Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Org Biomol Chem ; 22(16): 3249-3261, 2024 04 24.
Article En | MEDLINE | ID: mdl-38568016

A one-pot microwave assisted telescopic approach is reported for the chemo-selective synthesis of substituted 1,3-thiazetidines using readily available 2-aminopyridines/pyrazines/pyrimidine, substituted isothiocyanates and 1,2-dihalomethanes. The procedure involves thiourea formation from 2-aminopyridines/pyrazines/pyrimidine with the substituted isothiocyanates followed by a base catalysed nucleophilic attack of the CS bond on the 1,2-dihalomethane. Subsequently, a cyclization reaction occurs to yield substituted 1,3-thiazetidines. These four membered strained ring systems are reported to possess broad substrate scope with high functional group tolerance. The above synthetic sequence for the formation of four membered heterocycles is proven to be a modular and straightforward approach. Further the mechanistic pathway for the formation of 1,3-thiazetidines was supported by computational evaluations and X-ray crystallography analyses. The relevance of these thiazetidines in biological applications is evaluated by studying their ability to bind bio-macromolecules like proteins and nucleic acids.


Microwaves , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Crystallography, X-Ray , Proteins/chemistry , Thiazoles/chemistry , Thiazoles/chemical synthesis , Models, Molecular , Molecular Structure , Nucleic Acids/chemistry , Nucleic Acids/chemical synthesis , Isothiocyanates/chemistry , Isothiocyanates/chemical synthesis , Aminopyridines/chemistry , Aminopyridines/chemical synthesis
2.
Mol Biotechnol ; 2023 Aug 12.
Article En | MEDLINE | ID: mdl-37572221

p53 is a tumor suppressor gene activated in response to cellular stressors that inhibits cell cycle progression and induces pro-apoptotic signaling. The protein level of p53 is well balanced by the action of several E3 ligases and deubiquitinating enzymes (DUBs). Several DUBs have been reported to negatively regulate and promote p53 degradation in tumors. In this study, we identified USP19 as a negative regulator of p53 protein level. We demonstrate a direct interaction between USP19 and p53 by pull down assay. The overexpression of USP19 promoted ubiquitination of p53 and reduced its protein half-life. We also demonstrate that CRISPR/Cas9-mediated knockout of USP19 in cervical cancer cells elevates p53 protein levels, resulting in reduced colony formation, cell migration, and cell invasion. Overall, our results indicate that USP19 negatively regulates p53 protein levels in cervical cancer progression.

3.
Front Chem ; 10: 991369, 2022.
Article En | MEDLINE | ID: mdl-36247684

With an alarming increase in the number of cancer patients and a variety of tumors, it is high time for intensive investigation on more efficient and potent anti-tumor agents. Though numerous agents have enriched the literature, still there exist challenges, with the availability of different targets and possible cross-reactivity. Herein we have chosen the phosphoinositide 3-kinase (PI3K) as the target of interest and investigated the potential of pyrido fused imidazo[4,5-c]quinoline derivatives to bind strongly to the active site, thereby inhibiting the progression of various types of tumors. The AutoDock, Glide and the Prime-MM/GBSA analysis are used to execute the molecular docking investigation and validation for the designed compounds. The anti-tumor property evaluations were carried out by using PASS algorithm. Based on the GLIDE score, the binding affinity of the designed molecules towards the target PI3K was evaluated. The energetics associated with static interactions revealed 1j as the most potential candidate and the dynamic investigations including RMSD, RMSF, Rg, SASA and hydrogen bonding also supported the same through relative stabilization induced through ligand interactions. Subsequently, the binding free energy of the Wortmannin and 1j complex calculated using MM-PBSA analysis. Further evaluations with PASS prediction algorithm also supported the above results. The studies reveal that there is evidence for considering appropriate pyrido fused imidazo[4,5-c]quinoline compounds as potential anti-tumor agents.

