Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.907
Filtrer
1.
Med Sci (Paris) ; 40(6-7): 534-543, 2024.
Article de Français | MEDLINE | ID: mdl-38986098

RÉSUMÉ

Cyclic nucleotide phosphodiesterases (PDEs) modulate neurohormonal regulation of cardiac function by degrading cAMP and cGMP. In cardiomyocytes, multiple isoforms of PDEs with different enzymatic properties and subcellular locally regulate cyclic nucleotide levels and associated cellular functions. This organisation is severely disrupted during hypertrophy and heart failure (HF), which may contribute to disease progression. Clinically, PDE inhibition has been seen as a promising approach to compensate for the catecholamine desensitisation that accompanies heart failure. Although PDE3 inhibitors such as milrinone or enoximone can be used clinically to improve systolic function and relieve the symptoms of acute CHF, their chronic use has proved detrimental. Other PDEs, such as PDE1, PDE2, PDE4, PDE5, PDE9 and PDE10, have emerged as potential new targets for the treatment of HF, each with a unique role in local cyclic nucleotide signalling pathways. In this review, we describe cAMP and cGMP signalling in cardiomyocytes and present the different families of PDEs expressed in the heart and their modifications in pathological cardiac hypertrophy and HF. We also review results from preclinical models and clinical data indicating the use of specific PDE inhibitors or activators that may have therapeutic potential in CI.


Title: Les phosphodiestérases des nucléotides cycliques - Cibles thérapeutiques dans l'hypertrophie et l'insuffisance cardiaques. Abstract: Les phosphodiestérases des nucléotides cycliques (PDE) modulent la régulation neuro-hormonale de la fonction cardiaque en dégradant l'AMPc et le GMPc. Dans les cardiomyocytes, de multiples isoformes de PDE, aux propriétés enzymatiques et aux localisations subcellulaires différentes, régulent localement les niveaux de nucléotides cycliques et les fonctions cellulaires associées. Cette organisation est fortement perturbée au cours de l'hypertrophie et de l'insuffisance cardiaque à fraction d'éjection réduite (IC), ce qui peut contribuer à la progression de la maladie. Sur le plan clinique, l'inhibition des PDE a été considérée comme une approche prometteuse pour compenser la désensibilisation aux catécholamines qui accompagne l'IC. Bien que des inhibiteurs de la PDE3, tels que la milrinone ou l'énoximone, puissent être utilisés cliniquement pour améliorer la fonction systolique et soulager les symptômes de l'IC aiguë, leur utilisation chronique s'est avérée préjudiciable. D'autres PDE, telles que les PDE1, PDE2, PDE4, PDE5, PDE9 et PDE10, sont apparues comme de nouvelles cibles potentielles pour le traitement de l'IC, chacune ayant un rôle unique dans les voies de signalisation locales des nucléotides cycliques. Dans cette revue, nous décrivons la signalisation de l'AMPc et du GMPc dans les cardiomyocytes et présentons les différentes familles de PDE exprimées dans le cœur ainsi que leurs modifications dans l'hypertrophie cardiaque pathologique et dans l'IC. Nous évaluons également les résultats issus de modèles précliniques ainsi que les données cliniques indiquant l'utilisation d'inhibiteurs ou d'activateurs de PDE spécifiques qui pourraient avoir un potentiel thérapeutique dans l'IC.


Sujet(s)
Cardiomégalie , Défaillance cardiaque , Inhibiteurs de la phosphodiestérase , Humains , Cardiomégalie/traitement médicamenteux , Défaillance cardiaque/traitement médicamenteux , Animaux , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Inhibiteurs de la phosphodiestérase/pharmacologie , 3',5'-Cyclic-AMP Phosphodiesterases/antagonistes et inhibiteurs , 3',5'-Cyclic-AMP Phosphodiesterases/métabolisme , 3',5'-Cyclic-AMP Phosphodiesterases/physiologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Thérapie moléculaire ciblée/méthodes , GMP cyclique/métabolisme , GMP cyclique/physiologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , AMP cyclique/métabolisme , AMP cyclique/physiologie , Phosphodiesterases/métabolisme , Phosphodiesterases/physiologie
2.
Eur Respir Rev ; 33(172)2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-39009409

