Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
BMC Cancer ; 24(1): 624, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778317

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) has a high mortality rate, and the mechanisms underlying tumor development and progression remain unclear. However, inactivated tumor suppressor genes might play key roles. DNA methylation is a critical regulatory mechanism for inactivating tumor suppressor genes in HCC. Therefore, this study investigated methylation-related tumor suppressors in HCC to identify potential biomarkers and therapeutic targets. METHODS: We assessed genome-wide DNA methylation in HCC using whole genome bisulfite sequencing (WGBS) and RNA sequencing, respectively, and identified the differential expression of methylation-related genes, and finally screened phosphodiesterase 7B (PDE7B) for the study. The correlation between PDE7B expression and clinical features was then assessed. We then analyzed the changes of PDE7B expression in HCC cells before and after DNA methyltransferase inhibitor treatment by MassArray nucleic acid mass spectrometry. Furthermore, HCC cell lines overexpressing PDE7B were constructed to investigate its effect on HCC cell function. Finally, GO and KEGG were applied for the enrichment analysis of PDE7B-related pathways, and their effects on the expression of pathway proteins and EMT-related factors in HCC cells were preliminarily explored. RESULTS: HCC exhibited a genome-wide hypomethylation pattern. We screened 713 hypomethylated and 362 hypermethylated mCG regions in HCC and adjacent normal tissues. GO analysis showed that the main molecular functions of hypermethylation and hypomethylation were "DNA-binding transcriptional activator activity" and "structural component of ribosomes", respectively, whereas KEGG analysis showed that they were enriched in "bile secretion" and "Ras-associated protein-1 (Rap1) signaling pathway", respectively. PDE7B expression was significantly down-regulated in HCC tissues, and this low expression was negatively correlated with recurrence and prognosis of HCC. In addition, DNA methylation regulates PDE7B expression in HCC. On the contrary, overexpression of PDE7B inhibited tumor proliferation and metastasis in vitro. In addition, PDE7B-related genes were mainly enriched in the PI3K/ATK signaling pathway, and PDE7B overexpression inhibited the progression of PI3K/ATK signaling pathway-related proteins and EMT. CONCLUSION: PDE7B expression in HCC may be regulated by promoter methylation. PDE7B can regulate the EMT process in HCC cells through the PI3K/AKT pathway, which in turn affects HCC metastasis and invasion.


Subject(s)
Carcinoma, Hepatocellular , Cyclic Nucleotide Phosphodiesterases, Type 7 , DNA Methylation , Gene Expression Regulation, Neoplastic , Liver Neoplasms , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Cell Line, Tumor , Neoplasm Invasiveness/genetics , Genes, Tumor Suppressor , Male , Cell Proliferation/genetics , Female , Neoplasm Metastasis , Cell Movement/genetics
2.
Article in English | MEDLINE | ID: mdl-36220621

ABSTRACT

BACKGROUND: PDEs regulate cAMP levels which is critical for PKA activity-dependent activation of CREB-mediated transcription in learning and memory. Inhibitors of PDEs like PDE4 and Pde7 improve learning and memory in rodents. However, the role of PDE7 in cognition or learning and memory has not been reported yet. METHODS: Therefore, we aimed to explore the cognitive effects of a PDE7 subtype, PDE7a, using combined pharmacological and genetic approaches. RESULTS: PDE7a-nko mice showed deficient working memory, impaired novel object recognition, deficient spatial learning & memory, and contextual fear memory, contrary to enhanced cued fear memory, highlighting the potential opposite role of PDE7a in the hippocampal neurons. Further, pharmacological inhibition of PDE7 by AGF2.20 selectively strengthens cued fear memory in C57BL/6 J mice, decreasing its extinction but did not affect cognitive processes assessed in other behavioral tests. The further biochemical analysis detected deficient cAMP in neural cell culture with genetic excision of the PDE7a gene, as well as in the hippocampus of PDE7a-nko mice in vivo. Importantly, we found overexpression of PKA-R and the reduced level of pPKA-C in the hippocampus of PDE7a-nko mice, suggesting a novel mechanism of the cAMP regulation by PDE7a. Consequently, the decreased phosphorylation of CREB, CAMKII, eif2a, ERK, and AMPK, and reduced total level of NR2A have been found in the brain of PDE7a-nko animals. Notably, genetic excision of PDE7a in neurons was not able to change the expression of NR2B, BDNF, synapsin1, synaptophysin, or snap25. CONCLUSION: Altogether, our current findings demonstrated, for the first time, the role of PDE7a in cognitive processes. Future studies will untangle PDE7a-dependent neurobiological and molecular-cellular mechanisms related to cAMP-associated disorders.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cyclic Nucleotide Phosphodiesterases, Type 7 , Memory, Short-Term , Spatial Learning , Animals , Mice , AMP-Activated Protein Kinases/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Fear , Hippocampus/metabolism , Mice, Inbred C57BL , Synaptophysin/metabolism , Memory , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism
3.
Inflammopharmacology ; 30(6): 2051-2061, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36272040

