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










Publication year range
1.
Eur J Med Chem ; 225: 113753, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34388383

ABSTRACT

The human transactive responsive (TAR) DNA-binding protein 43 (TDP-43) is involved in a number of physiological processes in the body. Its primary function involves RNA regulation. The TDP-43 protein is also involved in many diseases such as amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease (AD), Parkinson's disease (PD) and even cancers. These TDP-43 mediated diseases are collectively called as TDP-43 proteinopathies. Intense research in the last decade has increased our understanding on TDP-43 structure and function in biology. The three-dimensional structures of TDP-43 domains such as N-terminal domain (NTD), RNA-recognition motif-1 (RRM1), RNA-recognition motif-2 (RRM2) and the C-terminal domain (CTD) or low-complexity domain (LCD) have been solved. These structures have yielded insights into novel binding sites and pockets at various TDP-43 domains, which can be targeted by designing a diverse library of ligands including small molecules, peptides and oligonucleotides as molecular tools to (i) study TDP-43 function, (ii) develop novel diagnostic agents and (iii) discover disease-modifying therapies to treat TDP-43 proteinopathies. This review provides a summary on recent progress in the development of TDP-43 binding ligands and uses the solved structures of various TDP-43 domains to investigate putative ligand binding regions that can be exploited to discover novel molecular probes to modulate TDP-43 structure and function.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Drug Development , Oligonucleotides/pharmacology , Peptides/pharmacology , Small Molecule Libraries/pharmacology , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Ligands , Molecular Structure , Oligonucleotides/chemical synthesis , Oligonucleotides/chemistry , Peptides/chemical synthesis , Peptides/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
2.
ACS Omega ; 6(12): 8680-8686, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33817530

ABSTRACT

The ß-amyloid (Aß) protein aggregation into toxic forms is one of the major factors in the Alzheimer's disease (AD) pathology. Screening compound libraries as inhibitors of Aß-aggregation is a common strategy to discover novel molecules as potential therapeutics in AD. In this regard, thioflavin T (ThT)-based fluorescence spectroscopy is a widely used in vitro method to identify inhibitors of Aß aggregation. However, conventional data processing of the ThT assay experimental results generally provides only qualitative output and lacks inhibitor-specific quantitative data, which can offer a number of advantages such as identification of critical inhibitor-specific parameters required to design superior inhibitors and reduce the need to conduct extensive in vitro kinetic studies. Therefore, we carried out mathematical modeling based on logistic equation and power law (PL) model to correlate the experimental results obtained from the ThT-based Aß40 aggregation kinetics for small-molecule inhibitors curcumin, orange G, and resveratrol and quantitatively fit the data in a logistic equation. This approach provides important inhibitor-specific parameters such as lag time λ, inflection point τ, maximum slope v m, and apparent rate constant k app, which are particularly useful in comparing the effectiveness of potential Aß40 aggregation inhibitors and can be applied in drug discovery campaigns to compare and contrast Aß40 inhibition data for large compound libraries.

3.
Pharmaceuticals (Basel) ; 14(1)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33430081

ABSTRACT

The current outbreak of severe acute respiratory distress syndrome (SARS) or nCOVID-19 pandemic, caused by the coronavirus-2 (CoV-2), continues to wreak havoc globally. As novel vaccines are being discovered and developed, small molecule drugs still constitute a viable treatment option for SARS-CoV-2 infections due to their advantages such as superior patient compliance for oral therapies, reduced manufacturing costs and ease of large scale distribution due to better stability and storage profiles. Discovering new drugs for SARS-CoV-2 infections is a time consuming and expensive proposition. In this regard, drug repurposing is an appealing approach which can provide rapid access to therapeutics with proven record of safety and efficacy. We investigated the drug repurposing potential of a library of dipeptidyl peptidase 4 (DPP4) inhibitors which are currently marketed for type-2 diabetes as treatment option for SARS-CoV-2 infections. These computational studies led to the identification of three marketed DPP4 inhibitors; gemigliptin, linagliptin and evogliptin as potential inhibitors of SARS-CoV-2 Mpro viral cysteine protease. In addition, our computational modeling shows that these drugs have the potential to inhibit other viral cysteine proteases from the beta coronavirus family, including the SAR-CoV Mpro and MERS-CoV CLpro suggesting their potential to be repurposed as broad-spectrum antiviral agents.