RÉSUMÉ

Lysophosphatidic acid (LPA)-mediated activation of LPA receptor 1 (LPAR1) contributes to the pathophysiology of fibrotic diseases such as idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc). These diseases are associated with high morbidity and mortality despite current treatment options. The LPA-producing enzyme autotaxin (ATX) and LPAR1 activation contribute to inflammation and mechanisms underlying fibrosis in preclinical fibrotic models. Additionally, elevated levels of LPA have been detected in bronchoalveolar lavage fluid from patients with IPF and in serum from patients with SSc. Thus, ATX and LPAR1 have gained considerable interest as pharmaceutical targets to combat fibrotic disease and inhibitors of these targets have been investigated in clinical trials for IPF and SSc. The goals of this review are to summarise the current literature on ATX and LPAR1 signalling in pulmonary fibrosis and to help differentiate the novel inhibitors in development. The mechanisms of action of ATX and LPAR1 inhibitors are described and preclinical studies and clinical trials of these agents are outlined. Because of their contribution to numerous physiologic events underlying fibrotic disease, ATX and LPAR1 inhibition presents a promising therapeutic strategy for IPF, SSc and other fibrotic diseases that may fulfil unmet needs of the current standard of care.


Sujet(s)
Fibrose pulmonaire idiopathique , Phosphodiesterases , Récepteurs à l'acide phosphatidique , Transduction du signal , Humains , Récepteurs à l'acide phosphatidique/antagonistes et inhibiteurs , Récepteurs à l'acide phosphatidique/métabolisme , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Phosphodiesterases/métabolisme , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Thérapie moléculaire ciblée , Poumon/effets des médicaments et des substances chimiques , Poumon/physiopathologie , Poumon/métabolisme , Antifibrotiques/usage thérapeutique , Lysophospholipides/métabolisme , Résultat thérapeutique , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/physiopathologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique
4.
Drug Res (Stuttg) ; 74(5): 241-249, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38830372

RÉSUMÉ

Pentoxifylline (PTX), a non-selective phosphodiesterase inhibitor, has demonstrated protective effects against lung injury in animal models. Given the significance of pulmonary toxicity resulting from paraquat (PQ) exposure, the present investigation was designed to explore the impact of PTX on PQ-induced pulmonary oxidative impairment in male mice.Following preliminary studies, thirty-six mice were divided into six groups. Group 1 received normal saline, group 2 received a single dose of PQ (20 mg/kg; i.p.), and group 3 received PTX (100 mg/kg/day; i.p.). Additionally, treatment groups 4-6 were received various doses of PTX (25, 50, and 100 mg/kg/day; respectively) one hour after a single dose of PQ. After 72 hours, the animals were sacrificed, and lung tissue was collected.PQ administration caused a significant decrease in hematocrit and an increase in blood potassium levels. Moreover, a notable increase was found in the lipid peroxidation (LPO), nitric oxide (NO), and myeloperoxidase (MPO) levels, along with a notable decrease in total thiol (TTM) and total antioxidant capacity (TAC) contents, catalase (CAT) and superoxide dismutase (SOD) enzymes activity in lung tissue. PTX demonstrated the ability to improve hematocrit levels; enhance SOD activity and TTM content; and decrease MPO activity, LPO and NO levels in PQ-induced pulmonary toxicity. Furthermore, these findings were well-correlated with the observed lung histopathological changes.In conclusion, our results suggest that the high dose of PTX may ameliorate lung injury by improving the oxidant/antioxidant balance in animals exposed to PQ.


