Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 280
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.
Cells ; 12(11)2023 06 04.
Article de Anglais | MEDLINE | ID: mdl-37296663

RÉSUMÉ

Cyclic nucleotide phosphodiesterases 2A (PDE2A) and PDE3A play an important role in the regulation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP)-to-cAMP crosstalk. Each of these PDEs has up to three distinct isoforms. However, their specific contributions to cAMP dynamics are difficult to explore because it has been challenging to generate isoform-specific knock-out mice or cells using conventional methods. Here, we studied whether the CRISPR/Cas9 approach for precise genome editing can be used to knock out Pde2a and Pde3a genes and their distinct isoforms using adenoviral gene transfer in neonatal and adult rat cardiomyocytes. Cas9 and several specific gRNA constructs were cloned and introduced into adenoviral vectors. Primary adult and neonatal rat ventricular cardiomyocytes were transduced with different amounts of Cas9 adenovirus in combination with PDE2A or PDE3A gRNA constructs and cultured for up to 6 (adult) or 14 (neonatal) days to analyze PDE expression and live cell cAMP dynamics. A decline in mRNA expression for PDE2A (~80%) and PDE3A (~45%) was detected as soon as 3 days post transduction, with both PDEs being reduced at the protein level by >50-60% in neonatal cardiomyocytes (after 14 days) and >95% in adult cardiomyocytes (after 6 days). This correlated with the abrogated effects of selective PDE inhibitors in the live cell imaging experiments based on using cAMP biosensor measurements. Reverse transcription PCR analysis revealed that only the PDE2A2 isoform was expressed in neonatal myocytes, while adult cardiomyocytes expressed all three PDE2A isoforms (A1, A2, and A3) which contributed to the regulation of cAMP dynamics as detected by live cell imaging. In conclusion, CRISPR/Cas9 is an effective tool for the in vitro knock-out of PDEs and their specific isoforms in primary somatic cells. This novel approach suggests distinct regulation of live cell cAMP dynamics by various PDE2A and PDE3A isoforms in neonatal vs. adult cardiomyocytes.


Sujet(s)
Systèmes CRISPR-Cas , Cyclic Nucleotide Phosphodiesterases, Type 2 , Cyclic Nucleotide Phosphodiesterases, Type 3 , Myocytes cardiaques , Animaux , Souris , Rats , Systèmes CRISPR-Cas/génétique , AMP cyclique/métabolisme , Diéthylstilbestrol , Myocytes cardiaques/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 2/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Isoformes de protéines/métabolisme
5.
Commun Biol ; 6(1): 504, 2023 05 10.
Article de Anglais | MEDLINE | ID: mdl-37165086

RÉSUMÉ

The occurrence of NO/cGMP signalling in cardiac cells is a matter of debate. Recent measurements with a FRET-based cGMP indicator in isolated cardiac cells revealed NO-induced cGMP signals in cardiac fibroblasts while cardiomyocytes were devoid of these signals. In a fibroblast/myocyte co-culture model though, cGMP formed in fibroblasts in response to NO entered cardiomyocytes via gap junctions. Here, we demonstrate gap junction-mediated cGMP transfer from cardiac fibroblasts to myocytes in intact tissue. In living cardiac slices of mice with cardiomyocyte-specific expression of a FRET-based cGMP indicator (αMHC/cGi-500), NO-dependent cGMP signals were shown to occur in myocytes, to depend on gap junctions and to be degraded mainly by PDE3. Stimulation of NO-sensitive guanylyl cyclase enhanced Forskolin- and Isoproterenol-induced cAMP and phospholamban phosphorylation. Genetic inactivation of NO-GC in Tcf21-expressing cardiac fibroblasts abrogated the synergistic action of NO-GC stimulation on Iso-induced phospholamban phosphorylation, identifying fibroblasts as cGMP source and substantiating the necessity of cGMP-transfer to myocytes. In sum, NO-stimulated cGMP formed in cardiac fibroblasts enters cardiomyocytes in native tissue where it exerts an inhibitory effect on cAMP degradation by PDE3, thereby increasing cAMP and downstream effects in cardiomyocytes. Hence, enhancing ß-receptor-induced contractile responses appears as one of NO/cGMP's functions in the non-failing heart.


