Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Comput Biol Med ; 159: 106869, 2023 06.
Article in English | MEDLINE | ID: mdl-37071939

ABSTRACT

In recent years, the PDE1B enzyme has become a desirable drug target for the treatment of psychological and neurological disorders, particularly schizophrenia disorder, due to the expression of PDE1B in brain regions involved in volitional behaviour, learning and memory. Although several inhibitors of PDE1 have been identified using different methods, none of these inhibitors has reached the market yet. Thus, searching for novel PDE1B inhibitors is considered a major scientific challenge. In this study, pharmacophore-based screening, ensemble docking and molecular dynamics simulations have been performed to identify a lead inhibitor of PDE1B with a new chemical scaffold. Five PDE1B crystal structures have been utilised in the docking study to improve the possibility of identifying an active compound compared to the use of a single crystal structure. Finally, the structure-activity- relationship was studied, and the structure of the lead molecule was modified to design novel inhibitors with a high affinity for PDE1B. As a result, two novel compounds have been designed that exhibited a higher affinity to PDE1B compared to the lead compound and the other designed compounds.


Subject(s)
Molecular Dynamics Simulation , Pharmacophore , Molecular Docking Simulation , Quantitative Structure-Activity Relationship , Structure-Activity Relationship , Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors
2.
PLoS One ; 17(12): e0278216, 2022.
Article in English | MEDLINE | ID: mdl-36454774

ABSTRACT

Phosphodiesterase 1B (PDE1B) and PDE10A are dual-specificity PDEs that hydrolyse both cyclic adenosine monophosphate and cyclic guanosine monophosphate, and are highly expressed in the striatum. Several reports have suggested that PDE10A inhibitors may present a promising approach for the treatment of positive symptoms of schizophrenia, whereas PDE1B inhibitors may present a novel mechanism to modulate cognitive deficits. Previously, we have reported a novel dual inhibitor of PDE1B and PDE10A, compound 2 [(3-fluorophenyl)(2-methyl-2,3-dihydro-4H-benzo[b][1,4]oxazin-4-yl)methanone] which has shown inhibitory activity for human recombinant PDE1B and PDE10A in vitro. In the present study, the safety profile of compound 2 has been evaluated in rats in the acute oral toxicity study, as well as; the antipsychotic-like effects in the rat model of schizophrenia. Compound 2 was tolerated up to 1 g/kg when administered at a single oral dose. Additionally, compound 2 has strongly suppressed ketamine-induced hyperlocomotion, which presented a model for the positive symptoms of schizophrenia. It has also shown an ability to attenuate social isolation induced by chronic administration of ketamine and enhanced recognition memory of rats ​in the novel object recognition test. Altogether, our results suggest that compound 2 represents a promising therapy for the treatment of the three symptomatic domains of schizophrenia.


Subject(s)
Antipsychotic Agents , Cognition Disorders , Ketamine , Schizophrenia , Humans , Animals , Rats , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Schizophrenia/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Phosphoric Diester Hydrolases
3.
Curr Top Med Chem ; 22(26): 2190-2206, 2022.
Article in English | MEDLINE | ID: mdl-36278463

ABSTRACT

Over the last two decades, computational technologies have played a crucial role in antiviral drug development. Whenever a virus spreads and becomes a threat to global health, it brings along the challenge of developing new therapeutics and prophylactics. Computational drug and vaccine discovery has evolved quickly over the years. Some interesting examples of computational drug discovery are anti-AIDS drugs, where HIV protease and reverse transcriptase have been targeted by agents developed using computational methods. Various computational methods that have been applied to anti-viral research include ligand-based methods that rely on known active compounds, i.e., pharmacophore modeling, machine learning or classical QSAR; structure-based methods that rely on an experimentally determined 3D structure of the targets, i.e., molecular docking and molecular dynamics and methods for the development of vaccines such as reverse vaccinology; structural vaccinology and vaccine epitope prediction. This review summarizes these approaches to battle viral diseases and underscores their importance for anti-viral research. We discuss the role of computational methods in developing small molecules and vaccines against human immunodeficiency virus, yellow fever, human papilloma virus, SARS-CoV-2, and other viruses. Various computational tools available for the abovementioned purposes have been listed and described. A discussion on applying artificial intelligence-based methods for antiviral drug discovery has also been included.