Sujet(s)
Antioxydants , Peroxydation lipidique , Poumon , Paraquat , Pentoxifylline , Superoxide dismutase , Animaux , Pentoxifylline/pharmacologie , Pentoxifylline/usage thérapeutique , Paraquat/toxicité , Souris , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Superoxide dismutase/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Catalase/métabolisme , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Monoxyde d'azote/métabolisme , Myeloperoxidase/métabolisme , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/traitement médicamenteux , Phosphodiesterases/métabolisme
5.
Pharmacopsychiatry ; 57(4): 205-214, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38710206

RÉSUMÉ

BACKGROUND: Evidence indicates an association between immune dysregulation and major depressive disorder (MDD). Pentoxifylline (PTX), a phosphodiesterase inhibitor, has been shown to reduce pro-inflammatory activities. The aim of this study was to evaluate changes in depressive symptoms and pro-inflammatory markers after administration of PTX as an adjunctive agent to citalopram in patients with MDD. METHODS: One hundred patients were randomly assigned to either citalopram (20 mg/day) plus placebo (twice daily) (n=50) or citalopram (20 mg/day) plus PTX (400 mg) (twice daily) (n=50). The Hamilton Depression Rating Scale-17 (HAM-D-17) scores at baseline, weeks 2, 4, 6, 8, 10, and 12 and serum levels of interleukin1-ß (IL-1-ß), tumor necrosis factor-α, C-reactive protein, IL-6, serotonin, IL-10, and brain-derived neurotrophic factor (BDNF) at baseline and week 12 were evaluated. RESULTS: HAM-D-17 score in the PTX group significantly reduced in comparison to the control group after weeks 4, 6, 8,10, and 12 ((LSMD): - 2.193, p=0.021; - 2.597, p=0.036; - 2.916, p=0.019; - 4.336, p=0.005; and - 4.087, p=0.008, respectively). Patients who received PTX had a better response (83%) and remission rate (79%) compared to the placebo group (49% and 40%, p=0.006 and p=0.01, respectively). Moreover, the reduction in serum concentrations of pro-inflammatory factors and increase in serotonin and BDNF in the PTX group was significantly greater than in the placebo group (p<0.001). CONCLUSION: These findings support the safety and efficacy of PTX as an adjunctive antidepressant agent with anti-inflammatory effects in patients with MDD.


Sujet(s)
Citalopram , Trouble dépressif majeur , Association de médicaments , Pentoxifylline , Humains , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/sang , Pentoxifylline/usage thérapeutique , Pentoxifylline/administration et posologie , Mâle , Femelle , Méthode en double aveugle , Adulte , Citalopram/usage thérapeutique , Citalopram/administration et posologie , Adulte d'âge moyen , Résultat thérapeutique , Facteur neurotrophique dérivé du cerveau/sang , Échelles d'évaluation en psychiatrie , Protéine C-réactive/analyse , Jeune adulte , Sérotonine/sang , Antidépresseurs/usage thérapeutique , Antidépresseurs/administration et posologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique
8.
J Med Chem ; 67(10): 8309-8322, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38669059

RÉSUMÉ

Liver fibrosis is a common pathological feature of most chronic liver diseases with no effective drugs available. Phosphodiesterase 1 (PDE1), a subfamily of the PDE super enzyme, might work as a potent target for liver fibrosis by regulating the concentration of cAMP and cGMP. However, there are few PDE1 selective inhibitors, and none has been investigated for liver fibrosis treatment yet. Herein, compound AG-205/1186117 with the dihydropyrimidine scaffold was selected as the hit by virtual screening. A hit-to-lead structural modification led to a series of dihydropyrimidine derivatives. Lead 13h exhibited the IC50 of 10 nM against PDE1, high selectivity over other PDEs, as well as good safety properties. Administration of 13h exerted significant anti-liver fibrotic effects in bile duct ligation-induced fibrosis rats, which also prevented TGF-ß-induced myofibroblast differentiation in vitro, confirming that PDE1 could work as a potential target for liver fibrosis.