Sujet(s)
Coeur , Cellules cultivées , Animaux , Souris , Fibroblastes/métabolisme , GMP cyclique/métabolisme , Monoxyde d'azote/métabolisme , Cellules musculaires/métabolisme , AMP cyclique/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Transduction du signal , Survie cellulaire
6.
Int Immunopharmacol ; 119: 110157, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37086679

RÉSUMÉ

Synovial samples collected from 30 rheumatoid arthritis (RA) patients and 30 normal controls were used to isolate fibroblast-like synoviocytes (FLSs) and named FLS-RA and FLS-Normal, respectively. Real-time quantitative polymerase chain reaction (RT-qPCR) was utilized to detect circ_0007707 expression. Effects of circ_0007707 silencing on cell proliferation and apoptosis were evaluated using cell counting kit-8, 5-ethynyl-2'-deoxyuridine (Edu), and flow cytometry assays. Levels of pro-inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA). Increased circ_0007707 expression was observed in synovial samples from RA patients and FLS-RA cells. Functional analysis showed circ_0007707 silencing restrained cell proliferation, induced cell apoptosis, and decreased cell inflammatory response in FLS-RA cells. Mechanistic analysis revealed the sponge function of circ_0007707 on miR-27b-3p, and miR-27b-3p inhibition weakened circ_0007707 knockdown-mediated effects on FLS-RA cell proliferation, apoptosis, and inflammatory response. Circ_0007707 could mediate PDE3B expression via sponging miR-27b-3p, and PDE3B overturned miR-27b-3p mimic-mediated effects on FLS-RA cell proliferation, apoptosis, and inflammatory response. Circ_0007707 mediated cell apoptosis and inflammatory response in FLS-RA cells through the miR-27b-3p/PDE3B axis, indicating the potential function of circ_0007707 as a target for RA treatment.


Sujet(s)
Polyarthrite rhumatoïde , microARN , Cellules synoviales , Humains , Cellules synoviales/métabolisme , microARN/génétique , microARN/métabolisme , Inflammation/métabolisme , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Fibroblastes/métabolisme , Prolifération cellulaire/physiologie , Apoptose/génétique , Cellules cultivées , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme
7.
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
8.
Naunyn Schmiedebergs Arch Pharmacol ; 396(4): 669-682, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-36445386

RÉSUMÉ

Levosimendan (up to 10 µM) given alone failed to increase force of contraction in isolated electrically stimulated (1 Hz) left atrial (LA) preparations from wild-type mice. Only in the additional presence of 0.1 µM rolipram, an inhibitor of the activity of phosphodiesterase IV, levosimendan increased force of contraction in LA and increased the phosphorylation state of phospholamban at amino acid serine 16. Levosimendan alone increased the beating rate in isolated spontaneously beating right atrial preparations from mice and this effect was potentiated by rolipram. The positive inotropic and the positive chronotropic effects of levosimendan in mouse atrial preparations were attenuated by 10 µM propranolol. Finally, we studied the contractile effects of levosimendan in isolated electrically stimulated (1 Hz) right atrial preparations from the human atrium (HAP), obtained during cardiac surgery. We detected concentration-dependent positive inotropic effects of levosimendan alone that reached plateau at 1 µM levosimendan in HAP (n = 11). Levosimendan shortened time of tension relaxation in HAP. Cilostamide (1 µM), an inhibitor of phosphodiesterase III, or propranolol (10 µM) blocked the positive inotropic effect of levosimendan in HAP. Levosimendan (1 µM) alone increased in HAP the phosphorylation state of phospholamban. In conclusion, we present evidence that levosimendan acts via phosphodiesterase III inhibition in the human atrium leading to phospholamban phosphorylation and thus explaining the positive inotropic effects of levosimendan in HAP.