4.
Theranostics ; 12(13): 5949-5970, 2022.
Article En | MEDLINE | ID: mdl-35966591

Background: Cisplatin is one of the frontline anticancer agents. However, development of cisplatin-resistance limits the therapeutic efficacy of cisplatin-based treatment. The expression of microtubule-associated serine/threonine kinase 1 (MAST1) is a primary factor driving cisplatin-resistance in cancers by rewiring the MEK pathway. However, the mechanisms responsible for MAST1 regulation in conferring drug resistance is unknown. Methods: We implemented a CRISPR/Cas9-based, genome-wide, dual screening system to identify deubiquitinating enzymes (DUBs) that govern cisplatin resistance and regulate MAST1 protein level. We analyzed K48- and K63-linked polyubiquitination of MAST1 protein and mapped the interacting domain between USP1 and MAST1 by immunoprecipitation assay. The deubiquitinating effect of USP1 on MAST1 protein was validated using rescue experiments, in vitro deubiquitination assay, immunoprecipitation assays, and half-life analysis. Furthermore, USP1-knockout A549 lung cancer cells were generated to validate the deubiquitinating activity of USP1 on MAST1 abundance. The USP1-MAST1 correlation was evaluated using bioinformatics tool and in different human clinical tissues. The potential role of USP1 in regulating MAST1-mediated cisplatin resistance was confirmed using a series of in vitro and in vivo experiments. Finally, the clinical relevance of the USP1-MAST1 axis was validated by application of small-molecule inhibitors in a lung cancer xenograft model in NSG mice. Results: The CRISPR/Cas9-based dual screening system identified USP1 as a novel deubiquitinase that interacts, stabilizes, and extends the half-life of MAST1 by preventing its K48-linked polyubiquitination. The expression analysis across human clinical tissues revealed a positive correlation between USP1 and MAST1. USP1 promotes MAST1-mediated MEK1 activation as an underlying mechanism that contributes to cisplatin-resistance in cancers. Loss of USP1 led to attenuation of MAST1-mediated cisplatin-resistance both in vitro and in vivo. The combined pharmacological inhibition of USP1 and MAST1 using small-molecule inhibitors further abrogated MAST1 level and synergistically enhanced cisplatin efficacy in a mouse xenograft model. Conclusions: Overall, our study highlights the role of USP1 in the development of cisplatin resistance and uncovers the regulatory mechanism of MAST1-mediated cisplatin resistance in cancers. Co-treatment with USP1 and MAST1 inhibitors abrogated tumor growth and synergistically enhanced cisplatin efficacy, suggesting a novel alternative combinatorial therapeutic strategy that could further improve MAST1-based therapy in patients with cisplatin-resistant tumors.


Cisplatin , Lung Neoplasms , Animals , CRISPR-Cas Systems/genetics , Cisplatin/pharmacology , Cisplatin/therapeutic use , Early Detection of Cancer , Humans , Mice , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Protein Serine-Threonine Kinases/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
5.
J Comput Chem ; 43(9): 619-630, 2022 04 05.
Article En | MEDLINE | ID: mdl-35167132

In this study, we assess the effective inhibition of a series of thiazolidine derivatives (1a-1q) were adopting structure-based drug design. Thiazolidine is a five-membered ring structure with thioether and amino groups at positions 1 and 3. Although, thiazolidine may bind to a wide range of protein targets, it is a major heterocyclic core in medicinal chemistry. Different scoring utilities including AutoDock Vina, Glide, and MM/GBSA analysis were performed to commensurate the improvement of screening progress. The evaluated binding affinities were validated by molecular dynamics simulations over a period of 20 ns for the interactions between the Mycobacterium tuberculosis protein PrpR with three novel scaffolds (1b, 1j, and 1k). All the scaffolds exhibited distinct stable interactions with the significant residues like Arg169, Arg197, Tyr248, Arg308, and Gly311 respectively. Further, the inhibitory activities of scaffolds were predicted and evaluated by machine learning based algorithm to rank the above proposed compounds. This study reveals the potential of 1k and 1j as effective inhibitor candidates for the treatment of tuberculosis.


Mycobacterium tuberculosis , Antitubercular Agents/chemistry , Antitubercular Agents/metabolism , Antitubercular Agents/pharmacology , Drug Design , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Mycobacterium tuberculosis/metabolism
6.
Int J Mol Sci ; 22(16)2021 Aug 07.
Article En | MEDLINE | ID: mdl-34445214

Deubiquitinating enzymes play key roles in the precise modulation of Aurora B-an essential cell cycle regulator. The expression of Aurora B increases before the onset of mitosis and decreases during mitotic exit; an imbalance in these levels has a severe impact on the fate of the cell cycle. Dysregulation of Aurora B can lead to aberrant chromosomal segregation and accumulation of errors during mitosis, eventually resulting in cytokinesis failure. Thus, it is essential to identify the precise regulatory mechanisms that modulate Aurora B levels during the cell division cycle. Using a deubiquitinase knockout strategy, we identified USP48 as an important candidate that can regulate Aurora B protein levels during the normal cell cycle. Here, we report that USP48 interacts with and stabilizes the Aurora B protein. Furthermore, we showed that the deubiquitinating activity of USP48 helps to maintain the steady-state levels of Aurora B protein by regulating its half-life. Finally, USP48 knockout resulted in delayed progression of cell cycle due to accumulation of mitotic defects and ultimately cytokinesis failure, suggesting the role of USP48 in cell cycle regulation.