ABSTRACT

Neurodegenerative illness develops as a result of genetic defects that cause changes at numerous levels, including genomic products and biological processes. It entails the degradation of cyclic nucleotides, cyclic adenosine monophosphate (cAMP), and cyclic guanosine monophosphate (cGMP). PDE7 modulates intracellular cAMP signalling, which is involved in numerous essential physiological and pathological processes. For the therapy of neurodegenerative illnesses, the normalization of cyclic nucleotide signalling through PDE inhibition remains intriguing. In this article, we shall examine the role of PDEs in neurodegenerative diseases. Alzheimer's disease, Multiple sclerosis, Huntington's disease, Parkinson's disease, Stroke, and Epilepsy are related to alterations in PDE7 expression in the brain. Earlier, animal models of neurological illnesses including Alzheimer's disease, Parkinson's disease, and multiple sclerosis have had significant results to PDE7 inhibitors, i.e., VP3.15; VP1.14. In addition, modulation of CAMP/CREB/GSK/PKA signalling pathways involving PDE7 in neurodegenerative diseases has been addressed. To understand the etiology, treatment options of these disorders mediated by PDE7 and its subtypes can be the focus of future research.


Subject(s)
Alzheimer Disease , Multiple Sclerosis , Neurodegenerative Diseases , Parkinson Disease , Animals , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Multiple Sclerosis/drug therapy
4.
Neuropharmacology ; 196: 108694, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34245775

ABSTRACT

Phosphodiesterase 7 (PDE7), one of the 11 phosphodiesterase (PDE) families, specifically hydrolyzes cyclic 3', 5'-adenosine monophosphate (cAMP). PDE7 is involved in many important functional processes in physiology and pathology by regulating intracellular cAMP signaling. Studies have demonstrated that PDE7 is widely expressed in the central nervous system (CNS) and potentially related to pathogenesis of many CNS diseases. Here, we summarized the classification and distribution of PDE7 in the brain and its functional roles in the mediation of CNS diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and schizophrenia. It is expected that the findings collected here will not only lead to a better understanding of the mechanisms by which PDE7 mediates CNS function and diseases, but also aid in the development of novel drugs targeting PDE7 for treatment of CNS diseases.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Multiple Sclerosis/metabolism , Parkinson Disease/metabolism , Schizophrenia/metabolism , Animals , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Humans , Molecular Targeted Therapy , Multiple Sclerosis/drug therapy , Parkinson Disease/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Protein Isoforms
5.
Biosci Biotechnol Biochem ; 85(9): 1962-1970, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34077501

ABSTRACT

Circular RNA plays an important role in the progression of sepsis. Circ_0091702 has been found to be an important regulator of sepsis progression, so its role and mechanism in sepsis progression deserve to be further explored. Lipopolysaccharide (LPS) could suppress cell viability, while enhance cell apoptosis and inflammation to induce cell injury. Circ_0091702 was downregulated in LPS-induced HK2 cells, and its overexpression alleviated LPS-induced cell injury. MiR-182 could be sponged by circ_0091702. Moreover, miR-182 inhibitor could relieve LPS-induced cell injury, and its overexpression also reversed the inhibition of circ_0091702 on LPS-induced cell injury. PDE7A was a target of miR-182, and its expression was reduced in LPS-induced HK2 cells. Additionally, silencing of PDE7A reversed the suppressive effect of circ_0091702 on LPS-induced cell injury. Our data suggested that circ_0091702 sponged miR-182 to regulate PDE7A, thereby alleviating LPS-induced cell injury in sepsis.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/metabolism , RNA, Circular/physiology , Cell Line , Computational Biology/methods , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Gene Knockdown Techniques , Humans
6.
Int J Mol Sci ; 21(17)2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32854348

ABSTRACT

Phosphodiesterase 7 (PDE7), a cAMP-specific PDE family, insensitive to rolipram, is present in many immune cells, including T lymphocytes. Two genes of PDE7 have been identified: PDE7A and PDE7B with three or four splice variants, respectively. Both PDE7A and PDE7B are expressed in T cells, and the predominant splice variant in these cells is PDE7A1. PDE7 is one of several PDE families that terminates biological functions of cAMP-a major regulating intracellular factor. However, the precise role of PDE7 in T cell activation and function is still ambiguous. Some authors reported its crucial role in T cell activation, while according to other studies PDE7 activity was not pivotal to T cells. Several studies showed that inhibition of PDE7 by its selective or dual PDE4/7 inhibitors suppresses T cell activity, and consequently T-mediated immune response. Taken together, it seems quite likely that simultaneous inhibition of PDE4 and PDE7 by dual PDE4/7 inhibitors or a combination of selective PDE4 and PDE7 remains the most interesting therapeutic target for the treatment of some immune-related disorders, such as autoimmune diseases, or selected respiratory diseases. An interesting direction of future studies could also be using a combination of selective PDE7 and PDE3 inhibitors.