4.
Am J Cancer Res ; 11(12): 5762-5781, 2021.
Article in English | MEDLINE | ID: mdl-35018225

ABSTRACT

Ubiquitination is a key enzymatic post-translational modification that influences p53 stability and function. p53 protein regulates the expression of MDM2 (mouse double-minute 2 protein) E3 ligase and MDMX (double-minute 4 protein), through proteasome-based degradation. Exploration of targeting the ubiquitination pathway offers a potentially promising strategy for precision therapy in a variety of cancers. The p53-MDM2-MDMX pathway provides multiple molecular targets for small molecule screening as potential therapies for wild-type p53. As a result of its effect on molecular carcinogenesis, a personalized therapeutic approach based on the wild-type and mutant p53 protein is desirable. We highlighted the implications of p53 mutations in cancer, p53 ubiquitination mechanistic details, targeting p53-MDM2/MDMX interactions, significant discoveries related to MDM2 inhibitor drug development, MDM2 and MDMX dual target inhibitors, and clinical trials with p53-MDM2/MDMX-targeted drugs. We also investigated potential therapeutic repurposing of selective estrogen receptor modulators (SERMs) in targeting p53-MDM2/MDMX interactions. Molecular docking studies of SERMs were performed utilizing the solved structures of the p53/MDM2/MDMX proteins. These studies identified ormeloxifene as a potential dual inhibitor of p53/MDM2/MDMX interaction, suggesting that repurposing SERMs for dual targeting of p53/MDM2 and p53/MDMX interactions is an attractive strategy for targeting wild-type p53 tumors and warrants further preclinical research.

5.
Eur J Med Chem ; 167: 146-152, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30771602

ABSTRACT

Polyphenols like caffeic acid and its phenethyl ester have been associated with potent anti-aggregating activity. Accordingly, we screened a library of polyphenols and synthetic derivatives thereof for their capacity to inhibit tau-aggregation using a thioflavin T-based fluorescence method. Our results show that the nitrocatechol scaffold is required for a significant anti-aggregating activity, which is enhanced by introducing bulky substituents at the side chain. A remarkable increase in activity was observed for α-cyanocarboxamide derivatives 26-27. Molecular docking studies showed that the amide bond provides superior conformational stability in the steric zipper assembly of tau, which drives the increase in activity. We also found that derivatives 24-27 were potent chelators of copper(II) - a property of pharmacological significance in abnormal protein aggregation. These small molecules can provide promising leads to develop new drugs for tauopathies and AD. These findings open a new window on the repurposing of nitrocatechols beyond their established role as catechol-O-methyltransferase inhibitors.


Subject(s)
Caffeic Acids/chemistry , Catechols/chemistry , Nitro Compounds/chemistry , Phenylethyl Alcohol/analogs & derivatives , Protein Aggregation, Pathological/drug therapy , Caffeic Acids/pharmacology , Chelating Agents , Copper , Drug Design , Peptides , Phenylethyl Alcohol/pharmacology , Polyphenols/chemistry , Small Molecule Libraries , Tauopathies/drug therapy , tau Proteins/chemistry
6.
ACS Chem Neurosci ; 10(1): 226-234, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30157623

ABSTRACT

Treating Alzheimer's disease (AD) is a major challenge at the moment with no new drugs available to cure this devastating neurodegenerative disorder. In this regard, drug repurposing, which aims to determine novel therapeutic usage for drugs already approved by the regulatory agencies, is a pragmatic approach to discover novel treatment strategies. Selective serotonin reuptake inhibitors (SSRIs) are a known class of United States Food and Drug Administration approved drugs used in the treatment of depression. We investigated the ability of SSRIs fluvoxamine, fluoxetine, paroxetine, sertraline, and escitalopram on Aß42 aggregation and fibrillogenesis. Remarkably, the aggregation kinetic experiments carried out demonstrate the anti-Aß42 aggregation activity of SSRIs fluoxetine, paroxetine, and sertraline at all the tested concentrations (1, 10, 50, and 100 µM). Both fluoxetine and paroxetine were identified as the most promising SSRIs, showing 74.8 and 76% inhibition of Aß42 aggregation at 100 µM. The transmission electron microscopy experiments and dot-blot study also demonstrate the ability of fluoxetine and paroxetine to prevent Aß42 aggregation and fibrillogenesis, providing further evidence. Investigating the binding interactions of fluoxetine and paroxetine in the Aß42 oligomer and fibril models derived from the solid-state NMR structure suggests that these SSRIs interact at a region close to the N-terminal (Lys16-Glu22) in the S-shaped cross-ß-strand assembly and reduce Aß42 fibrillogenesis. On the basis of this study, a pharmacophore model is proposed which shows that the minimum structural requirements to design novel Aß42 aggregation inhibitors include the presence of one ionizable group, one hydrophobic group, two aromatic rings, and two hydrogen bond donor groups. These studies demonstrate that SSRIs have the potential to prevent Aß42 aggregation by direct binding and could be beneficial to AD patients on SSRIs.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Molecular Docking Simulation/methods , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Humans , Protein Aggregates/drug effects , Protein Aggregates/physiology , Protein Binding/drug effects , Protein Binding/physiology , Selective Serotonin Reuptake Inhibitors/therapeutic use
7.
Urol Ann ; 10(4): 406-408, 2018.
Article in English | MEDLINE | ID: mdl-30386095

ABSTRACT

Splenogonadal fusion (SGF) is a rare benign malformation in which spleen is aberrantly attached to the gonads or mesonephric derivatives. This entity often presents with scrotal mass, inguinal hernia, or cryptorchidism. Herein, we report our experience with a boy who presented with a scrotal enlargement which later turned out to be SGF. Moreover, an accessory spleen was unexpectedly found in the left inguinal canal. To the best of our knowledge, this is the first report of SGF and accessory spleen in the literature. Although rare, SGF should be included in the complete differential diagnosis list of scrotal masses, especially in children. The use of frozen section pathology if available would aid the surgeon in avoiding unnecessary orchiectomy in small testicular masses of benign origin.

8.
J Med Chem ; 59(16): 7584-97, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27463695

ABSTRACT

Recent efforts have been focused on the development of centrally active COMT inhibitors, which can be valuable assets for neurological disorders such as Parkinson's disease, due to the severe hepatotoxicity risk associated with tolcapone. New nitrocatechol COMT inhibitors based on naturally occurring caffeic acid and caffeic acid phenethyl ester were developed. All nitrocatechol derivatives displayed potent inhibition of peripheral and cerebral COMT within the nanomolar range. Druglike derivatives 13, 15, and 16 were predicted to cross the blood-brain barrier in vitro and were significantly less toxic than tolcapone and entacapone when incubated at 50 µM with rat primary hepatocytes. Moreover, their unique acidity and electrochemical properties decreased the chances of formation of reactive quinone-imines and, as such, the potential for hepatotoxicity. The binding mode of 16 confirmed that the major interactions with COMT were established via the nitrocatechol ring, allowing derivatization of the side chain for future lead optimization efforts.


Subject(s)
Benzophenones/pharmacology , Blood-Brain Barrier/drug effects , Catechol O-Methyltransferase Inhibitors/pharmacology , Catechol O-Methyltransferase/metabolism , Catechols/pharmacology , Hepatocytes/drug effects , Nitriles/pharmacology , Nitrophenols/pharmacology , Animals , Benzophenones/chemical synthesis , Benzophenones/chemistry , Blood-Brain Barrier/metabolism , Catechol O-Methyltransferase Inhibitors/chemical synthesis , Catechol O-Methyltransferase Inhibitors/chemistry , Catechols/chemical synthesis , Catechols/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Male , Models, Molecular , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry , Nitrophenols/chemical synthesis , Nitrophenols/chemistry , Rats , Rats, Wistar , Structure-Activity Relationship , Tolcapone
9.
ACS Med Chem Lett ; 7(5): 502-7, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27190601

ABSTRACT

A library of isomeric 2,4-diaminoquinazoline (DAQ) derivatives were synthesized and evaluated for antiaggregation potential toward Aß40/42. Structure-activity relationship data identified compound 3k (N (4)-(4-bromobenzyl)quinazoline-2,4-diamine) with a 4-bromobenzyl substituent as the most potent inhibitor (Aß40 IC50 = 80 nM) and was almost 18-fold more potent compared to the reference agent curcumin (Aß40 IC50 = 1.5 µM). The corresponding N (2)-isomer 4k (N (2)-(4-bromobenzyl)quinazoline-2,4-diamine) was also able to prevent Aß aggregation (Aß40 IC50 = 1.7 µM). However, compound 4k exhibited superior inhibition of Aß42 aggregation (Aß42 IC50 = 1.7 µM) compared to compound 3k (Aß42 IC50 = 14.8 µM) and was ∼1.8-fold more potent compared to curcumin (Aß42 IC50 = 3.1 µM). These results were supported by Aß aggregation kinetics investigations and transmission electron microscopy studies, which demonstrate the suitability of DAQ ring system to develop antiamyloid agents as pharmacological tools to study Aß aggregation.

10.
Eur J Med Chem ; 113: 258-72, 2016 May 04.
Article in English | MEDLINE | ID: mdl-26945113

ABSTRACT

Alzheimer's disease is of major concern all over the world due to a number of factors including (i) an aging population (ii) increasing life span and (iii) lack of effective pharmacotherapy options. The past decade has seen intense research in discovering disease-modifying multitargeting small molecules as therapeutic options. The pathophysiology of Alzheimer's disease is attributed to a number of factors such as the cholinergic dysfunction, amyloid/tau toxicity and oxidative stress/mitochondrial dysfunction. In recent years, targeting the amyloid cascade has emerged as an attractive strategy to discover novel neurotherapeutics. Formation of beta-amyloid species, with different degrees of solubility and neurotoxicity is associated with the gradual decline in cognition leading to dementia. The two commonly used approaches to prevent beta-amyloid accumulation in the brain include (i) development of beta-secretase inhibitors and (ii) designing direct inhibitors of beta-amyloid (self-induced) aggregation. This review highlights the amyloid cascade hypothesis and the key chemical features required to design small molecules that inhibit lower and higher order beta-amyloid aggregates. Several recent examples of small synthetic molecules with disease-modifying properties were considered and their molecular docking studies were conducted using either a dimer or steric-zipper assembly of beta-amyloid. These investigations provide a mechanistic understanding on the structural requirements needed to design novel small molecules with anti-amyloid aggregation properties. Significantly, this work also demonstrates that the structural requirements to prevent aggregation of various amyloid species differs considerably, which explains the fact that many small molecules do not exhibit similar inhibition profile toward diverse amyloid species such as dimers, trimers, tetramers, oligomers, protofibrils and fibrils.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Chemistry, Pharmaceutical , Humans , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...