Sujet(s)
Fibrillation auriculaire , Propranolol , Humains , Souris , Animaux , Simendan/pharmacologie , Rolipram/pharmacologie , Phosphorylation , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Propranolol/pharmacologie , Contraction myocardique , Cardiotoniques/pharmacologie
9.
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
10.
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
11.
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
12.
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
13.
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
14.
Int J Mol Sci ; 24(1)2022 Dec 24.
Article de Anglais | MEDLINE | ID: mdl-36613764

RÉSUMÉ

As a worldwide health issue, obesity is associated with the infiltration of monocytes/macrophages into the adipose tissue causing unresolved inflammation. Monocyte chemoattractant protein-1 (MCP-1) exerts a crucial effect on obesity-related monocytes/macrophages infiltration. Clinically, aspirin and salsalate are beneficial for the treatment of metabolic diseases in which adipose tissue inflammation plays an essential role. Herein, we investigated the effect and precise mechanism of their active metabolite salicylate on TNF-α-elevated MCP-1 in adipocytes. The results indicated that salicylate sodium (SAS) could lower the level of MCP-1 in TNF-α-stimulated adipocytes, which resulted from a previously unrecognized target phosphodiesterase (PDE), 3B (PDE3B), rather than its known targets IKKß and AMPK. The SAS directly bound to the PDE3B to inactivate it, thus elevating the intracellular cAMP level and activating PKA. Subsequently, the expression of MKP-1 was increased, which led to the decrease in p-EKR and p-p38. Both PDE3B silencing and the pharmacological inhibition of cAMP/PKA compromised the suppressive effect of SAS on MCP-1. In addition to PDE3B, the PDE3A and PDE4B activity was also inhibited by SAS. Our findings identify a previously unrecognized pathway through which SAS is capable of attenuating the inflammation of adipocytes.


Sujet(s)
Chimiokine CCL2 , Facteur de nécrose tumorale alpha , Humains , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Chimiokine CCL2/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Adipocytes/métabolisme , Inflammation/métabolisme , Obésité/métabolisme , Salicylates/pharmacologie
15.
PLoS One ; 16(12): e0261681, 2021.
Article de Anglais | MEDLINE | ID: mdl-34972146

RÉSUMÉ

Lipolysis and the release of fatty acids to supply energy fuel to other organs, such as between meals, during exercise, and starvation, are fundamental functions of the adipose tissue. The intracellular lipolytic pathway in adipocytes is activated by adrenaline and noradrenaline, and inhibited by insulin. Circulating fatty acids are elevated in type 2 diabetic individuals. The mechanisms behind this elevation are not fully known, and to increase the knowledge a link between the systemic circulation and intracellular lipolysis is key. However, data on lipolysis and knowledge from in vitro systems have not been linked to corresponding in vivo data and knowledge in vivo. Here, we use mathematical modelling to provide such a link. We examine mechanisms of insulin action by combining in vivo and in vitro data into an integrated mathematical model that can explain all data. Furthermore, the model can describe independent data not used for training the model. We show the usefulness of the model by simulating new and more challenging experimental setups in silico, e.g. the extracellular concentration of fatty acids during an insulin clamp, and the difference in such simulations between individuals with and without type 2 diabetes. Our work provides a new platform for model-based analysis of adipose tissue lipolysis, under both non-diabetic and type 2 diabetic conditions.


