Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 684
Filter
1.
Zhonghua Jie He He Hu Xi Za Zhi ; 47(9): 876-884, 2024 Sep 12.
Article in Chinese | MEDLINE | ID: mdl-39266490

ABSTRACT

Current treatments for chronic obstructive pulmonary disease (COPD) are relatively limited and cannot meet the needs of all patients. Ensifentrine (development code RPL554), a representative drug of cyclic nucleotide phosphodiesterase 3/4 (PDE 3/4) inhibitors, has shown promising developments in the treatment of COPD in recent years, which need to be summarized. This article reviews the mechanism and clinical research progress of ensifentrine, focusing on its chemical structure, pharmacokinetics, pathophysiological mechanism, efficacy, and safety. Additionally, we provide clinical application suggestions and future research prospects.


Subject(s)
Phosphodiesterase 4 Inhibitors , Pulmonary Disease, Chronic Obstructive , Pulmonary Disease, Chronic Obstructive/drug therapy , Humans , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 3 Inhibitors/therapeutic use , Phosphodiesterase 3 Inhibitors/pharmacology , Carbolines/therapeutic use , Carbolines/pharmacology , Isoquinolines , Pyrimidinones
2.
Mol Med ; 30(1): 134, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223490

ABSTRACT

BACKGROUND: Epidermal remodeling and hypertrophy are hallmarks of skin fibrotic disorders, and keratinocyte to mesenchymal (EMT)-like transformations drive epidermis alteration in skin fibrosis such as keloids and hypertrophic scars (HTS). While phosphodiesterase 4 (PDE4) inhibitors have shown effectiveness in various fibrotic disorders, their role in skin fibrosis is not fully understood. This study aimed to explore the specific role of PDE4B in epidermal remodeling and hypertrophy seen in skin fibrosis. METHODS: In vitro experiments examined the effects of inhibiting PDE4A-D (with Roflumilast) or PDE4B (with siRNA) on TGFß1-induced EMT differentiation and dedifferentiation in human 3D epidermis. In vivo studies investigated the impact of PDE4 inhibition on HOCl-induced skin fibrosis and epidermal hypertrophy in mice, employing both preventive and therapeutic approaches. RESULTS: The study found increased levels of PDE4B (mRNA, protein) in keloids > HTS compared to healthy epidermis, as well as in TGFß-stimulated 3D epidermis. Keloids and HTS epidermis exhibited elevated levels of collagen Iα1, fibronectin, αSMA, N-cadherin, and NOX4 mRNA, along with decreased levels of E-cadherin and ZO-1, confirming an EMT process. Inhibition of both PDE4A-D and PDE4B prevented TGFß1-induced Smad3 and ERK1/2 phosphorylation and mesenchymal differentiation in vitro. PDE4A-D inhibition also promoted mesenchymal dedifferentiation and reduced TGFß1-induced ROS and keratinocyte senescence by rescuing PPM1A, a Smad3 phosphatase. In vivo, PDE4 inhibition mitigated HOCl-induced epidermal hypertrophy in mice in both preventive and therapeutic settings. CONCLUSIONS: Overall, the study supports the potential of PDE4 inhibitors, particularly PDE4B, in treating skin fibrosis, including keloids and HTS, shedding light on their functional role in this condition.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Fibrosis , Keloid , Keratinocytes , Phosphodiesterase 4 Inhibitors , Humans , Keloid/metabolism , Keloid/pathology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Keratinocytes/metabolism , Keratinocytes/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Animals , Mice , Epidermis/metabolism , Epidermis/pathology , Transforming Growth Factor beta1/metabolism , Epithelial-Mesenchymal Transition/drug effects , Cell Differentiation/drug effects , Male
3.
Phytomedicine ; 134: 155985, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39236558