Subject(s)
Autoimmune Diseases/immunology , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Phosphodiesterase Inhibitors/pharmacology , Respiratory Tract Diseases/immunology , T-Lymphocytes/metabolism , Alternative Splicing , Animals , Autoimmune Diseases/drug therapy , Cell Division , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Humans , Lymphocyte Activation , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Respiratory Tract Diseases/drug therapy , T-Lymphocytes/drug effects
7.
Eur J Med Chem ; 201: 112437, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32673902

ABSTRACT

A library of novel anilide and benzylamide derivatives of ω-(4-(2-methoxyphenyl)piperazin-1-yl)alkanoic acids as combined 5-HT1A/5-HT7 receptor ligands and phosphodiesterase PDE4B/PDE7A inhibitors was designed using a structure-based drug design approach. The in vitro studies of 33 newly synthesized compounds (7-39) allowed us to identify 22 as the most promising multifunctional 5-HT1A/5-HT7 receptor antagonist (5-HT1AKi = 8 nM, Kb = 0.04 nM; 5-HT7Ki = 451 nM, Kb = 460 nM) with PDE4B/PDE7A inhibitory activity (PDE4B IC50 = 80.4 µM; PDE7A IC50 = 151.3 µM). Compound 22 exerted a very good ability to passively penetrate through biological membranes and a high metabolic stability in vitro. Moreover, the pharmacological evaluation of 22 showed its procognitive and antidepressant properties in rat behavioral tests. Compound 22 at a dose of 3 mg/kg (i.p.) significantly reversed MK-801-induced episodic memory deficits in the novel object recognition test, while at a dose of 10 mg/kg (i.p.) reduced the immobility time of animals (by about 34%) in the forced swimming test. The antidepressant-like effect produced by compound 22 was stronger than that of escitalopram used as a reference drug. This study opens a new perspective in the search for efficacious drugs for the treatment of cognitive and depressive disorders.


Subject(s)
Anilides/pharmacology , Central Nervous System Agents/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Piperazines/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Anilides/chemical synthesis , Anilides/metabolism , Animals , CHO Cells , Central Nervous System Agents/chemical synthesis , Central Nervous System Agents/metabolism , Cricetulus , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , HEK293 Cells , Humans , Male , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Open Field Test/drug effects , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/metabolism , Piperazines/chemical synthesis , Piperazines/metabolism , Protein Binding , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin/metabolism , Serotonin 5-HT1 Receptor Antagonists/chemical synthesis , Serotonin 5-HT1 Receptor Antagonists/metabolism , Sf9 Cells , Structure-Activity Relationship
8.
Int J Mol Sci ; 21(11)2020 Jun 03.
Article in English | MEDLINE | ID: mdl-32503342

ABSTRACT

Phosphodiesterase (PDE) inhibitors are currently a widespread and extensively studied group of anti-inflammatory and anti-fibrotic compounds which may find use in the treatment of numerous lung diseases, including asthma and chronic obstructive pulmonary disease. Several PDE inhibitors are currently in clinical development, and some of them, e.g., roflumilast, are already recommended for clinical use. Due to numerous reports indicating that elevated intracellular cAMP levels may contribute to the alleviation of inflammation and airway fibrosis, new and effective PDE inhibitors are constantly being sought. Recently, a group of 7,8-disubstituted purine-2,6-dione derivatives, representing a novel and prominent pan-PDE inhibitors has been synthesized. Some of them were reported to modulate transient receptor potential ankyrin 1 (TRPA1) ion channels as well. In this study, we investigated the effect of selected derivatives (832-a pan-PDE inhibitor, 869-a TRPA1 modulator, and 145-a pan-PDE inhibitor and a weak TRPA1 modulator) on cellular responses related to airway remodeling using MRC-5 human lung fibroblasts. Compound 145 exerted the most considerable effect in limiting fibroblast to myofibroblasts transition (FMT) as well as proliferation, migration, and contraction. The effect of this compound appeared to depend mainly on its strong PDE inhibitory properties, and not on its effects on TRPA1 modulation. The strong anti-remodeling effects of 145 required activation of the cAMP/protein kinase A (PKA)/cAMP response element-binding protein (CREB) pathway leading to inhibition of transforming growth factor type ß1 (TGF-ß1) and Smad-dependent signaling in MRC-5 cells. These data suggest that the TGF-ß pathway is a major target for PDE inhibitors leading to inhibitory effects on cell responses involved in airway remodeling. These potent, pan-PDE inhibitors from the group of 7,8-disubstituted purine-2,6-dione derivatives, thus represent promising anti-remodeling drug candidates for further research.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Fibroblasts/drug effects , Lung/drug effects , Phosphodiesterase Inhibitors/pharmacology , Transforming Growth Factor beta1/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Calcium/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Drug Design , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Fibrosis , Humans , Lung/metabolism , Myofibroblasts/metabolism , Signal Transduction , TRPA1 Cation Channel/metabolism
9.
PLoS One ; 15(1): e0227279, 2020.
Article in English | MEDLINE | ID: mdl-31999703

ABSTRACT

Fibrous dysplasia (FD) of bone is a complex disease of the skeleton caused by dominant activating mutations of the GNAS locus encoding for the α subunit of the G protein-coupled receptor complex (Gsα). The mutation involves a substitution of arginine at position 201 by histidine or cysteine (GsαR201H or R201C), which leads to overproduction of cAMP. Several signaling pathways are implicated downstream of excess cAMP in the manifestation of disease. However, the pathogenesis of FD remains largely unknown. The overall FD phenotype can be attributed to alterations of skeletal stem/progenitor cells which normally develop into osteogenic or adipogenic cells (in cis), and are also known to provide support to angiogenesis, hematopoiesis, and osteoclastogenesis (in trans). In order to dissect the molecular pathways rooted in skeletal stem/progenitor cells by FD mutations, we engineered human skeletal stem/progenitor cells with the GsαR201C mutation and performed transcriptomic analysis. Our data suggest that this FD mutation profoundly alters the properties of skeletal stem/progenitor cells by pushing them towards formation of disorganized bone with a concomitant alteration of adipogenic differentiation. In addition, the mutation creates an altered in trans environment that induces neovascularization, cytokine/chemokine changes and osteoclastogenesis. In silico comparison of our data with the signature of FD craniofacial samples highlighted common traits, such as the upregulation of ADAM (A Disintegrin and Metalloprotease) proteins and other matrix-related factors, and of PDE7B (Phosphodiesterase 7B), which can be considered as a buffering process, activated to compensate for excess cAMP. We also observed high levels of CEBPs (CCAAT-Enhancer Binding Proteins) in both data sets, factors related to browning of white fat. This is the first analysis of the reaction of human skeletal stem/progenitor cells to the introduction of the FD mutation and we believe it provides a useful background for further studies on the molecular basis of the disease and for the identification of novel potential therapeutic targets.


Subject(s)
Bone Marrow Cells/physiology , Cell Differentiation/genetics , Chromogranins/genetics , Fibrous Dysplasia of Bone/pathology , GTP-Binding Protein alpha Subunits, Gs/genetics , Stem Cells/physiology , ADAM Proteins/metabolism , Adipogenesis/genetics , Adipose Tissue, White/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Chromogranins/metabolism , Computer Simulation , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Datasets as Topic , Fibrous Dysplasia of Bone/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gain of Function Mutation , Gene Expression Profiling , Healthy Volunteers , Humans , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Osteogenesis/genetics , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Stromal Cells/physiology , Up-Regulation
10.
Mol Neurobiol ; 57(2): 806-822, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31473904

ABSTRACT

Parkinson's disease is characterized by a loss of dopaminergic neurons in the ventral midbrain. This disease is diagnosed when around 50% of these neurons have already died; consequently, therapeutic treatments start too late. Therefore, an urgent need exists to find new targets involved in the onset and progression of the disease. Phosphodiesterase 7 (PDE7) is a key enzyme involved in the degradation of intracellular levels of cyclic adenosine 3', 5'-monophosphate in different cell types; however, little is known regarding its role in neurodegenerative diseases, and specifically in Parkinson's disease. We have previously shown that chemical as well as genetic inhibition of this enzyme results in neuroprotection and anti-inflammatory activity in different models of neurodegenerative disorders, including Parkinson's disease. Here, we have used in vitro and in vivo models of Parkinson's disease to study the regulation of PDE7 protein levels. Our results show that PDE7 is upregulated after an injury both in the human dopaminergic cell line SH-SY5Y and in primary rat mesencephalic cultures and after lipopolysaccharide or 6-hidroxydopamine injection in the Substantia nigra pars compacta of adult mice. PDE7 increase takes place mainly in degenerating dopaminergic neurons and in microglia cells. This enhanced expression appears to be direct since 6-hydroxydopamine and lipopolysaccharide increase the expression of a 962-bp fragment of its promoter. Taking together, these results reveal an essential function for PDE7 in the pathways leading to neurodegeneration and inflammatory-mediated brain damage and suggest novel roles for PDE7 in neurodegenerative diseases, specifically in PD, opening the door for new therapeutic interventions.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Parkinson Disease/enzymology , Parkinson Disease/pathology , Animals , Apoptosis , Cell Line , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Disease Models, Animal , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Embryo, Mammalian/enzymology , Humans , Male , Mesencephalon/enzymology , Mesencephalon/pathology , Neuroglia/enzymology , Neuroglia/pathology , Oxidopamine , Promoter Regions, Genetic/genetics , Rats, Wistar , Substantia Nigra/enzymology , Substantia Nigra/pathology
11.
Arch Pharm (Weinheim) ; 353(1): e1900211, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31696968