Sujet(s)
Adipocytes/métabolisme , Tissu adipeux/métabolisme , Acides gras/métabolisme , Lipolyse/physiologie , Biologie des systèmes , Simulation numérique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Diabète de type 2/sang , Acides gras/sang , Humains , Techniques in vitro , Insuline/métabolisme , Insulinorésistance , Modèles statistiques , Modèles théoriques , Phosphorylation , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs alpha-2 adrénergiques/métabolisme , Récepteurs bêta-adrénergiques/métabolisme , Transduction du signal , Logiciel , Triglycéride/métabolisme , Incertitude
16.
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
17.
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
18.
Blood ; 137(5): 678-689, 2021 02 04.
Article de Anglais | MEDLINE | ID: mdl-33538796

RÉSUMÉ

Thrombospondin-1 (TSP-1) is released by platelets upon activation and can increase platelet activation, but its role in hemostasis in vivo is unclear. We show that TSP-1 is a critical mediator of hemostasis that promotes platelet activation by modulating inhibitory cyclic adenosine monophosphate (cAMP) signaling. Genetic deletion of TSP-1 did not affect platelet activation in vitro, but in vivo models of hemostasis and thrombosis showed that TSP-1-deficient mice had prolonged bleeding, defective thrombosis, and increased sensitivity to the prostacyclin mimetic iloprost. Adoptive transfer of wild-type (WT) but not TSP-1-/- platelets ameliorated the thrombotic phenotype, suggesting a key role for platelet-derived TSP-1. In functional assays, TSP-1-deficient platelets showed an increased sensitivity to cAMP signaling, inhibition of platelet aggregation, and arrest under flow by prostacyclin (PGI2). Plasma swap experiments showed that plasma TSP-1 did not correct PGI2 hypersensitivity in TSP-1-/- platelets. By contrast, incubation of TSP-1-/- platelets with releasates from WT platelets or purified TSP-1, but not releasates from TSP-1-/- platelets, reduced the inhibitory effects of PGI2. Activation of WT platelets resulted in diminished cAMP accumulation and downstream signaling, which was associated with increased activity of the cAMP hydrolyzing enzyme phosphodiesterase 3A (PDE3A). PDE3A activity and cAMP accumulation were unaffected in platelets from TSP-1-/- mice. Platelets deficient in CD36, a TSP-1 receptor, showed increased sensitivity to PGI2/cAMP signaling and diminished PDE3A activity, which was unaffected by platelet-derived or purified TSP-1. This scenario suggests that the release of TSP-1 regulates hemostasis in vivo through modulation of platelet cAMP signaling at sites of vascular injury.


Sujet(s)
Plaquettes/physiologie , AMP cyclique/physiologie , Troubles hémorragiques/génétique , Hémostase/physiologie , Thrombospondine-1/physiologie , Animaux , Temps de saignement , Plaquettes/effets des médicaments et des substances chimiques , Antigènes CD36/déficit , Antigènes CD36/physiologie , Cellules cultivées , Chlorures/toxicité , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Granulations cytoplasmiques/métabolisme , Prostacycline/physiologie , Composés du fer III/toxicité , Humains , Iloprost/pharmacologie , Souris , Souris de lignée C57BL , Transfusion de plaquettes , Systèmes de seconds messagers/physiologie , Thrombose/induit chimiquement , Thrombose/prévention et contrôle , Thrombospondine-1/déficit , Thrombospondine-1/pharmacologie
19.
Naunyn Schmiedebergs Arch Pharmacol ; 394(6): 1215-1229, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33576869