ABSTRACT

BACKGROUND: Hua-Shi-Bai-Du decoction (HSBD) exerts significant effects on the prevention and treatment of COVID-19 in China. The activation of the NLRP3 inflammasome of macrophages plays a vital role in COVID-19 pathology. However, no previous studies have focused on this pathological process to explore the effect of HSBD. PURPOSE: Our aim is to uncover the effect of HSBD on NLRP3 inflammasome activation and the underlying mechanisms. METHODS: The NLRP3-activated J774A.1 cells primed by LPS and activated by nigericin/ATP/MSU were used to evaluate NLRP3 activation in vitro. ASC oligomerization and speck formation were assessed by western blot and immunofluorescence imaging. Intracellular K+ levels were determined by the colorimetric assay. Mitochondrial ROS (mtROS) level was detected by the flow cytometry and the fluorescence spectrophotometry. The intracellular cAMP level was determined by chemiluminescence method and ELISA, while phosphodiesterase (PDE) activity was measured using the fluorescent substrate MANT-cAMP. siRNA was applied to knockdown PDE4B. Two in vivo mouse models, MSU-induced peritonitis and LPS-induced acute lung injury (ALI), were used to evaluate the effects of HSBD on IL-1ß and other inflammatory cytokines. Pathological changes in lung tissue were observed by histopathological examination. RESULTS: HSBD not only decreased supernatant IL-1ß, caspase-1 p20, and cleaved gasdermin D (GSDMD) in NLRP3-activated J774A.1 cells, but also reduced IL-1ß in the peritoneal lavage fluid of mice with MSU-induced peritonitis, demonstrating the suppressive effect on NLRP3 inflammasome activation. The mechanism study showed that HSBD blocked ASC oligomerization and speck formation without affecting K+ efflux or mtROS production. Furthermore, it prevented the decrease of intracellular cAMP by inhibiting PDE4B activity. And in the PDE4B-deficient cells, its suppressive effect on IL-1ß release was abolished. In LPS-induced ALI mice, oral administration of HSBD decreased several proinflammatory cytokines (IL-1ß, IL-6, TNF-α, and CXCL-1) and attenuated the pathological damage to the lung. CONCLUSION: HSBD suppresses the activation of NLRP3 inflammasome by inhibiting PDE4B activity to counteract the decrease of intracellular cAMP, thereby blocking ASC oligomerization in macrophages. Our findings may provide new insight into the clinical effets of HSBD for the treatment of COVID-19.


Subject(s)
Acute Lung Injury , Cyclic Nucleotide Phosphodiesterases, Type 4 , Drugs, Chinese Herbal , Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Acute Lung Injury/drug therapy , Mice , Inflammasomes/metabolism , Inflammasomes/drug effects , Drugs, Chinese Herbal/pharmacology , Macrophages/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Male , Disease Models, Animal , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Cell Line , COVID-19 , Interleukin-1beta/metabolism , Lipopolysaccharides
4.
Rapid Commun Mass Spectrom ; 38(23): e9916, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39307998

ABSTRACT

RATIONALE: Phosphodiesterase 4 (PDE4) inhibitors are a newer class of drugs that induce bronchodilation and have anti-inflammatory effects, making them susceptible to misuse as performance enhancers in competitive sports. METHODS: This study explores the metabolic conversion of PDE4 inhibitor ibudilast in thoroughbred horses after oral administration and in vitro using equine liver microsomes and Cunninghamella elegans. A liquid chromatography-high resolution mass spectrometry method was used to postulate the plausible structures of the detected metabolites. RESULTS: A total of 20 in vivo metabolites were identified under experimental conditions, including 12 Phase I and 8 Phase II conjugated metabolites. Phase I metabolites were predominantly formed through hydroxylation (mono-, di-, and tri-hydroxylation). Demethylated metabolites were also identified during this investigation. Additionally, the research detected Phase II metabolites conjugated with glucuronic and sulfonic acids. CONCLUSIONS: The data presented here can assist in detecting the PDE4 inhibitor ibudilast and uncover its illicit use in competitive sports.


Subject(s)
Microsomes, Liver , Phosphodiesterase 4 Inhibitors , Pyridines , Animals , Horses , Phosphodiesterase 4 Inhibitors/metabolism , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Pyridines/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/analysis , Microsomes, Liver/metabolism , Microsomes, Liver/drug effects , Performance-Enhancing Substances/metabolism , Performance-Enhancing Substances/chemistry , Performance-Enhancing Substances/pharmacology , Doping in Sports , Indolizines , Pyrazoles
5.
Eur J Pharmacol ; 983: 177015, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39332796

ABSTRACT

BACKGROUND: Phosphodiesterase-4 (PDE4) is responsible for terminating cyclic adenosine monophosphate (cAMP) signalling. PDE4 inhibitors, such as roflumilast (RFM), have anti-inflammatory activity and have been studied in inflammation-induced tissue damage in sepsis. However, the role of RFM on cardiovascular derangements induced by sepsis is still unknown. Thus, we aimed to evaluate the potential effects of RFM on cardiovascular collapse and multiorgan damage caused by sepsis. METHODS: Sepsis was induced by cecal ligation and puncture (CLP) in male rats. Six hours after the CLP or sham procedure, animals were randomly assigned to receive either RFM (0.3 mg/kg) or vehicle subcutaneously, and cardiovascular parameters were assessed 24 h after the surgery and organ/plasma samples were collected for further analyses. RESULTS: Sepsis induced hypotension, tachycardia, reduced renal blood flow (RBF) and hyporeactivity to vasoconstrictors both in vivo and ex vivo. RFM treatment increased systemic cAMP levels and RBF. RFM also attenuated hypoperfusion and liver damage induced by CLP. Furthermore, RFM reduced systemic nitric oxide (NO) levels in septic rats, while there were no changes in hepatic NOS-2 expression. Nevertheless, RFM exacerbated sepsis-induced hypotension and tachycardia without ameliorating vascular hyporeactivity. CONCLUSION: Our data show that PDE-4 inhibition protects septic rats from hepatic injury and improves renal perfusion. However, RFM worsened hemodynamic parameters and showed no protection against sepsis-induced cardiovascular dysfunction and mortality. Thus, despite the anti-inflammatory benefits of RFM, its application in sepsis should be approached cautiously.


Subject(s)
Aminopyridines , Benzamides , Cyclic AMP , Cyclopropanes , Phosphodiesterase 4 Inhibitors , Sepsis , Animals , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Sepsis/complications , Sepsis/drug therapy , Male , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Cyclopropanes/administration & dosage , Benzamides/pharmacology , Benzamides/therapeutic use , Rats , Cyclic AMP/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Nitric Oxide/metabolism , Cardiovascular System/drug effects , Cardiovascular System/physiopathology , Rats, Sprague-Dawley
6.
Bioorg Chem ; 151: 107689, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39111119

ABSTRACT

Immune-mediated inflammatory diseases (IMIDs) comprise a broad spectrum of conditions characterized by systemic inflammation affecting various organs and tissues, for which there is no known cure. The isoform-specific inhibition of phosphodiesterase-4B (PDE4B) over PDE4D constitutes an effective therapeutic strategy for the treatment of IMIDs that minimizes the adverse effects associated with non-selective PDE4 inhibitors. Thus, we report a new class of isoquinolone derivatives as next-generation PDE4 inhibitors for effective management of rheumatoid arthritis (RA) and psoriasis. Among the series, 8 compounds i.e. 1e, 1l, 1m, 1n, 1o, 2m, 2o and 3o showed promising PDE4B inhibition (>80 %) in vitro with IC50 ∼ 1.4-6.2 µM. The compound 1l was identified as an initial hit and was pursued for further studies. According to structure-activity relationship (SAR), an allyl group at C-4 position improved PDE4B inhibition. The correlation between in vitro activity data and binding affinities obtained via molecular docking suggested that the high-affinity binding to PDE4B is a prerequisite for the effective inhibition of PDE4B. Notably, the hit 1l showed selectivity towards PDE4B over PDE4D in vitro. Furthermore, 1l treatment (30 mg/kg) in the adjuvant-induced arthritis (AIA) rat model induced by complete Freund's adjuvant (CFA) demonstrated anti-arthritic potential via ameliorating paw swelling and body weight, narrowing joint space, reducing excessive immune cells infiltration and pannus formation in addition to reducing mRNA expression of pro-inflammatory cytokines such as TNF-α and IL-6 in synovial tissues of experimental rats. Additionally, 1l reduced the hyper-proliferative state and colony forming potential of IMQ-induced psoriatic keratinocytes. The treatment of these cells with 1l markedly reduced the protein levels of Ki67 and mRNA levels of pro-inflammatory cytokines e.g. IL-17A and TNF-α suggesting its potent anti-psoriatic potential. Furthermore, 1l did not show any significant adverse effects when evaluated in a systematic toxicity (e.g. teratogenicity, hepatotoxicity and cardiotoxicity) studies in zebrafish at the tested concentrations (1-100 µM) and the NOAEL (no-observed-adverse-effect level) was found to be 100 µM. Thus, with promising anti-inflammatory effects both in vitro and in vivo along with PDE4B selectivity with an acceptable safety margin, 1l emerged as a new and promising inhibitor for further studies.


Subject(s)
Arthritis, Rheumatoid , Cyclic Nucleotide Phosphodiesterases, Type 4 , Phosphodiesterase 4 Inhibitors , Psoriasis , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/therapeutic use , Psoriasis/drug therapy , Arthritis, Rheumatoid/drug therapy , Animals , Structure-Activity Relationship , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Humans , Rats , Molecular Structure , Dose-Response Relationship, Drug , Molecular Docking Simulation , Zebrafish , Arthritis, Experimental/drug therapy , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Male
7.
Chem Biol Interact ; 402: 111211, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39197814

ABSTRACT

There is a pressing medical need for improved treatments in skin fibrosis including keloids and hypertrophic scars (HTS). This study aimed to characterize the role of phosphodiesterase 4 (PDE4), specifically PDE4B in fibrotic skin remodeling in vitro and in vivo. In vitro, effects of PDE4A-D (Roflumilast) or PDE4B (siRNA) inhibition on TGFß1-induced myofibroblast differentiation and dedifferentiation were studied in normal (NHDF) and keloid (KF) human dermal fibroblasts. In vivo, the role of PDE4 on HOCl-induced skin fibrosis in mice was addressed in preventive and therapeutic protocols. PDE4B (mRNA, protein) was increased in Keloid > HTS compared to healthy skin and in TGFß-stimulated NHDF and KF. In Keloid > HTS, collagen Iα1, αSMA, TGFß1 and NOX4 mRNA were all elevated compared to healthy skin confirming skin fibrosis. In vitro, inhibition of PDE4A-D and PDE4B similarly prevented TGFß1-induced Smad3 and ERK1/2 phosphorylation and myofibroblast differentiation, elevated NOX4 protein and proliferation in NHDF. PDE4A-D inhibition enabled myofibroblast dedifferentiation and curbed TGFß1-induced reactive oxygen species and fibroblast senescence. In KF PDE4A-D inhibition restrained TGFß1-induced Smad3 and ERK1/2 phosphorylation, myofibroblast differentiation and senescence. Mechanistically, PDE4A-D inhibition rescued from TGFß1-induced loss in PPM1A, a Smad3 phosphatase. In vivo, PDE4 inhibition mitigated HOCl-induced skin fibrosis in mice in preventive and therapeutic protocols. The current study provides novel evidence evolving rationale for PDE4 inhibitors in skin fibrosis (including keloids and HTS) and delivered evidence for a functional role of PDE4B in this fibrotic condition.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Fibroblasts , Fibrosis , Keloid , Phosphodiesterase 4 Inhibitors , Skin , Transforming Growth Factor beta1 , Keloid/pathology , Keloid/metabolism , Humans , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Mice , Phosphodiesterase 4 Inhibitors/pharmacology , Transforming Growth Factor beta1/metabolism , Skin/pathology , Skin/metabolism , Skin/drug effects , Cell Differentiation/drug effects , Male , Cells, Cultured , NADPH Oxidase 4/metabolism , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , Hypochlorous Acid/metabolism , Reactive Oxygen Species/metabolism , Smad3 Protein/metabolism , Cell Proliferation/drug effects , Female
8.
Eur J Med Chem ; 277: 116710, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39098133

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic and progressive condition with a significant global burden. Currently, available treatments primarily provide symptomatic relief and retard disease progression, yet they do not offer a cure and are frequently associated with adverse effects. Therefore, the discovery of new targets and therapeutic drugs for IBD is crucial. Phosphodiesterase 4 (PDE4) inhibitors have emerged as promising candidates in the search for effective IBD treatments, although dose-dependent side effects hamper their clinical utility. In this study, building upon heterocyclic biaryl derivatives (TPA16), we designed and synthesized a series of N2-substituted indazole-based PDE4D inhibitors, emphasizing improving safety profiles. An enzyme activity screening discovered an optimized compound, LZ-14 (Z21115), which exhibited high PDE4D7 (IC50 = 10.5 nM) inhibitory activity and good selectivity. More interestingly, LZ-14 has demonstrated promising effects in treating IBD in mouse models by improving the inflammatory response and colon injury. Furthermore, LZ-14 displayed low emetogenic potential in ketamine/xylazine anesthesia mice alternative models.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Drug Discovery , Indazoles , Inflammatory Bowel Diseases , Phosphodiesterase 4 Inhibitors , Animals , Inflammatory Bowel Diseases/drug therapy , Structure-Activity Relationship , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/therapeutic use , Mice , Indazoles/pharmacology , Indazoles/chemistry , Indazoles/chemical synthesis , Humans , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Molecular Structure , Dose-Response Relationship, Drug , Male , Mice, Inbred C57BL
9.
Drugs ; 84(9): 1157-1163, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39196510

ABSTRACT

Ensifentrine, an inhaled, selective phosphodiesterase (PDE) 3 and PDE4 inhibitor, is being developed by Verona Pharma plc for the treatment of respiratory diseases, including chronic obstructive pulmonary disease (COPD). In June 2024, ensifentrine (OHTUVAYRE™) inhalation suspension was approved for the maintenance treatment of COPD in adult patients in the USA. This article summarizes the milestones in the development of ensifentrine leading to this first approval for the maintenance treatment of COPD.


Subject(s)
Drug Approval , Phosphodiesterase 4 Inhibitors , Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/drug therapy , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/administration & dosage , Administration, Inhalation , Isoquinolines/therapeutic use , Isoquinolines/pharmacology , Isoquinolines/administration & dosage , Phosphodiesterase 3 Inhibitors/pharmacology , Phosphodiesterase 3 Inhibitors/therapeutic use , Phosphodiesterase 3 Inhibitors/administration & dosage , United States , Pyrimidinones
10.
Dermatologie (Heidelb) ; 75(10): 791-797, 2024 Oct.
Article in German | MEDLINE | ID: mdl-39212723

ABSTRACT

BACKGROUND: Chronic inflammatory skin diseases are of great social and medical importance and require effective drug therapy. Phosphodiesterase 4 (PDE4) inhibitors represent a possible therapeutic option by regulating inflammatory processes. PDEs cause the release of proinflammatory cytokines by interfering with signaling pathways. The PDE4 inhibitors apremilast (treatment of psoriasis and Behçet's disease), roflumilast (treatment of chronic obstructive pulmonary disease), and crisaborole (treatment of atopic dermatitis) are currently approved in Europe. PSORIASIS: Apremilast is used for second-line treatment of plaque psoriasis and psoriatic arthritis and has a favorable side effect profile. Topical PDE4 inhibitors are currently being researched and have not yet been approved by the European Medicines Agency (EMA). ATOPIC DERMATITIS: The topical PDE4 inhibitor crisaborole was approved by the EMA in 2020 as a topical treatment alternative to glucocorticoids and calcineurin inhibitors. Although the substance has shown good tolerability in studies and also alleviates the accompanying itching, it did not find its way onto the German market. BEHçET'S DISEASE: Apremilast is approved for the treatment of Behçet's disease in adults with refractory, severe oral ulcers. OUTLOOK: Case studies have also demonstrated the efficacy of systemic PDE4 inhibition in other skin diseases (including blistering autoimmune dermatoses, lichen planus, and acantholytic genodermatoses). The substances are also being researched and used to treat extracutaneous inflammatory diseases.


Subject(s)
Phosphodiesterase 4 Inhibitors , Psoriasis , Thalidomide , Humans , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/adverse effects , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Thalidomide/pharmacology , Thalidomide/adverse effects , Psoriasis/drug therapy , Skin Diseases/drug therapy , Benzamides/therapeutic use , Benzamides/pharmacology , Aminopyridines/therapeutic use , Aminopyridines/adverse effects , Boron Compounds/therapeutic use , Boron Compounds/pharmacology , Behcet Syndrome/drug therapy , Dermatitis, Atopic/drug therapy , Cyclopropanes , Bridged Bicyclo Compounds, Heterocyclic
11.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125619

ABSTRACT

Phosphodiesterase 4 (PDE4) enzymes catalyze cyclic adenosine monophosphate (cAMP) hydrolysis and are involved in a variety of physiological processes, including brain function, monocyte and macrophage activation, and neutrophil infiltration. Among different PDE4 isoforms, Phosphodiesterases 4D (PDE4Ds) play a fundamental role in cognitive, learning and memory consolidation processes and cancer development. Selective PDE4D inhibitors (PDE4Dis) could represent an innovative and valid therapeutic strategy for the treatment of various neurodegenerative diseases, such as Alzheimer's, Parkinson's, Huntington's, and Lou Gehrig's diseases, but also for stroke, traumatic brain and spinal cord injury, mild cognitive impairment, and all demyelinating diseases such as multiple sclerosis. In addition, small molecules able to block PDE4D isoforms have been recently studied for the treatment of specific cancer types, particularly hepatocellular carcinoma and breast cancer. This review overviews the PDE4DIsso far identified and provides useful information, from a medicinal chemistry point of view, for the development of a novel series of compounds with improved pharmacological properties.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Phosphodiesterase 4 Inhibitors , Humans , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/chemistry , Animals , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism
12.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39063153

ABSTRACT

Difamilast, a phosphodiesterase 4 (PDE4) inhibitor, has been shown to be effective in the treatment of atopic dermatitis (AD), although the mechanism involved remains unclear. Since IL-33 plays an important role in the pathogenesis of AD, we investigated the effect of difamilast on IL-33 activity. Since an in vitro model of cultured normal human epidermal keratinocytes (NHEKs) has been utilized to evaluate the pharmacological potential of adjunctive treatment of AD, we treated NHEKs with difamilast and analyzed the expression of the suppression of tumorigenicity 2 protein (ST2), an IL-33 receptor with transmembrane (ST2L) and soluble (sST2) isoforms. Difamilast treatment increased mRNA and protein levels of sST2, a decoy receptor suppressing IL-33 signal transduction, without affecting ST2L expression. Furthermore, supernatants from difamilast-treated NHEKs inhibited IL-33-induced upregulation of TNF-α, IL-5, and IL-13 in KU812 cells, a basophil cell line sensitive to IL-33. We also found that difamilast activated the aryl hydrocarbon receptor (AHR)-nuclear factor erythroid 2-related factor 2 (NRF2) axis. Additionally, the knockdown of AHR or NRF2 abolished the difamilast-induced sST2 production. These results indicate that difamilast treatment produces sST2 via the AHR-NRF2 axis, contributing to improving AD symptoms by inhibiting IL-33 activity.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Keratinocytes , NF-E2-Related Factor 2 , Phosphodiesterase 4 Inhibitors , Receptors, Aryl Hydrocarbon , Signal Transduction , Humans , NF-E2-Related Factor 2/metabolism , Keratinocytes/metabolism , Keratinocytes/drug effects , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-1 Receptor-Like 1 Protein/genetics , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Phosphodiesterase 4 Inhibitors/pharmacology , Interleukin-33/metabolism , Signal Transduction/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line
13.
Biomed Pharmacother ; 177: 117009, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908196