Sujet(s)
Cyclic Nucleotide Phosphodiesterases, Type 1 , Conception de médicament , Cirrhose du foie , Inhibiteurs de la phosphodiestérase , Pyrimidines , Animaux , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonistes et inhibiteurs , Cyclic Nucleotide Phosphodiesterases, Type 1/métabolisme , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/anatomopathologie , Pyrimidines/synthèse chimique , Pyrimidines/pharmacologie , Pyrimidines/composition chimique , Pyrimidines/usage thérapeutique , Humains , Rats , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/synthèse chimique , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Inhibiteurs de la phosphodiestérase/composition chimique , Mâle , Relation structure-activité , Rat Sprague-Dawley , Simulation de docking moléculaire , Structure moléculaire
9.
Eur J Med Chem ; 271: 116386, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38614063

RÉSUMÉ

Phosphodiesterase (PDE) is a superfamily of enzymes that are responsible for the hydrolysis of two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). PDE inhibition promotes the gene transcription by activating cAMP-response element binding protein (CREB), initiating gene transcription of brain-derived neurotrophic factor (BDNF). The procedure exerts neuroprotective profile, and motor and cognitive improving efficacy. From this point of view, PDE inhibition will provide a promising therapeutic strategy for treating neurodegenerative disorders. Herein, we summarized the PDE inhibitors that have entered the clinical trials or been discovered in recent five years. Well-designed clinical or preclinical investigations have confirmed the effectiveness of PDE inhibitors, such as decreasing Aß oligomerization and tau phosphorylation, alleviating neuro-inflammation and oxidative stress, modulating neuronal plasticity and improving long-term cognitive impairment.


Sujet(s)
Maladies neurodégénératives , Inhibiteurs de la phosphodiestérase , Humains , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/composition chimique , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Animaux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Phosphodiesterases/métabolisme , Structure moléculaire
10.
Front Immunol ; 15: 1365484, 2024.
Article de Anglais | MEDLINE | ID: mdl-38524120

RÉSUMÉ

T-cell activation is a pivotal process of the adaptive immune response with 3',5'-cyclic adenosine monophosphate (cAMP) as a key regulator of T-cell activation and function. It governs crucial control over T-cell differentiation and production of pro-inflammatory cytokines, such as IFN-γ. Intriguingly, levels of intracellular cAMP differ between regulatory (Treg) and conventional T-cells (Tcon). During cell-cell contact, cAMP is transferred via gap junctions between these T-cell subsets to mediate the immunosuppressive function of Treg. Moreover, the activation of T-cells via CD3 and CD28 co-stimulation leads to a transient upregulation of cAMP. Elevated intracellular cAMP levels are balanced precisely by phosphodiesterases (PDEs), a family of enzymes that hydrolyze cyclic nucleotides. Various PDEs play distinct roles in regulating cAMP and cyclic guanosine monophosphate (cGMP) in T-cells. Research on PDEs has gained growing interest due to their therapeutic potential to manipulate T-cell responses. So far, PDE4 is the best-described PDE in T-cells and the first PDE that is currently targeted in clinical practice to treat autoimmune diseases. But also, other PDE families harbor additional therapeutic potential. PDE2A is a dual-substrate phosphodiesterase which is selectively upregulated in Tcon upon activation. In this Mini-Review, we will highlight the impact of cAMP regulation on T-cell activation and function and summarize recent findings on different PDEs regulating intracellular cAMP levels in T-cells.


Sujet(s)
Diéthylstilbestrol/analogues et dérivés , Inhibiteurs de la phosphodiestérase , Phosphodiesterases , Inhibiteurs de la phosphodiestérase/usage thérapeutique , AMP cyclique , Lymphocytes T
11.
Cells ; 13(4)2024 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-38391934