Aurora Kinase B/metabolism , Cytokinesis , Mitosis , Ubiquitin-Specific Proteases/metabolism , Aurora Kinase B/genetics , Enzyme Stability , HEK293 Cells , HeLa Cells , Humans , Ubiquitin-Specific Proteases/genetics
7.
Int J Mol Sci ; 22(11)2021 May 28.
Article En | MEDLINE | ID: mdl-34071237

Cell division cycle 25A (Cdc25A) is a dual-specificity phosphatase that is overexpressed in several cancer cells and promotes tumorigenesis. In normal cells, Cdc25A expression is regulated tightly, but the changes in expression patterns in cancer cells that lead to tumorigenesis are unknown. In this study, we showed that ubiquitin-specific protease 29 (USP29) stabilized Cdc25A protein expression in cancer cell lines by protecting it from ubiquitin-mediated proteasomal degradation. The presence of USP29 effectively blocked polyubiquitination of Cdc25A and extended its half-life. CRISPR-Cas9-mediated knockdown of USP29 in HeLa cells resulted in cell cycle arrest at the G0/G1 phase. We also showed that USP29 knockdown hampered Cdc25A-mediated cell proliferation, migration, and invasion of cancer cells in vitro. Moreover, NSG nude mice transplanted with USP29-depleted cells significantly reduced the size of the tumors, whereas the reconstitution of Cdc25A in USP29-depleted cells significantly increased the tumor size. Altogether, our results implied that USP29 promoted cell cycle progression and oncogenic transformation by regulating protein turnover of Cdc25A.


Carcinogenesis/metabolism , Gene Expression Regulation, Neoplastic , Ubiquitin-Specific Proteases/metabolism , cdc25 Phosphatases/metabolism , Animals , Apoptosis , CRISPR-Cas Systems , Carcinogenesis/genetics , Cell Cycle , Cell Cycle Checkpoints , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Heterografts , Humans , Male , Mice , Mice, Nude , Mice, SCID , Oncogenes , Ubiquitin/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination , cdc25 Phosphatases/genetics
8.
RSC Adv ; 11(52): 32680-32705, 2021 Oct 04.
Article En | MEDLINE | ID: mdl-35493554

In the field of drug discovery, isoxazole is a five-membered heterocyclic moiety commonly found in many commercially available drugs. In view of their enormous significance, it is always imperative to unleash new eco-friendly synthetic strategies. Among various novel synthetic techniques in use for isoxazole synthesis, most synthetic methods employ Cu(i) or Ru(ii) as catalysts for (3 + 2) cycloaddition reaction. The particular disadvantages associated with metal-catalyzed reactions are high costs, low abundance, toxicity, a significant generation of waste, and difficulty to separate from the reaction mixtures. In view of these drawbacks, it is always imperative to develop alternate metal-free synthetic routes. This review article highlights a comprehensive overview on the potential application of metal-free synthetic routes for the synthesis of isoxazoles with significant biological interests.

9.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118835, 2020 12.
Article En | MEDLINE | ID: mdl-32860838

Resistance to DNA-damaging agents is one of the main reasons for the low survival of cervical cancer patients. Previous reports have suggested that the Cdc25A oncoprotein significantly affects the level of susceptibility to DNA-damaging agents, but the molecular mechanism remains unclear. In this study, we used Western blot and flow cytometry analyses to demonstrate that the deubiquitinating enzyme HAUSP stabilizes Cdc25A protein level. Furthermore, in a co-immunoprecipitation assay, we found that HAUSP interacts with and deubiquitinates Cdc25A both exogenously and endogenously. HAUSP extends the half-life of the Cdc25A protein by circumventing turnover. HAUSP knockout in HeLa cells using the CRISPR/Cas9 system caused a significant delay in Cdc25A-mediated cell cycle progression, cell migration, and colony formation and attenuated tumor progression in a mouse xenograft model. Furthermore, HAUSP-mediated stabilization of the Cdc25A protein produced enhanced resistance to DNA-damaging agents. Overall, our study suggests that targeting Cdc25A and HAUSP could be a promising combinatorial approach to halt progression and minimize antineoplastic resistance in cervical cancer.


Drug Resistance, Neoplasm/genetics , Ubiquitin-Specific Peptidase 7/genetics , Uterine Cervical Neoplasms/genetics , cdc25 Phosphatases/genetics , Animals , CRISPR-Cas Systems , DNA Damage/genetics , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Heterografts , Humans , Mice , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology
10.
Cell Death Differ ; 27(11): 3004-3020, 2020 11.
Article En | MEDLINE | ID: mdl-32415280

Conventional screening methods for deubiquitinating enzymes (DUBs) have important limitations. A loss-of-function study based on the knockout of DUB genes in mammalian cells can provide an excellent model for exploring DUB function. Here, we used CRISPR-Cas9 to perform genome-scale knockout of the entire set of genes encoding ubiquitin-specific proteases (USPs), a DUB subfamily, and then systematically screened for DUBs that stabilize the Cdc25A oncoprotein. USP3 was identified as a deubiquitinase of Cdc25A. USP3 depletion reduces the Cdc25A protein level, resulting in a significant delay in cell-cycle progression, and reduces the growth of cervical tumor xenografts in nude mice. Clinically, USP3 expression is positively correlated with Cdc25A protein expression and the poorest survival in breast cancer. We envision that our DUB knockout library kit will facilitate genome-scale screening of functional DUBs for target proteins of interest in a wide range of biomedical fields.


Cell Cycle/genetics , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Uterine Cervical Neoplasms/metabolism , cdc25 Phosphatases/genetics , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Female , Humans , Male , Mice , Mice, Nude , Survival Analysis , Ubiquitin-Specific Proteases/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays , cdc25 Phosphatases/metabolism
11.
Mol Cells ; 43(3): 203-214, 2020 Mar 31.
Article En | MEDLINE | ID: mdl-32133826

Post-translational modifications play major roles in the stability, function, and localization of target proteins involved in the nervous system. The ubiquitin-proteasome pathway uses small ubiquitin molecules to degrade neuronal proteins. Deubiquitinating enzymes (DUBs) reverse this degradation and thereby control neuronal cell fate, synaptic plasticity,axonal growth, and proper function of the nervous system.Moreover, mutations or downregulation of certain DUBshave been found in several neurodegenerative diseases, as well as gliomas and neuroblastomas. Based on emerging findings, DUBs represent an important target for therapeutic intervention in various neurological disorders. Here, we summarize advances in our understanding of the roles of DUBs related to neurobiology.


Deubiquitinating Enzymes/metabolism , Nervous System/pathology , Neurodegenerative Diseases/physiopathology , Protein Processing, Post-Translational/genetics , Ubiquitin/metabolism , Humans
12.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 599-610, 2019 03 01.
Article En | MEDLINE | ID: mdl-30543854

RNA-binding protein LIN28A is often highly expressed in human malignant tumors and is involved in tumor metastasis and poor prognosis. Knowledge about post-translational regulatory mechanisms governing LIN28A protein stability and function is scarce. Here, we investigated the role of ubiquitination and deubiquitination on LIN28A protein stability and report that LIN28A protein undergoes ubiquitination. Ubiquitin-specific protease 28 (USP28), a deubiquitinating enzyme, interacts with and stabilizes LIN28A protein to extend its half-life. USP28, through its deubiquitinating activity, antagonizes LIN28A protein turnover by reversing its proteasomal degradation. Our study describes the consequential impacts of USP28-mediated stabilization of LIN28A protein on enhancing cancer cell viability, migration and ultimately augmenting LIN28A-mediated tumor progression. Overall, our data suggest that a synergistic, combinatorial approach of targeting LIN28A with USP28 would contribute to effective cancer therapeutics.


Carcinogenesis/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Ubiquitin Thiolesterase/physiology , Cells, Cultured , HCT116 Cells , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , K562 Cells , MCF-7 Cells , Oncogenes/physiology , Protein Binding , Protein Processing, Post-Translational/genetics , Protein Stability , Proteolysis , RNA-Binding Proteins/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitination
13.
Arch Pharm Res ; 41(9): 911-920, 2018 Sep.
Article En | MEDLINE | ID: mdl-29855892