ABSTRACT

N-Substituted isatoic anhydrides were used as starting materials for the synthesis of compounds 5-16 through alkali hydrolysis, Schiff base reactions, and oxidation. Compounds 18-23 were obtained by thionation of their oxo isosteres using Lawesson's reagent. Cyclocondesation of anthranilic acid with thiourea afforded compounds 25-27, which were S-alkylated to afford compounds 28-30, which were thionated using Lawesson's reagent to afford 31-33. The compounds were tested for their in vitro inhibitory activity against the phosphodiesterase 7A (PDE7A) enzyme compared with the selective PDE7 inhibitor BRL50481. All the compounds showed the inhibitory activity on the enzyme at micromolar levels. Compounds 9 and 25 showed the highest inhibitory activity on the enzyme: IC50 = 0.096 and 0.074 µM, respectively, comparable to BRL50481 (IC50 = 0.072 µM). The binding mode and binding affinity of the target compounds at the enzyme PDE7A-binding site were studied through molecular docking. Compounds 9 and 25 showed good recognition at the enzyme-binding site and were capable of binding in an inhibitory mode similar to the reference compound BRL50481, forming the necessary interactions with the key amino acids. Docking studies and enzyme assay were in agreement.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Quinazolines/pharmacology , Quinazolinones/pharmacology , Thiones/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Quinazolines/chemical synthesis , Quinazolines/chemistry , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Structure-Activity Relationship , Thiones/chemical synthesis , Thiones/chemistry
12.
Bull Exp Biol Med ; 167(4): 467-469, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31493257

ABSTRACT

Second messengers cAMP and cGMP play an important role in synaptic plasticity and memory consolidation. The inhibitors of phosphodiesterases, enzymes hydrolyzing these cyclic nucleotides, are actively studied as potential drugs for the treatment of various cognitive disorders and depression. We studied the effects of a new inhibitor of phosphodiesterase 7 AGF2.20 on the formation of long-term potentiation in hippocampal slices. Administration of AGF2.20 (10 nM) in 90 min after weak tetanization prevented a decrease in the amplitude of excitatory post-synaptic potentials and stabilized long-term potentiation. These data attest to the involvement of phosphodiesterase 7 in the development of synaptic plasticity in the hippocampus. The inhibitor AGF2.20 is considered for the further analysis as a promising substance for the treatment of cognitive impairments.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Enzyme Inhibitors/pharmacology , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/drug effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects
13.
Proc Natl Acad Sci U S A ; 115(52): E12265-E12274, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30541888

ABSTRACT

Adrenal cortex steroids are essential for body homeostasis, and adrenal insufficiency is a life-threatening condition. Adrenal endocrine activity is maintained through recruitment of subcapsular progenitor cells that follow a unidirectional differentiation path from zona glomerulosa to zona fasciculata (zF). Here, we show that this unidirectionality is ensured by the histone methyltransferase EZH2. Indeed, we demonstrate that EZH2 maintains adrenal steroidogenic cell differentiation by preventing expression of GATA4 and WT1 that cause abnormal dedifferentiation to a progenitor-like state in Ezh2 KO adrenals. EZH2 further ensures normal cortical differentiation by programming cells for optimal response to adrenocorticotrophic hormone (ACTH)/PKA signaling. This is achieved by repression of phosphodiesterases PDE1B, 3A, and 7A and of PRKAR1B. Consequently, EZH2 ablation results in blunted zF differentiation and primary glucocorticoid insufficiency. These data demonstrate an all-encompassing role for EZH2 in programming steroidogenic cells for optimal response to differentiation signals and in maintaining their differentiated state.


Subject(s)
Adrenal Cortex/enzymology , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Signal Transduction , Adrenal Cortex/metabolism , Animals , Cell Differentiation , Cyclic AMP-Dependent Protein Kinase RIbeta Subunit/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/genetics , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Steroids/metabolism , Zona Fasciculata/cytology , Zona Fasciculata/enzymology , Zona Fasciculata/metabolism , Zona Glomerulosa/cytology , Zona Glomerulosa/enzymology , Zona Glomerulosa/metabolism
14.
Eur J Med Chem ; 158: 517-533, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30245393

ABSTRACT

A series of novel amide derivatives of 1,3-dimethyl-2,6-dioxopurin-7-yl-alkylcarboxylic acids designed using a structure-based computational approach was synthesized and assayed to evaluate their ability to block human TRPA1 channel and inhibit PDE4B/7A activity. We identified compounds 16 and 27 which showed higher potency against TRPA1 compared to HC-030031. In turn, compound 36 was the most promising multifunctional TRPA1 antagonist and PDE4B/7A dual inhibitor with IC50 values in the range of that of the reference rolipram and BRL-50481, respectively. Compound 36 as a combined TRPA1/PDE4B/PDE7A ligand was characterized by a distinct binding mode in comparison to 16 and 27, in the given protein targets. The inhibition of both cAMP-specific PDE isoenzymes resulted in a strong anti-TNF-α effect of 36in vivo. Moreover, the potent anti-inflammatory and analgesic efficacy of 36 was observed in animal models of pain and inflammation (formalin test in mice and carrageenan-induced paw edema in rats). This compound also displayed significant antiallodynic properties in the early phase of chemotherapy-induced peripheral neuropathy in mice. In turn, the pure TRPA1 antagonists 16 and 27 revealed a statistically significant antiallodynic effect in the formalin test and in the von Frey test performed in both phases of oxaliplatin-induced allodynia. Antiallodynic activity of the test compounds 16, 27 and 36 was observed at a dose range comparable to that of the reference drug - pregabalin. In conclusion, the proposed approach of pain treatment based on the concomitant blocking of TRPA1 channel and PDE4B/7A inhibitory activity appears to be interesting research direction for the future search for novel analgesics.


Subject(s)
Analgesics/chemistry , Analgesics/therapeutic use , Pain/drug therapy , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/therapeutic use , TRPA1 Cation Channel/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Amides/therapeutic use , Analgesics/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Mice , Molecular Docking Simulation , Pain/metabolism , Pain Measurement/drug effects , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/metabolism , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase 4 Inhibitors/therapeutic use , Phosphodiesterase Inhibitors/pharmacology , Rats, Wistar , TRPA1 Cation Channel/metabolism
15.
Endocrinology ; 159(5): 2142-2152, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29608743

ABSTRACT

Luteinizing hormone (LH) acts on the granulosa cells that surround the oocyte in mammalian preovulatory follicles to cause meiotic resumption and ovulation. Both of these responses are mediated primarily by an increase in cyclic adenosine monophosphate (cAMP) in the granulosa cells, and the activity of cAMP phosphodiesterases (PDEs), including PDE4, contributes to preventing premature responses. However, two other cAMP-specific PDEs, PDE7 and PDE8, are also expressed at high levels in the granulosa cells, raising the question of whether these PDEs also contribute to preventing uncontrolled activation of meiotic resumption and ovulation. With the use of selective inhibitors, we show that inhibition of PDE7 or PDE8 alone has no effect on the cAMP content of follicles, and inhibition of PDE4 alone has only a small and variable effect. In contrast, a mixture of the three inhibitors elevates cAMP to a level comparable with that seen with LH. Correspondingly, inhibition of PDE7 or PDE8 alone has no effect on meiotic resumption or ovulation, and inhibition of PDE4 alone has only a partial and slow effect. However, the fraction of oocytes resuming meiosis and undergoing ovulation is increased when PDE4, PDE7, and PDE8 are simultaneously inhibited. PDE4, PDE7, and PDE8 also function together to suppress the premature synthesis of progesterone and progesterone receptors, which are required for ovulation. Our results indicate that three cAMP PDEs act in concert to suppress premature responses in preovulatory follicles.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Meiosis/physiology , Oocytes/metabolism , Ovulation/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Animals , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Female , Meiosis/drug effects , Mice , Oocytes/drug effects , Ovulation/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Rolipram/pharmacology
16.
Eur J Med Chem ; 146: 381-394, 2018 Feb 25.
Article in English | MEDLINE | ID: mdl-29407965