RÉSUMÉ

Alcohol use disorder (AUD) requires new neurobiological targets. Problematic drinking involves underactive indirect pathway medium spiny neurons (iMSNs) that subserve adaptive behavioral selection vs. overactive direct pathway MSNs (dMSNs) that promote drinking, with a shift from ventromedial to dorsolateral striatal (VMS, DLS) control of EtOH-related behavior. We hypothesized that inhibiting phosphodiesterase 10A (PDE10A), enriched in striatal MSNs, would reduce EtOH self-administration in rats with a history of chronic intermittent ethanol exposure. To test this, Wistar rats (n = 10/sex) with a history of chronic intermittent EtOH (CIE) vapor exposure received MR1916 (i.p., 0, 0.05, 0.1, 0.2, and 0.4 µmol/kg), a PDE10A inhibitor, before operant EtOH self-administration sessions. We determined whether MR1916 altered the expression of MSN markers (Pde10a, Drd1, Drd2, Penk, and Tac1) and immediate-early genes (IEG) (Fos, Fosb, ΔFosb, and Egr1) in EtOH-naïve (n = 5-6/grp) and post-CIE (n = 6-8/grp) rats. MR1916 reduced the EtOH self-administration of high-drinking, post-CIE males, but increased it at a low, but not higher, doses, in females and low-drinking males. MR1916 increased Egr1, Fos, and FosB in the DLS, modulated by sex and alcohol history. MR1916 elicited dMSN vs. iMSN markers differently in ethanol-naïve vs. post-CIE rats. High-drinking, post-CIE males showed higher DLS Drd1 and VMS IEG expression. Our results implicate a role and potential striatal bases of PDE10A inhibitors to influence post-dependent drinking.


Sujet(s)
Éthanol , Composés chimiques organiques , Inhibiteurs de la phosphodiestérase , Mâle , Femelle , Rats , Animaux , Éthanol/pharmacologie , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Rat Wistar , Phosphodiesterases/génétique , Phosphodiesterases/métabolisme , Expression des gènes
12.
Med Chem ; 20(5): 467-486, 2024.
Article de Anglais | MEDLINE | ID: mdl-38265379

RÉSUMÉ

Intracellular glucose concentration plays a crucial role in initiating the molecular secretory process of pancreatic ß-cells through multiple messengers and signaling pathways. Cyclic nucleotides are key physiological regulators that modulate pathway interactions in ß -cells. An increase of cyclic nucleotides is controled by hydrolysed phosphodiesterases (PDEs), which degrades cyclic nucleotides into inactive metabolites. Despite the undeniable therapeutic potential of PDE inhibitors, they are associated with several side effects. The treatment strategy for diabetes based on PDE inhibitors has been proposed for a long time. Hence, the world of natural antidiabetic medicinal plants represents an ideal source of phosphodiesterase inhibitors as a new strategy for developing novel agents to treat diabetes mellitus. This review highlights medicinal plants traditionally used in the treatment of diabetes mellitus that have been proven to have inhibitory effects on PDE activity. The contents of this review were sourced from electronic databases, including Science Direct, PubMed, Springer Link, Web of Science, Scopus, Wiley Online, Scifinder and Google Scholar. These databases were consulted to collect information without any limitation date. After comprehensive literature screening, this paper identified 27 medicinal plants that have been reported to exhibit anti-phosphodiesterase activities. The selection of these plants was based on their traditional uses in the treatment of diabetes mellitus. The review emphasizes the antiphosphodiesterase properties of 31 bioactive components derived from these plant extracts. Many phenolic compounds have been identified as PDE inhibitors: Brazilin, mesozygin, artonin I, chalcomaracin, norartocarpetin, moracin L, moracin M, moracin C, curcumin, gallic acid, caffeic acid, rutin, quercitrin, quercetin, catechin, kaempferol, chlorogenic acid, and ellagic acid. Moreover, smome lignans have reported as PDE inhibitors: (+)-Medioresinol di-O-ß-d-glucopyranoside, (+)- Pinoresinol di-O-ß-d-glucopyranoside, (+)-Pinoresinol-4-O-ß-d-glucopyranosyl (1→6)-ß-dglucopyranoside, Liriodendrin, (+)-Pinoresinol 4'-O-ß-d-glucopyranoside, and forsythin. This review provides a promising starting point of medicinal plants, which could be further studied for the development of natural phosphodiesterase inhibitors to treat diabetes mellitus. Therefore, it is important to consider clinical studies for the identification of new targets for the treatment of diabetes.