Subject(s)
COVID-19 , Vaccines , Humans , Molecular Docking Simulation , Artificial Intelligence , SARS-CoV-2 , COVID-19/prevention & control
4.
RSC Adv ; 12(3): 1576-1591, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35425186

ABSTRACT

Phosphodiesterase10A (PDE10A) is a potential therapeutic target for the treatment of several neurodegenerative disorders. Thus, extensive efforts of medicinal chemists have been directed toward developing potent PDE10A inhibitors with minimal side effects. However, PDE10A inhibitors are not approved as a treatment for neurodegenerative disorders, possibly due to the lack of research in this area. Therefore, the discovery of novel and diverse scaffolds targeting PDE10A is required. In this study, we described the identification of a new PDE10A inhibitor by structure-based virtual screening combining pharmacophore modelling, molecular docking, molecular dynamics simulations, and biological evaluation. Zinc42657360 with a cyclopenta[4,5]thieno[2,3-d]pyrimidin-4-one scaffold from the zinc database exhibited a significant inhibitory activity of 1.60 µM against PDE10A. The modelling studies demonstrated that Zinc42657360 is involved in three hydrogen bonds with ASN226, THR187 and ASP228, and two aromatic interactions with TYR78 and PHE283, besides the common interactions with the P-clamp residues PHE283 and ILE246. The novel scaffold of Zinc42657360 can be used for the rational design of PDE10A inhibitors with improved affinity.

5.
Braz. J. Pharm. Sci. (Online) ; 58: e191134, 2022. tab, graf
Article in English | LILACS | ID: biblio-1394031

ABSTRACT

Abstract Existing medications i.e. the antipsychotic drugs are known to be effective in treating only the positive symptoms of schizophrenia, while being ineffective on negative and cognitive symptoms of the disease. In addition, these medications cause extrapyramidal symptoms, forcing many patients towards natural medicine in the hope of minimizing the unwanted adverse effects. Nardostachys jatamansi is a medicinal plant that has been traditionally prescribed for various types of brain disorders. The active constituents of the plant have beneficial effects on the negative and cognitive symptoms of schizophrenia. This study was designed to identify the active constituents of Nardostachys jatamansi with the highest binding affinities for the key macromolecular drug targets involved in the pathophysiology of schizophrenia and thereby elucidate the possible mechanism of action. These targets are dopamine receptors, Gamma-aminobutyric acid receptors, N-methyl-D-aspartate receptors and Phosphodiesterase 10A. The results of molecular docking showed that, β-sitosterol, chlorogenic acid, oleanic acid and ursolic acid, displayed high binding affinity toward all the macromolecular drug targets. Ligands with steroid backbone and pentacyclic triterpene structure have been found to possess high binding affinity toward the dopamine receptor and phosphodiesterase 10A. While ligands with carbonyl group form stronger binding interactions with the N-methyl-D-aspartate receptor.


Subject(s)
Plants, Medicinal/adverse effects , Research/classification , Pharmaceutical Preparations/analysis , Valerianaceae/classification , Nardostachys/adverse effects , Schizophrenia , Antipsychotic Agents
6.
Heliyon ; 6(9): e04856, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32984588

ABSTRACT

Inhibition of phosphodiesterase 4 (PDE4) is a promising therapeutic approach for the treatment of inflammatory pulmonary disorders, i.e. asthma and chronic obstructive pulmonary disease. However, the treatment with non-selective PDE4 inhibitors is associated with side effects such as nausea and vomiting. Among the subtypes of PDE4 inhibited by these inhibitors, PDE4B is expressed in immune, inflammatory and airway smooth muscle cells, whereas, PDE4D is expressed in the area postrema and nucleus of the solitary tract. Thus, PDE4D inhibition is responsible for the emetic response. In this regard, a selective PDE4B inhibitor is expected to be a potential drug candidate for the treatment of inflammatory pulmonary disorders. Therefore, a shared feature pharmacophore model was developed and used as a query for the virtual screening of Maybridge and SPECS databases. A number of filters were applied to ensure only compounds with drug-like properties were selected. Accordingly, nine compounds have been identified as final hits, where HTS04529 showed the highest affinity and selectivity for PDE4B over PDE4D in molecular docking. The docked complexes of HTS04529 with PDE4B and PDE4D were subjected to molecular dynamics simulations for 100ns to assess their binding stability. The results showed that HTS04529 was bound tightly to PDE4B and formed a more stable complex with it than with PDE4D.

7.
Curr Top Med Chem ; 20(26): 2404-2421, 2020.
Article in English | MEDLINE | ID: mdl-32533817

ABSTRACT

Schizophrenia is a severe mental disorder that affects more than 1% of the population worldwide. Dopamine system dysfunction and alterations in glutamatergic neurotransmission are strongly implicated in the aetiology of schizophrenia. To date, antipsychotic drugs are the only available treatment for the symptoms of schizophrenia. These medications, which act as D2-receptor antagonist, adequately address the positive symptoms of the disease, but they fail to improve the negative symptoms and cognitive impairment. In schizophrenia, cognitive impairment is a core feature of the disorder. Therefore, the treatment of cognitive impairment and the other symptoms related to schizophrenia remains a significant unmet medical need. Currently, phosphodiesterases (PDEs) are considered the best drug target for the treatment of schizophrenia since many PDE subfamilies are abundant in the brain regions that are relevant to cognition. Thus, this review aims to illustrate the mechanism of PDEs in treating the symptoms of schizophrenia and summarises the encouraging results of PDE inhibitors as anti-schizophrenic drugs in preclinical and clinical studies.


Subject(s)
Antipsychotic Agents/chemistry , Cognition Disorders/drug therapy , Cognition/drug effects , Nootropic Agents/chemistry , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/metabolism , Schizophrenia/drug therapy , Antipsychotic Agents/pharmacology , Brain , Dopamine/metabolism , Excitatory Amino Acid Agents/chemistry , Excitatory Amino Acid Agents/pharmacology , Humans , Molecular Targeted Therapy , Nootropic Agents/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Signal Transduction , Structure-Activity Relationship , Synaptic Transmission/drug effects
8.
ACS Omega ; 4(5): 8767-8777, 2019 May 31.
Article in English | MEDLINE | ID: mdl-31459966

ABSTRACT

Several studies have shown that the mammalian target of rapamycin (mTOR) inhibitor; everolimus (EV) improves patient survival in several types of cancer. However, the meaningful efficacy of EV as a single agent for the treatment of colorectal cancer (CRC) has failed to be proven in multiple clinical trials. Combination therapy is one of the options that could increase the efficacy and decrease the toxicity of the anticancer therapy. This study revealed that the ß-cyclodextrin (ß-CD):FGF7 complex has the potential to improve the antiproliferative effect of EV by preventing FGF receptor activation and by enhancing EV cellular uptake and intracellular retention. Molecular docking techniques were used to investigate the possible interaction between EV, ß-CD, and FGF7. Molecular docking insights revealed that ß-CD and EV are capable to form a stable inclusion complex with FGF at the molecular level. The aqueous solubility of the inclusion complex was increased (3.1 ± 0.23 µM) when compared to the aqueous solubility of pure EV (1.7 ± 0.16 µM). In addition, the in vitro cytotoxic activity of a FGF7:ß-CD:EV complex on Caco-2 cell line was investigated using real-time xCELLigence technology. The FGF7:ß-CD:EV complex has induced apoptosis of Caco-2 cells and shown higher cytotoxic activity than the parent drug EV. With the multitargets effect of ß-CD:FGF7 and EV, the antiproliferative effect of EV was remarkably improved as the IC50 value of EV was reduced from 9.65 ± 1.42 to 1.87 ± 0.33 µM when compared to FGF7:ß-CD:EV complex activity. In conclusion, the findings advance the understanding of the biological combinational effects of the ß-CD:FGF7 complex and EV as an effective treatment to combat CRC.