Targeted genome editing by clustered regularly interspaced short palindromic repeats (CRISPR-Cas9) raised concerns over off-target effects. The use of double-nicking strategy using paired Cas9 nickase has been developed to minimize off-target effects. However, it was reported that the efficiency of paired nickases were comparable or lower than that of either corresponding nuclease alone. Recently, we conducted a systematic comparison of the efficiencies of several paired Cas9 with their corresponding Cas9 nucleases and showed that paired D10A Cas9 nickases are sometimes more efficient than individual nucleases for gene disruption. However, sometimes the designed paired Cas9 nickases exhibited significantly lower mutation frequencies than nucleases, hampering the generation of cells containing paired Cas9 nickase-induced mutations. Here we implemented IRES peptide-conjugation of fluorescent protein to Cas9 nickase and subjected for fluorescence-activated cell sorting. The sorted cell populations are highly enriched with cells containing paired Cas9 nickase-induced mutations, by a factor of up to 40-fold as compared with the unsorted population. Furthermore, gene-disrupted single cell clones using paired nickases followed by FACS sorting strategy were generated highly efficiently, without compromising with its low off-target effects. We envision that our fluorescent protein coupled paired nickase-mediated gene disruption, facilitating efficient and highly specific genome editing in medical research.


CRISPR-Cas Systems/genetics , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Flow Cytometry , Gene Editing , Luminescent Proteins/metabolism , Animals , Cell Separation , Cells, Cultured , HEK293 Cells , Humans , Luminescent Proteins/genetics , Mice , NIH 3T3 Cells
14.
Discov Med ; 25(138): 177-194, 2018 04.
Article En | MEDLINE | ID: mdl-29723489

Ran-binding protein in the microtubule-organizing center (RanBPM) is an evolutionarily conserved, nucleocytoplasmic scaffolding protein involved in various cellular processes and several signal transduction pathways. RanBPM has a crucial role in mediating disease pathology by interacting with diverse proteins to regulate their functions. Previously, we compiled diverse cellular functions of RanBPM. Since then the functions of RanBPM have increased exponentially. In this article, we have updated the functions of RanBPM through its manifold interactions that have been investigated to date, according to their roles in protein stability, transcriptional activity, cellular development, neurobiology, and the cell cycle. Our review provides a complete guide on RanBPM interactors, the physiological role of RanBPM in cellular functions, and potential applications in disease therapeutics.


Adaptor Proteins, Signal Transducing , Cell Cycle , Cytoskeletal Proteins , Gene Expression Regulation , Neoplasm Proteins , Neoplasms , Nuclear Proteins , Signal Transduction , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Stability
15.
Drug Discov Today ; 22(12): 1816-1824, 2017 12.
Article En | MEDLINE | ID: mdl-28847759

The Ran-binding protein microtubule-organizing center (RanBPM) is a highly conserved nucleocytoplasmic protein involved in a variety of intracellular signaling pathways that control diverse cellular functions. RanBPM interacts with proteins that are linked to various diseases, including Alzheimer's disease (AD), schizophrenia (SCZ), and cancer. In this article, we define the characteristics of the scaffolding protein RanBPM and focus on its interaction partners in diverse physiological disorders, such as neurological diseases, fertility disorders, and cancer.


Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Animals , Brain Diseases/metabolism , Cytoskeletal Proteins/chemistry , Humans , Neoplasms/metabolism , Nuclear Proteins/chemistry , Reproduction/physiology
16.
Carbohydr Polym ; 173: 121-130, 2017 Oct 01.
Article En | MEDLINE | ID: mdl-28732850

Advanced biomedical materials can potentially be developed from combinations of natural biodegradable polymers and synthetic polymers. We synthesized bioactive composites based on polypyrrole/chitosan through in-situ electrochemical polymerization in oxalic acid medium. Surface characterization results revealed the influence of chitosan inclusion on polypyrrole (PPy) surface morphology. Contact angle results confirmed the enhancement in surface hydrophilicity due to the addition of chitosan into the PPy matrix. Electrochemical corrosion studies revealed that the composite coatings showed enhanced protective performance compared to pure PPy. Further, we investigated the effect of the composite coatings on the growth of MG-63 human osteoblast cells to assess their biocompatibility. Monte Carlo simulations were engaged to assess the interactions between the metal surface and composite coatings. The composite containing equal parts PPy and chitosan was found to be biocompatible; together with the corrosion protection results, the findings indicated that this bioactive coating material has potential for use in 316L SS implants.


Chitosan/chemistry , Coated Materials, Biocompatible , Osteoblasts/drug effects , Polymers/chemistry , Pyrroles/chemistry , Cell Line , Humans , Prostheses and Implants , Surface Properties
...