Sujet(s)
Diabète , Hypoglycémiants , Inhibiteurs de la phosphodiestérase , Plantes médicinales , Plantes médicinales/composition chimique , Humains , Hypoglycémiants/pharmacologie , Hypoglycémiants/composition chimique , Hypoglycémiants/usage thérapeutique , Diabète/traitement médicamenteux , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/composition chimique , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Animaux , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/usage thérapeutique
13.
Med Res Rev ; 44(4): 1404-1445, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38279990

RÉSUMÉ

Neurodegenerative diseases (NDs) cause progressive loss of neuron structure and ultimately lead to neuronal cell death. Since the available drugs show only limited symptomatic relief, NDs are currently considered as incurable. This review will illustrate the principal roles of the signaling systems of cyclic adenosine and guanosine 3',5'-monophosphates (cAMP and cGMP) in the neuronal functions, and summarize expression/activity changes of the associated enzymes in the ND patients, including cyclases, protein kinases, and phosphodiesterases (PDEs). As the sole enzymes hydrolyzing cAMP and cGMP, PDEs are logical targets for modification of neurodegeneration. We will focus on PDE inhibitors and their potentials as disease-modifying therapeutics for the treatment of Alzheimer's disease, Parkinson's disease, and Huntington's disease. For the overlapped but distinct contributions of cAMP and cGMP to NDs, we hypothesize that dual PDE inhibitors, which simultaneously regulate both cAMP and cGMP signaling pathways, may have complementary and synergistic effects on modifying neurodegeneration and thus represent a new direction on the discovery of ND drugs.


Sujet(s)
Maladies neurodégénératives , Inhibiteurs de la phosphodiestérase , Humains , Maladies neurodégénératives/traitement médicamenteux , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Inhibiteurs de la phosphodiestérase/pharmacologie , Animaux , AMP cyclique/métabolisme , GMP cyclique/métabolisme , Phosphodiesterases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
14.
Physiol Rev ; 104(2): 765-834, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-37971403

RÉSUMÉ

Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.


Sujet(s)
Maladies cardiovasculaires , Diéthylstilbestrol/analogues et dérivés , Phosphodiesterases , Humains , Inhibiteurs de la phosphodiestérase/usage thérapeutique , AMP cyclique , GMP cyclique , Isoformes de protéines
15.
JACC Heart Fail ; 12(1): 100-113, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37921801

RÉSUMÉ

BACKGROUND: Intracellular second messenger cyclic guanosine monophosphate (cGMP) mediates bioactivity of the natriuretic peptides and nitric oxide, and is key to circulatory homeostasis and protection against cardiovascular disease. Inhibition of cGMP-degrading phosphodiesterases (PDEs) PDE5 and PDE9 are emerging as pharmacological targets in heart failure (HF). OBJECTIVES: The present study investigated dual enhancement of cGMP in experimental HF by combining inhibition of PDE-5 (P5-I) and PDE-9 (P9-I). METHODS: Eight sheep with pacing-induced HF received on separate days intravenous P5-I (sildenafil), P9-I (PF-04749982), P5-I+P9-I, and vehicle control, in counterbalanced order. RESULTS: Compared with control, separate P5-I and P9-I significantly increased circulating cGMP concentrations in association with reductions in mean arterial pressure (MAP), left atrial pressure (LAP), and pulmonary arterial pressure (PAP), with effects of P5-I on cGMP, MAP, and PAP greater than those of P9-I. Only P5-I decreased pulmonary vascular resistance. Combination P5-I+P9-I further reduced MAP, LAP, and PAP relative to inhibition of either phosphodiesterase alone. P9-I and, especially, P5-I elevated urinary cGMP levels relative to control. However, whereas inhibition of either enzyme increased urine creatinine excretion and clearance, only P9-I induced a significant diuresis and natriuresis. Combined P5-I+P9-I further elevated urine cGMP with concomitant increases in urine volume, sodium and creatinine excretion, and clearance similar to P9-I alone, despite the greater MAP reductions induced by combination treatment. CONCLUSIONS: Combined P5-I+P9-I amalgamated the superior renal effects of P9-I and pulmonary effects of P5-1, while concurrently further reducing cardiac preload and afterload. These findings support combination P5-I+P9-I as a therapeutic strategy in HF.