9.
Curr Top Med Chem ; 19(7): 555-564, 2019.
Article in English | MEDLINE | ID: mdl-30931862

ABSTRACT

BACKGROUND: Phosphodiesterases (PDEs) are enzymes that play a key role in terminating cyclic nucleotides signalling by catalysing the hydrolysis of 3', 5'- cyclic adenosine monophosphate (cAMP) and/or 3', 5' cyclic guanosine monophosphate (cGMP), the second messengers within the cell that transport the signals produced by extracellular signalling molecules which are unable to get into the cells. However, PDEs are proteins which do not operate alone but in complexes that made up of a many proteins. OBJECTIVE: This review highlights some of the general characteristics of PDEs and focuses mainly on the Protein-Protein Interactions (PPIs) of selected PDE enzymes. The objective is to review the role of PPIs in the specific mechanism for activation and thereby regulation of certain biological functions of PDEs. METHODS: The article discusses some of the PPIs of selected PDEs as reported in recent scientific literature. These interactions are critical for understanding the biological role of the target PDE. RESULTS: The PPIs have shown that each PDE has a specific mechanism for activation and thereby regulation a certain biological function. CONCLUSION: Targeting of PDEs to specific regions of the cell is based on the interaction with other proteins where each PDE enzyme binds with specific protein(s) via PPIs.


Subject(s)
Drug Delivery Systems , Gene Expression Regulation, Enzymologic/physiology , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Protein Interaction Domains and Motifs/physiology , Protein Binding
10.
Comput Biol Chem ; 77: 52-63, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30240986

ABSTRACT

The major complaint that most of the schizophrenic patients' face is the cognitive impairment which affects the patient's quality of life. The current antipsychotic drugs treat only the positive symptoms without alleviating the negative or cognitive symptoms of the disease. In addition, the existing therapies are known to produce extrapyramidal side effects that affect the patient adherence to the treatment. PDE10A inhibitor is the new therapeutic approach which has been proven to be effective in alleviating the negative and cognitive symptoms of the disease. A number of PDE10A inhibitors have been developed, but no inhibitor has made it beyond the clinical trials so far. Thus, the present study has been conducted to identify a PDE10A inhibitor from natural sources to be used as a lead compound for the designing of novel selective PDE10A inhibitors. Ligand and structure-based pharmacophore models for PDE10A inhibitors were generated and employed for virtual screening of universal natural products database. From the virtual screening results, 37 compounds were docked into the active site of the PDE10A. Out of 37 compounds, three inhibitors showed the highest affinity for PDE10A where UNPD216549 showed the lowest binding energy and has been chosen as starting point for designing of novel PDE10A inhibitors. The structure-activity-relationship studies assisted in designing of selective PDE10A inhibitors. The optimization of the substituents on the phenyl ring resulted in 26 derivatives with lower binding energy with PDE10A as compared to the lead compound. Among these, MA 8 and MA 98 exhibited the highest affinity for PDE10A with binding energy (-10.90 Kcal/mol).


Subject(s)
Antipsychotic Agents/pharmacology , Biological Products/pharmacology , Drug Discovery , Drug Evaluation, Preclinical , Phosphoric Diester Hydrolases/metabolism , Schizophrenia/drug therapy , Antipsychotic Agents/chemical synthesis , Antipsychotic Agents/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Schizophrenia/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...