ABSTRACT

A novel butanehydrazide derivatives of purine-2,6-dione designed using a ligand-based approach were synthesized and their in vitro activity against both PDE4B and PDE7A isoenzymes was assessed. The 7,8-disubstituted purine-2,6-dione derivatives 31, 34, 37, and 40 appeared to be the most potent PDE4/7 inhibitors with IC50 values in the range of that of the reference rolipram and BRL-50481, respectively. Moreover, docking studies explained the importance of N-(2,3,4-trihydroxybenzylidene)butanehydrazide substituent in position 7 of purine-2,6-dione core for dual PDE4/7 inhibitory properties. The inhibition of both the cAMP-specific PDE isoenzymes resulted in a strong anti-TNF-α effect. Compounds 31, 34, and 37 in the in vivo study in rats with LPS-induced endotoxemia decreased the maximum concentration of this proinflammatory cytokine by 53, 84 and 88%, respectively.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Butanes/pharmacology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Drug Design , Hydrazines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Purinones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Butanes/analysis , Butanes/chemical synthesis , Butanes/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Dose-Response Relationship, Drug , Endotoxemia/drug therapy , Humans , Hydrazines/chemical synthesis , Hydrazines/chemistry , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Male , Molecular Structure , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Purinones/chemical synthesis , Purinones/chemistry , Rats , Rats, Wistar , Structure-Activity Relationship
17.
Alzheimers Res Ther ; 10(1): 24, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458418

ABSTRACT

BACKGROUND: The phosphodiesterase (PDE) 7 inhibitor S14 is a cell-permeable small heterocyclic molecule that is able to cross the blood-brain barrier. We previously found that intraperitoneal treatment with S14 exerted neuroprotection in an Alzheimer's disease (AD) model (in APP/PS1 mice). The objective of this study was to investigate the neurogenic and cellular effects of oral administration of S14 on amyloid ß (Aß) overload. METHODS: We orally administered the PDE7 inhibitor S14 (15 mg/kg/day) or vehicle in 6-month-old APP/PS1 mice. After 5 weeks of S14 treatment, we evaluated cognitive functions and brain tissues. We also assessed the effects of S14 on the Aß-treated human neuroblastome SH-SY5Y cell line. RESULTS: Targeting the cyclic adenosine monophosphate (cAMP)/cAMP-response element binding protein (CREB) pathway, S14 rescued cognitive decline by improving hippocampal neurogenesis in APP/PS1 transgenic mice. Additionally, S14 treatment reverted the Aß-induced reduction in mitochondrial mass in APP/PS1 mice and in the human neuroblastoma SH-SY5Y cells co-exposed to Aß. The restoration of the mitochondrial mass was found to be a dual effect of S14: a rescue of the mitochondrial biogenesis formerly slowed down by Aß overload, and a reduction in the Aß-increased mitochondrial clearance mechanism of mitophagy. CONCLUSIONS: Here, we show new therapeutic effects of the PDE7 inhibitor, confirming S14 as a potential therapeutic drug for AD.


Subject(s)
Alzheimer Disease/complications , Alzheimer Disease/enzymology , Hippocampus/drug effects , Mitochondrial Dynamics/drug effects , Neurogenesis/drug effects , Phosphodiesterase Inhibitors/pharmacology , Quinazolines/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line, Tumor , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Disease Models, Animal , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/pathology , Humans , Learning Disabilities/drug therapy , Learning Disabilities/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Dynamics/genetics , Neurogenesis/genetics , Phosphodiesterase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Stem Cells ; 35(2): 458-472, 2017 02.
Article in English | MEDLINE | ID: mdl-27538853

ABSTRACT

The phosphodiesterase 7 (PDE7) enzyme is one of the enzymes responsible for controlling intracellular levels of cyclic adenosine 3',5'-monophosphate in the immune and central nervous system. We have previously shown that inhibitors of this enzyme are potent neuroprotective and anti-inflammatory agents. In addition, we also demonstrated that PDE7 inhibition induces endogenous neuroregenerative processes toward a dopaminergic phenotype. Here, we show that PDE7 inhibition controls stem cell expansion in the subgranular zone of the dentate gyrus of the hippocampus (SGZ) and the subventricular zone (SVZ) in the adult rat brain. Neurospheres cultures obtained from SGZ and SVZ of adult rats treated with PDE7 inhibitors presented an increased proliferation and neuronal differentiation compared to control cultures. PDE7 inhibitors treatment of neurospheres cultures also resulted in an increase of the levels of phosphorylated cAMP response element binding protein, suggesting that their effects were indeed mediated through the activation of the cAMP/PKA signaling pathway. In addition, adult rats orally treated with S14, a specific inhibitor of PDE7, presented elevated numbers of proliferating progenitor cells, and migrating precursors in the SGZ and the SVZ. Moreover, long-term treatment with this PDE7 inhibitor shows a significant increase in newly generated neurons in the olfactory bulb and the hippocampus. Also a better performance in memory tests was observed in S14 treated rats, suggesting a functional relevance for the S14-induced increase in SGZ neurogenesis. Taken together, our results indicate for the first time that inhibition of PDE7 directly regulates proliferation, migration and differentiation of neural stem cells, improving spatial learning and memory tasks. Stem Cells 2017;35:458-472.


Subject(s)
Aging/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Hippocampus/enzymology , Hippocampus/growth & development , Lateral Ventricles/enzymology , Lateral Ventricles/growth & development , Neurogenesis , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Dentate Gyrus/cytology , Hippocampus/drug effects , Lateral Ventricles/drug effects , Male , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phenotype , Phosphodiesterase Inhibitors/pharmacology , Rats, Wistar , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
19.
Br J Pharmacol ; 172(17): 4277-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25994655

ABSTRACT

BACKGROUND AND PURPOSE: cAMP plays an important role in the transduction of signalling pathways involved in neuroprotection and immune regulation. Control of the levels of this nucleotide by inhibition of cAMP-specific PDEs such as PDE7 may affect the pathological processes of neuroinflammatory diseases like multiple sclerosis (MS). In the present study, we evaluated the therapeutic potential of the selective PDE7 inhibitor, TC3.6, in a model of primary progressive multiple sclerosis (PPMS), a rare and severe variant of MS. EXPERIMENTAL APPROACH: Theiler's murine encephalomyelitis virus-induced demyelinated disease (TMEV-IDD) is one of the models used to validate the therapeutic efficacy of new drugs in MS. As recent studies have analysed the effect of PDE7 inhibitors in the EAE model of MS, here the TMEV-IDD model was used to test their efficacy in a progressive variant of MS. Mice were subjected to two protocols of TC3.6 administration: on the pre-symptomatic phase and once the disease was established. KEY RESULTS: Treatment with TC3.6 ameliorated the disease course and improved motor deficits of infected mice. This was associated with down-regulation of microglial activation and reduced cellular infiltrates. Decreased expression of pro-inflammatory mediators such as COX-2 and the cytokines, IL-1ß, TNF-α, IFN-γ and IL-6 in the spinal cord of TMEV-infected mice was also observed after TC3.6 administration. CONCLUSION: These findings support the importance of PDE7 inhibitors, and specifically TC3.6, as a novel class of agents with therapeutic potential for PPMS. Preclinical studies are needed to determine whether their effects translate into durable clinical benefits.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Disease Models, Animal , Multiple Sclerosis, Chronic Progressive/drug therapy , Phosphodiesterase Inhibitors/therapeutic use , Animals , Animals, Newborn , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Female , Mice , Multiple Sclerosis, Chronic Progressive/enzymology , Multiple Sclerosis, Chronic Progressive/pathology , PC12 Cells , Phosphodiesterase Inhibitors/pharmacology , Rats
20.
Stem Cells Transl Med ; 4(6): 564-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25925836

ABSTRACT

UNLABELLED: Parkinson's disease is characterized by a loss of dopaminergic neurons in a specific brain region, the ventral midbrain. Parkinson's disease is diagnosed when approximately 50% of the dopaminergic neurons of the substantia nigra pars compacta (SNpc) have degenerated and the others are already affected by the disease. Thus, it is conceivable that all therapeutic strategies, aimed at neuroprotection, start too late. Therefore, an urgent medical need exists to discover new pharmacological targets and novel drugs with disease-modifying properties. In this regard, modulation of endogenous adult neurogenesis toward a dopaminergic phenotype might provide a new strategy to target Parkinson's disease by partially ameliorating the dopaminergic cell loss that occurs in this disorder. We have previously shown that a phosphodiesterase 7 (PDE7) inhibitor, S14, exerts potent neuroprotective and anti-inflammatory effects in different rodent models of Parkinson's disease, indicating that this compound could represent a novel therapeutic agent to stop the dopaminergic cell loss that occurs during the progression of the disease. In this report we show that, in addition to its neuroprotective effect, the PDE7 inhibitor S14 is also able to induce endogenous neuroregenerative processes toward a dopaminergic phenotype. We describe a population of actively dividing cells that give rise to new neurons in the SNpc of hemiparkinsonian rats after treatment with S14. In conclusion, our data identify S14 as a novel regulator of dopaminergic neuron generation. SIGNIFICANCE: Parkinson's disease is a neurodegenerative disorder characterized by the loss of dopaminergic neurons in the ventral midbrain. Currently, no cure and no effective disease-modifying therapy are available for Parkinson's disease; therefore, an urgent medical need exists to discover new pharmacological targets and novel drugs for the treatment of this disorder. The present study reports that an inhibitor of the enzyme phosphodiesterase 7 (S14) induces proliferation in vitro and in vivo of neural stem cells, promoting its differentiation toward a dopaminergic phenotype and therefore enhancing dopaminergic neuron generation. Because this drug is also able to confer neuroprotection of these cells in animal models of Parkinson's disease, S14 holds great promise as a therapeutic new strategy for this disorder.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Dopaminergic Neurons/enzymology , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Animals , Cyclic Nucleotide Phosphodiesterases, Type 7/metabolism , Dopaminergic Neurons/pathology , Male , Parkinsonian Disorders/enzymology , Parkinsonian Disorders/pathology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...