RÉSUMÉ

Histamine exerts cAMP-dependent positive inotropic effects (PIE) and positive chronotropic effects (PCE) on isolated left and right atria, respectively, of transgenic mice which overexpress the human H2-receptor in the heart (=H2-TG). To determine whether these effects are antagonized by phosphodiesterases (PDEs), contractile studies were done in isolated left and right atrial preparations of H2-TG. The contractile effects of histamine were tested in the additional presence of the PDE-inhibitorserythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride (EHNA, 1 µM, PDE2-inhibitor) or cilostamide (1 µM, PDE3-inhibitor), rolipram (10 µM, a PDE4-inhibitor), and their combinations. Cilostamide (1 µM) and EHNA (1 µM), rolipram (1 µM), and EHNA (1 µM) and the combination of rolipram (0.1 µM) and cilostamide (1 µM) each increased the potency of histamine to elevate the force of contraction (FOC) in H2-TG. Cilostamide (1 µM) and rolipram (10 µM) alone increased and EHNA (1 µM) decreased alone, and their combination increased the potency of histamine to increase the FOC in H2-TG indicating that PDE3 and PDE4 regulate the inotropic effects of histamine in H2-TG. The PDE inhibitors (EHNA, cilostamide, rolipram) alone did not alter the potency of histamine to increase the heart beat in H2-TG whereas a combination of rolipram, cilostamide, and EHNA, or of rolipram and EHNA increased the potency of histamine to act on the beating rate. In summary, the data suggest that the PCE of histamine in H2-TG atrium involves PDE 2 and 4 activities, whereas the PIE of histamine are diminished by activity of PDE 3 and 4.


Sujet(s)
Atrium du coeur/métabolisme , Histamine/métabolisme , Récepteur histaminergique H2/métabolisme , Adénine/administration et posologie , Adénine/analogues et dérivés , Adénine/pharmacologie , Animaux , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonistes et inhibiteurs , Cyclic Nucleotide Phosphodiesterases, Type 2/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/effets des médicaments et des substances chimiques , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 4/effets des médicaments et des substances chimiques , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Femelle , Atrium du coeur/effets des médicaments et des substances chimiques , Rythme cardiaque/effets des médicaments et des substances chimiques , Rythme cardiaque/physiologie , Humains , Mâle , Souris , Souris transgéniques , Inhibiteurs de la phosphodiestérase/administration et posologie , Inhibiteurs de la phosphodiestérase/pharmacologie , Quinolinone/administration et posologie , Quinolinone/pharmacologie , Rolipram/administration et posologie , Rolipram/pharmacologie
20.
Naunyn Schmiedebergs Arch Pharmacol ; 394(2): 291-298, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32949251

RÉSUMÉ

Atrial fibrillation (AF)-associated remodeling includes contractile dysfunction whose reasons are only partially resolved. Serotonin (5-HT) increases contractile force and causes arrhythmias in atrial trabeculae from patients in sinus rhythm (SR). In persistent atrial fibrillation (peAF), the force responses to 5-HT are blunted and arrhythmic effects are abolished. Since force but not arrhythmic responses to 5-HT in peAF could be restored by PDE3 + PDE4 inhibition, we sought to perform real-time measurements of cAMP to understand whether peAF alters PDE3 + PDE4-mediated compartmentation of 5-HT4 receptor-cAMP responses. Isolated human atrial myocytes from patients in SR, with paroxysmal AF (paAF) or peAF, were adenovirally transduced to express the FRET-based cAMP sensor Epac1-camps. Forty-eight hours later, cAMP responses to 5-HT (100 µM) were measured in the absence or concomitant presence of the PDE3 inhibitor cilostamide (0.3 µM) and the PDE4 inhibitor rolipram (1 µM). We successfully established real-time cAMP imaging in AF myocytes. 5-HT increased cAMP in SR, paAF, and peAF, but in line with previous findings on contractility, this increase was considerably smaller in peAF than in SR or paAF. The maximal cAMP response to forskolin (10 µM) was preserved in all groups. The diminished cAMP response to 5-HT in peAF was recovered by preincubation with cilostamide + rolipram. We uncovered a significantly diminished cAMP response to 5-HT4 receptor stimulation which may explain the blunted 5-HT inotropic responses observed in peAF. Since both cAMP and force responses but not arrhythmic responses were recovered after concomitant inhibition of PDE3 + PDE4, they might be regulated in different subcellular microdomains.


Sujet(s)
Fibrillation auriculaire/métabolisme , AMP cyclique/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Cyclic Nucleotide Phosphodiesterases, Type 4/métabolisme , Myocytes cardiaques/métabolisme , Récepteurs de la sérotonine de type 5-HT4/métabolisme , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...