ABSTRACT

Cyclic adenosine monophosphate (cAMP) is a key second messenger that regulates signal transduction pathways pivotal for numerous biological functions. Intracellular cAMP levels are spatiotemporally regulated by their hydrolyzing enzymes called phosphodiesterases (PDEs). It has been shown that increased cAMP levels in the central nervous system (CNS) promote neuroplasticity, neurotransmission, neuronal survival, and myelination while suppressing neuroinflammation. Thus, elevating cAMP levels through PDE inhibition provides a therapeutic approach for multiple CNS disorders, including multiple sclerosis, stroke, spinal cord injury, amyotrophic lateral sclerosis, traumatic brain injury, and Alzheimer's disease. In particular, inhibition of the cAMP-specific PDE4 subfamily is widely studied because of its high expression in the CNS. So far, the clinical translation of full PDE4 inhibitors has been hampered because of dose-limiting side effects. Hence, focusing on signaling cascades downstream activated upon PDE4 inhibition presents a promising strategy, offering novel and pharmacologically safe targets for treating CNS disorders. Yet, the underlying downstream signaling pathways activated upon PDE(4) inhibition remain partially elusive. This review provides a comprehensive overview of the existing knowledge regarding downstream mediators of cAMP signaling induced by PDE4 inhibition or cAMP stimulators. Furthermore, we highlight existing gaps and future perspectives that may incentivize additional downstream research concerning PDE(4) inhibition, thereby providing novel therapeutic approaches for CNS disorders.


Subject(s)
Central Nervous System Diseases , Central Nervous System , Cyclic AMP , Cyclic Nucleotide Phosphodiesterases, Type 4 , Phosphodiesterase 4 Inhibitors , Signal Transduction , Humans , Cyclic AMP/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Signal Transduction/drug effects , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism
14.
Eur J Med Chem ; 275: 116576, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38861808

ABSTRACT

Sepsis can quickly result in fatality for critically ill individuals, while liver damage can expedite the progression of sepsis, necessitating the exploration of new strategies for treating hepatic sepsis. PDE4 has been identified as a potential target for the treatment of liver damage. The scaffold hopping of lead compounds FCPR16 and Z19153 led to the discovery of a novel 7-methoxybenzofuran PDE4 inhibitor 4e, demonstrating better PDE4B (IC50 = 10.0 nM) and PDE4D (IC50 = 15.2 nM) inhibitor activity as a potential anti-hepatic sepsis drug in this study. Compared with FCPR16 and Z19153, 4e displayed improved oral bioavailability (F = 66 %) and longer half-life (t1/2 = 2.0 h) in SD rats, which means it can be more easily administered and has a longer-lasting effect. In the D-GalN/LPS-induced liver injury model, 4e exhibited excellent hepatoprotective activity against hepatic sepsis by decreasing ALT and AST levels and inflammatory infiltrating areas.


Subject(s)
Benzofurans , Galactosamine , Phosphodiesterase 4 Inhibitors , Sepsis , Animals , Humans , Male , Rats , Benzofurans/pharmacology , Benzofurans/chemistry , Benzofurans/chemical synthesis , Chemical and Drug Induced Liver Injury/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dose-Response Relationship, Drug , Drug Discovery , Galactosamine/pharmacology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Liver/drug effects , Liver/pathology , Molecular Docking Simulation , Molecular Structure , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/chemical synthesis , Protective Agents/pharmacology , Protective Agents/chemistry , Protective Agents/chemical synthesis , Rats, Sprague-Dawley , Sepsis/drug therapy , Structure-Activity Relationship
15.
Cells ; 13(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38920631

ABSTRACT

Microglia activity can drive excessive synaptic loss during the prodromal phase of Alzheimer's disease (AD) and is associated with lowered cyclic adenosine monophosphate (cAMP) due to cAMP phosphodiesterase 4B (PDE4B). This study aimed to investigate whether long-term inhibition of PDE4B by A33 (3 mg/kg/day) can prevent synapse loss and its associated cognitive decline in APPswe/PS1dE9 mice. This model is characterized by a chimeric mouse/human APP with the Swedish mutation and human PSEN1 lacking exon 9 (dE9), both under the control of the mouse prion protein promoter. The effects on cognitive function of prolonged A33 treatment from 20 days to 4 months of age, was assessed at 7-8 months. PDE4B inhibition significantly improved both the working and spatial memory of APPswe/PSdE9 mice after treatment ended. At the cellular level, in vitro inhibition of PDE4B induced microglial filopodia formation, suggesting that regulation of PDE4B activity can counteract microglia activation. Further research is needed to investigate if this could prevent microglia from adopting their 'disease-associated microglia (DAM)' phenotype in vivo. These findings support the possibility that PDE4B is a potential target in combating AD pathology and that early intervention using A33 may be a promising treatment strategy for AD.


Subject(s)
Alzheimer Disease , Cognition , Cyclic Nucleotide Phosphodiesterases, Type 4 , Disease Models, Animal , Mice, Transgenic , Microglia , Phosphodiesterase 4 Inhibitors , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Cognition/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/administration & dosage , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Presenilin-1/genetics , Presenilin-1/metabolism , Humans , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Male
16.
Expert Opin Ther Targets ; 28(7): 545-573, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38878273

ABSTRACT

INTRODUCTION: Chronic liver disease (CLD) is a complex disease associated with profound dysfunction. Despite an incredible burden, the first and only pharmacotherapy for metabolic-associated steatohepatitis was only approved in March of this year, indicating a gap in the translation of preclinical studies. There is a body of preclinical work on the application of phosphodiesterase 4 inhibitors in CLD, none of these molecules have been successfully translated into clinical use. AREAS COVERED: To design therapies to combat CLD, it is essential to consider the dysregulation of other tissues that contribute to its development and progression. As such, proper therapies must combat this throughout the body rather than focusing only on the liver. To detail this, literature characterizing the pathogenesis of CLD was pulled from PubMed, with a particular focus placed on the role of PDE4 in inflammation and metabolism. Then, the focus is shifted to detailing the available information on existing PDE4 inhibitors. EXPERT OPINION: This review gives a brief overview of some of the pathologies of organ systems that are distinct from the liver but contribute to disease progression. The demonstrated efficacy of PDE4 inhibitors in other human inflammatory diseases should earn them further examination for the treatment of CLD.


Subject(s)
Adipose Tissue , Cyclic Nucleotide Phosphodiesterases, Type 4 , Phosphodiesterase 4 Inhibitors , Humans , Phosphodiesterase 4 Inhibitors/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Adipose Tissue/metabolism , Adipose Tissue/drug effects , Liver Diseases/drug therapy , Liver Diseases/physiopathology , Inflammation/drug therapy , Inflammation/physiopathology , Disease Progression , Chronic Disease , Drug Development
17.
J Nat Prod ; 87(7): 1808-1816, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-38943602

ABSTRACT

Four new p-terphenyl derivatives, talaroterphenyls A-D (1-4), together with three biosynthetically related known ones (5-7), were obtained from the mangrove sediment-derived Talaromyces sp. SCSIO 41412. Compounds 1-3 are rare p-terphenyls, which are completely substituted on the central benzene ring by oxygen atoms; this is the first report of their isolation from natural sources. Their structures were elucidated through NMR spectroscopy, HRESIMS, and X-ray diffraction. Genome sequence analysis revealed that 1-7 were biosynthesized from tyrosine and phenylalanine, involving four key biosynthetic genes (ttpB-ttpE). These p-terphenyls (1-7) and 36 marine-derived terphenyl analogues (8-43) were screened for phosphodiesterase 4 (PDE4) inhibitory activities, and 1-5, 14, 17, 23, and 26 showed notable activities with IC50 values of 0.40-16 µM. The binding pattern of p-terphenyl inhibitors 1-3 with PDE4 were explored by molecular docking analysis. Talaroterphenyl A (1), with a low cytotoxicity, showed obvious anti-inflammatory activity in LPS-stimulated RAW264.7 cells. Furthermore, in the TGF-ß1-induced medical research council cell strain-5 (MRC-5) pulmonary fibrosis model, 1 could down-regulate the expression levels of FN1, COL1, and α-SMA significantly at concentrations of 5-20 µM. This study suggests that the oxidized p-terphenyl 1, as a marine-derived PDE4 inhibitor, could be used as a promising antifibrotic agent.


Subject(s)
Phosphodiesterase 4 Inhibitors , Terphenyl Compounds , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/isolation & purification , Mice , Animals , Terphenyl Compounds/pharmacology , Terphenyl Compounds/chemistry , Terphenyl Compounds/isolation & purification , Molecular Structure , Talaromyces/chemistry , RAW 264.7 Cells , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Marine Biology
18.
Neuropharmacology ; 257: 110035, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38876310

ABSTRACT

We previously showed that the PDE4 inhibitor apremilast reduces ethanol consumption in mice by protein kinase A (PKA) and GABAergic mechanisms. Preventing PKA phosphorylation of GABAA ß3 subunits partially blocked apremilast-mediated decreases in drinking. Here, we produced Gabrb1-S409A mice to render GABAA ß1 subunits resistant to PKA-mediated phosphorylation. Mass spectrometry confirmed the presence of the S409A mutation and lack of changes in ß1 subunit expression or phosphorylation at other residues. ß1-S409A male and female mice did not differ from wild-type C57BL/6J mice in expression of Gabrb1, Gabrb2, or Gabrb3 subunits or in behavioral characteristics. Apremilast prolonged recovery from ethanol ataxia to a greater extent in Gabrb1-S409A mice but prolonged recovery from zolpidem and propofol to a similar extent in both genotypes. Apremilast shortened recovery from diazepam ataxia in wild-type but prolonged recovery in Gabrb1-S409A mice. In wild-type mice, the PKA inhibitor H89 prevented apremilast modulation of ataxia by ethanol and diazepam, but not by zolpidem. In Gabrb1-S409A mice, inhibiting PKA or EPAC2 (exchange protein directly activated by cAMP) partially reversed apremilast potentiation of ethanol, diazepam, and zolpidem ataxia. Apremilast prevented acute tolerance to ethanol ataxia in both genotypes, but there were no genotype differences in ethanol consumption before or after apremilast. In contrast to results in Gabrb3-S408A/S409A mice, PKA phosphorylation of ß1-containing GABAA receptors is not required for apremilast's effects on acute tolerance or on ethanol consumption but is required for its ability to decrease diazepam intoxication. Besides PKA we identified EPAC2 as an additional cAMP-dependent mechanism by which apremilast regulates responses to GABAergic drugs.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Ethanol , Mice, Inbred C57BL , Phosphodiesterase 4 Inhibitors , Receptors, GABA-A , Thalidomide , Animals , Thalidomide/pharmacology , Thalidomide/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Male , Female , Ethanol/pharmacology , Mice , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Receptors, GABA-A/drug effects , Gene Knock-In Techniques , Phosphorylation/drug effects , Ataxia/genetics , Alcohol Drinking/drug therapy , Alcohol Drinking/genetics , Mice, Transgenic , Diazepam/pharmacology
19.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 53(3): 390-398, 2024 Jun 06.
Article in English, Chinese | MEDLINE | ID: mdl-38860393

ABSTRACT

Phosphodiesterases (PDE) are involved in the regulation of cellular physiological processes and neurological functions, including neuronal plasticity, synapto-genesis, synaptic transmission, memory formation and cognitive functions by catalyzing the hydrolysis of intracellular cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Many basic and clinical studies have shown that PDE4 inhibitors block or ameliorate the occurrence and development of central nervous system (CNS) diseases by inhibiting cAMP hydrolysis, increasing cAMP content and enhancing its downstream effects. PDE4 inhibitors have long-term potentiation effect, which can enhance phosphorylation of cAMP response element binding protein (CREB) and upregulate expression of memory related Arc genes in hippocampal neurons, thereby improving cognitive impairment and Alzheimer's disease-like symptoms. They can also delay the occurrence and development of Parkinson's disease by reducing the cytotoxicity induced by α-syn and increasing the effect of miR-124-3p on cell functions. Alteration of PDE4 activity is the molecular basis for psychosis and some cognitive disorders, therefore it is considered as a therapeutic target for schizophrenia. PDE4 inhibitors play a role in depression by inhibiting the advanced glycation end product receptor (RAGE), TLR4 and NLRP3 pathways in the hippocampus, reducing the activation of microglia and the production of IL-1ß, down-regulating HMGB1/RAGE signaling pathway and inhibiting inflammatory factors. PDE4 inhibitor plays a role in the treatment of autism spectrum disorder by reducing the damage of cerebellar glial cells, increasing nociceptive threshold, and improving mutual learning and memory deficits. PDE4 inhibitors might be used in the treatment of fragile X syndrome by regulating the level of cAMP and affecting the expression of fragile X mental retardation protein (FMRP). PDE4 inhibitors can also promote the differentiation of oligodendrocyte progenitor cells and enhance myelination, which has potential in the treatment of multiple sclerosis. PDE4 is also related to bipolar disorder, which may be one of the therapeutic targets. At present, several PDE4 inhibitors are in clinical trials for the treatment of CNS diseases. This article reviews and discusses the progress on basic research and clinical trials of PDE4 inhibitors in CNS diseases, providing a reference for the prevention and treatment of CNS diseases and the development of new drugs.


Subject(s)
Central Nervous System Diseases , Phosphodiesterase 4 Inhibitors , Humans , Central Nervous System Diseases/drug therapy , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Animals , Cyclic AMP/metabolism , Neuronal Plasticity/drug effects , Signal Transduction/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism
20.
Bioorg Chem ; 149: 107474, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805909

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and high mortality lung disease. Although the antifibrotic drugs pirfenidone and nintedanib could slow the rate of lung function decline, the usual course of the condition is inexorably to respiratory failure and death. Therefore, new approaches and novel therapeutic drugs for the treatment of IPF are urgently needed. And the selective PDE4 inhibitor has in vivo and in vitro anti-fibrotic effects in IPF models. But the clinical application of most PDE4 inhibitors are limited by their unexpected and severe side effects such as nausea, vomiting, and diarrhea. Herein, structure-based optimizations of the natural product Moracin M resulted in a novel a novel series of 2-arylbenzofurans as potent PDE4 inhibitors. The most potent inhibitor L13 has an IC50 of 36 ± 7 nM with remarkable selectivity across the PDE families and administration of L13·citrate (10.0 mg/kg) exhibited comparable anti-pulmonary fibrosis effects to pirfenidone (300 mg/kg) in a bleomycin-induced IPF mice model, indicate that L13 is a potential lead for the treatment of IPF.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Idiopathic Pulmonary Fibrosis , Phosphodiesterase 4 Inhibitors , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/chemically induced , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/therapeutic use , Animals , Structure-Activity Relationship , Mice , Molecular Structure , Humans , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Bleomycin , Dose-Response Relationship, Drug , Mice, Inbred C57BL , Male , Benzofurans/pharmacology , Benzofurans/chemistry , Benzofurans/chemical synthesis
SELECTION OF CITATIONS
SEARCH DETAIL