Sujet(s)
Défaillance cardiaque , Humains , Animaux , Ovis , Cyclic Nucleotide Phosphodiesterases, Type 5/usage thérapeutique , Défaillance cardiaque/traitement médicamenteux , Créatinine , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Inhibiteurs de la phosphodiestérase/pharmacologie , GMP cyclique
16.
Biochem Pharmacol ; 220: 116006, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38142838

RÉSUMÉ

Cancer immunotherapy, particularly with immune checkpoint inhibitors, has revolutionized the paradigm of cancer treatment. Nevertheless, the efficacy of cancer immunotherapy remains limited in most clinical settings due to the lack of a preexisting antitumor T-cell response in tumors. Therefore, the clinical outcomes of cancer immunotherapy must be improved crucially. With increased awareness of the importance of the innate immune response in the recruitment of T cells, as well as the onset and maintenance of the T cell response, great interest has been shown in activating the cGAS-STING signaling pathway to awaken the innate immune response, thereby orchestrating both innate and adaptive immune responses to induce tumor clearance. However, tumor cells have evolved to overexpress ectonucleotide pyrophosphate phosphodiesterase 1 (ENPP1), which degrades the immunotransmitter 2',3'-cGAMP and promotes the production of immune-suppressing adenosine, resulting in inhibition of the anticancer immune response in the tumor microenvironment. Clinically, ENPP1 overexpression is closely associated with poor prognosis in patients with cancer. Conversely, depleting or inhibiting ENPP1 has been verified to elevate extracellular 2',3'-cGAMP levels and inhibit the generation of adenosine, thereby reinvigorating the anticancer immune response for tumor elimination. A variety of ENPP1 inhibitors have recently been developed and have demonstrated significant promise for cancer immunotherapy. In this review, we provide an overview of ENPP1, dissect its immunosuppressive mechanisms, and discuss the development of ENPP1 inhibitors with the potential to further improve the efficacy of cancer immunotherapy.


Sujet(s)
Tumeurs , Phosphodiesterases , Humains , Adénosine , Diphosphates , Immunothérapie , Tumeurs/métabolisme , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Phosphodiesterases/métabolisme , Microenvironnement tumoral
17.
mBio ; 15(2): e0305623, 2024 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-38132724

RÉSUMÉ

Apicomplexa encompasses a large number of intracellular parasites infecting a wide range of animals. Cyclic nucleotide signaling is crucial for a variety of apicomplexan life stages and cellular processes. The cyclases and kinases that synthesize and respond to cyclic nucleotides (i.e., 3',5'-cyclic guanosine monophosphate and 3',5'-cyclic adenosine monophosphate) are highly conserved and essential throughout the parasite phylum. Growing evidence indicates that phosphodiesterases (PDEs) are also critical for regulating cyclic nucleotide signaling via cyclic nucleotide hydrolysis. Here, we discuss recent advances in apicomplexan PDE biology and opportunities for therapeutic interventions, with special emphasis on the major human apicomplexan parasite genera Plasmodium, Toxoplasma, Cryptosporidium, and Babesia. In particular, we show a highly flexible repertoire of apicomplexan PDEs associated with a wide range of cellular requirements across parasites and lifecycle stages. Despite this phylogenetic diversity, cellular requirements of apicomplexan PDEs for motility, host cell egress, or invasion are conserved. However, the molecular wiring of associated PDEs is extremely malleable suggesting that PDE diversity and redundancy are key for the optimization of cyclic nucleotide turnover to respond to the various environments encountered by each parasite and life stage. Understanding how apicomplexan PDEs are regulated and integrating multiple signaling systems into a unified response represent an untapped avenue for future exploration.


