelle , Humains , Hypertension artérielle/imagerie diagnostique , Hypertension artérielle/génétique , Mâle , Pedigree , Syndrome
20.
Circulation ; 142(2): 133-149, 2020 07 14.
Article de Anglais | MEDLINE | ID: mdl-32524868

RÉSUMÉ

BACKGROUND: High blood pressure is the primary risk factor for cardiovascular death worldwide. Autosomal dominant hypertension with brachydactyly clinically resembles salt-resistant essential hypertension and causes death by stroke before 50 years of age. We recently implicated the gene encoding phosphodiesterase 3A (PDE3A); however, in vivo modeling of the genetic defect and thus showing an involvement of mutant PDE3A is lacking. METHODS: We used genetic mapping, sequencing, transgenic technology, CRISPR-Cas9 gene editing, immunoblotting, and fluorescence resonance energy transfer. We identified new patients, performed extensive animal phenotyping, and explored new signaling pathways. RESULTS: We describe a novel mutation within a 15 base pair (bp) region of the PDE3A gene and define this segment as a mutational hotspot in hypertension with brachydactyly. The mutations cause an increase in enzyme activity. A CRISPR/Cas9-generated rat model, with a 9-bp deletion within the hotspot analogous to a human deletion, recapitulates hypertension with brachydactyly. In mice, mutant transgenic PDE3A overexpression in smooth muscle cells confirmed that mutant PDE3A causes hypertension. The mutant PDE3A enzymes display consistent changes in their phosphorylation and an increased interaction with the 14-3-3θ adaptor protein. This aberrant signaling is associated with an increase in vascular smooth muscle cell proliferation and changes in vessel morphology and function. CONCLUSIONS: The mutated PDE3A gene drives mechanisms that increase peripheral vascular resistance causing hypertension. We present 2 new animal models that will serve to elucidate the underlying mechanisms further. Our findings could facilitate the search for new antihypertensive treatments.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Études d'associations génétiques , Prédisposition génétique à une maladie , Hypertension artérielle/génétique , Mutation , Allèles , Substitution d'acide aminé , Animaux , Animal génétiquement modifié , Pression artérielle , Marqueurs biologiques/sang , Marqueurs biologiques/urine , Brachydactylie/diagnostic , Brachydactylie/génétique , Systèmes CRISPR-Cas , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Analyse de mutations d'ADN , Modèles animaux de maladie humaine , Activation enzymatique , Ciblage de gène , Études d'associations génétiques/méthodes , Génotype , Immunohistochimie , Isoenzymes , Mâle , Pedigree , Phénotype , Radiographie , Rats , Système rénine-angiotensine/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...