Sujet(s)
Cryptosporidiose , Cryptosporidium , Diéthylstilbestrol/analogues et dérivés , Animaux , Humains , Phosphodiesterases/génétique , Nucléotides cycliques , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Phylogenèse , GMP cyclique , 3',5'-Cyclic-AMP Phosphodiesterases
18.
J Med Chem ; 66(21): 14597-14608, 2023 11 09.
Article de Anglais | MEDLINE | ID: mdl-37862143

RÉSUMÉ

Phosphodiesterase 11A4 (PDE11A4) is a dual-acting cyclic nucleotide hydrolase expressed in neurons in the CA1, subiculum, amygdalostriatal transition area and amygdalohippocampal area of the extended hippocampal formation. PDE11A4 is the only PDE enzyme to emanate solely from hippocampal formation, a key brain region for the formation of long-term memory. PDE11A4 expression increases in the hippocampal formation of both humans and rodents as they age. Interestingly, PDE11A knockout mice do not show age-related deficits in associative memory and show no gross histopathology. This suggests that inhibition of PDE11A4 might serve as a therapeutic option for age-related cognitive decline. A novel, yeast-based high throughput screen previously identified moderately potent, selective PDE11A4 inhibitors, and this work describes initial efforts that improved potency more than 10-fold and improved some pharmaceutical properties of one of these scaffolds, leading to selective, cell-penetrant PDE11A4 inhibitors, one of which is 10-fold more potent compared to tadalafil in cell-based activity.


Sujet(s)
Dysfonctionnement cognitif , Inhibiteurs de la phosphodiestérase , Humains , Animaux , Souris , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Inhibiteurs de la phosphodiestérase/métabolisme , 3',5'-Cyclic-GMP Phosphodiesterases/métabolisme , Phosphodiesterases/métabolisme , Encéphale/métabolisme , Souris knockout , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/métabolisme
19.
J Med Chem ; 66(17): 12468-12478, 2023 09 14.
Article de Anglais | MEDLINE | ID: mdl-37584424

RÉSUMÉ

Phosphodiesterase 1 (PDE1) is a subfamily of PDE super enzyme families that can hydrolyze cyclic adenosine monophosphate and cyclic guanosine monophosphate simultaneously. Currently, the number of PDE1 inhibitors is relatively few, significantly limiting their application. Herein, a novel series of quinolin-2(1H)-ones were designed rationally, leading to compound 10c with an IC50 of 15 nM against PDE1C, high selectivity across other PDEs, and remarkable safety properties. Furthermore, we used the lead compound 10c as a chemical tool to explore whether PDE1 could work as a novel potential target for the treatment of inflammatory bowel disease (IBD), a disease which is a chronic, relapsing disorder of the gastrointestinal tract inflammation lacking effective treatment. Our results showed that administration of 10c exerted significant anti-IBD effects in the dextran sodium sulfate-induced mice model and alleviated the inflammatory response, indicating that PDE1 could work as a potent target for IBD.


Sujet(s)
Maladies inflammatoires intestinales , Inhibiteurs de la phosphodiestérase , Souris , Animaux , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Phosphodiesterases , GMP cyclique , AMP cyclique , Maladies inflammatoires intestinales/traitement médicamenteux
20.
Eur J Med Chem ; 259: 115682, 2023 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-37536210

RÉSUMÉ

Cyclic nucleotide phosphodiesterase 9 (PDE9), a specifically hydrolytic enzyme with the highest affinity for cyclic guanosine monophosphate (cGMP) among the phosphodiesterases family, plays a critical role in many biological processes. Consequently, the development of PDE9 inhibitors has received increasing attention in recent years, with several compounds undergoing clinical trials for the treatment of central nervous system (CNS) diseases such as Alzheimer's disease, schizophrenia, and psychotic disorders, as well as heart failure and sickle cell disease. This review analyzes the recent primary literatures and patents published from 2004 to 2023, focusing on the structure, pharmacophores, selectivity, and therapeutic potential of PDE9 inhibitors. It hoped to provide a comprehensive overview of the field's current state to inform the development of novel PDE9 inhibitors.


Sujet(s)
3',5'-Cyclic-AMP Phosphodiesterases , Inhibiteurs de la phosphodiestérase , GMP cyclique , Pharmacophore , Inhibiteurs de la phosphodiestérase/pharmacologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique , Phosphodiesterases
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE