Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem J ; 480(14): 1079-1096, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37306466

RESUMEN

Mycobacterium tuberculosis (M. tb), the causative pathogen of tuberculosis (TB) remains the leading cause of death from single infectious agent. Furthermore, its evolution to multi-drug resistant (MDR) and extremely drug-resistant (XDR) strains necessitate de novo identification of drug-targets/candidates or to repurpose existing drugs against known targets through drug repurposing. Repurposing of drugs has gained traction recently where orphan drugs are exploited for new indications. In the current study, we have combined drug repurposing with polypharmacological targeting approach to modulate structure-function of multiple proteins in M. tb. Based on previously established essentiality of genes in M. tb, four proteins implicated in acceleration of protein folding (PpiB), chaperone assisted protein folding (MoxR1), microbial replication (RipA) and host immune modulation (S-adenosyl dependent methyltransferase, sMTase) were selected. Genetic diversity analyses in target proteins showed accumulation of mutations outside respective substrate/drug binding sites. Using a composite receptor-template based screening method followed by molecular dynamics simulations, we have identified potential candidates from FDA approved drugs database; Anidulafungin (anti-fungal), Azilsartan (anti-hypertensive) and Degarelix (anti-cancer). Isothermal titration calorimetric analyses showed that the drugs can bind with high affinity to target proteins and interfere with known protein-protein interaction of MoxR1 and RipA. Cell based inhibitory assays of these drugs against M. tb (H37Ra) culture indicates their potential to interfere with pathogen growth and replication. Topographic assessment of drug-treated bacteria showed induction of morphological aberrations in M. tb. The approved candidates may also serve as scaffolds for optimization to future anti-mycobacterial agents which can target MDR strains of M. tb.


Asunto(s)
Antituberculosos , Reposicionamiento de Medicamentos , Mycobacterium tuberculosis , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/genética , Antituberculosos/farmacología , Tuberculosis Extensivamente Resistente a Drogas/tratamiento farmacológico , Anidulafungina/farmacología , Proteínas Bacterianas/genética , Estructura Terciaria de Proteína , Simulación de Dinámica Molecular
2.
J Cell Biochem ; 124(10): 1516-1529, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37566682

RESUMEN

The emergence of multiple drug resistance and extreme drug resistance pathogens necessitates the continuous evaluation of the pathogenic genome to identify conserved molecular targets and their respective inhibitors. In this study, we mapped the global mutational landscape of Neisseria gonorrhoeae (an intracellular pathogen notoriously known to cause the sexually transmitted disease gonorrhoea). We identified highly variable amino acid positions in the antibiotic target genes like the penA, ponA, 23s rRNA, rpoB, gyrA, parC, mtrR and porB. Some variations are directly reported to confer resistance to the currently used front-line drugs like ceftriaxone, cefixime, azithromycin and ciprofloxacin. Further, by whole genome comparison and Shannon entropy analysis, we identified a completely conserved protein HtpX in the drug-resistant as well as susceptible isolates of N. gonorrhoeae (NgHtpX). Comparison with the only available information of Escherichia coli HtpX suggested it to be a transmembrane metalloprotease having a role in stress response. The critical zinc-binding residue of NgHtpX was mapped to E141. By applying composite high throughput screening followed by MD simulations, we identified pemirolast and thalidomide as high-energy binding ligands of NgHtpX. Following cloning and expression of the purified metal-binding domain of NgHtpX (NgHtpXd), its Zn2+ -binding (Kd = 0.4 µM) and drug-binding (pemirolast, Kd = 3.47 µM; and thalidomide, Kd = 1.04 µM) potentials were determined using in-vitro fluorescence quenching experiment. When tested on N. gonorrhoeae cultures, both the ligands imposed a dose-dependent reduction in viability. Overall, our results provide high entropy positions in the targets of presently used antibiotics, which can be further explored to understand the AMR mechanism. Additionally, HtpX and its specific inhibitors identified can be utilised effectively in managing gonococcal infections.

3.
Biochem Biophys Res Commun ; 665: 88-97, 2023 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-37149987

RESUMEN

Bacillus anthracis Ser/Thr protein kinase PrkC is necessary for phenotypic memory and spore germination, and the loss of PrkC-dependent phosphorylation events affect the spore development. During sporulation, Bacillus sp. can store 3-Phosphoglycerate (3-PGA) that will be required at the onset of germination when ATP will be necessary. The Phosphoglycerate mutase (Pgm) catalyzes the isomerization of 2-PGA and 3-PGA and is important for spore germination as a key metabolic enzyme that maintains 3-PGA pool at later events. Therefore, regulation of Pgm is important for an efficient spore germination process and metabolic switching. While the increased expression of Pgm in B. anthracis decreases spore germination efficiency, it remains unexplored if PrkC could directly influence Pgm activity. Here, we report the phosphorylation and regulation of Pgm by PrkC and its impact on Pgm stability and catalytic activity. Mass spectrometry revealed Pgm phosphorylation on seven threonine residues. In silico mutational analysis highlighted the role of Thr459 residue towards metal and substrate binding. Altogether, we demonstrated that PrkC-mediated Pgm phosphorylation negatively regulates its activity that is essential to maintain Pgm in its apo-like isoform before germination. This study advances the role of Pgm regulation that represents an important switch for B. anthracis resumption of metabolism and spore germination.


Asunto(s)
Bacillus anthracis , Proteínas Quinasas , Fosforilación , Proteínas Quinasas/metabolismo , Bacillus anthracis/metabolismo , Fosfoglicerato Mutasa/metabolismo , Treonina/metabolismo , Esporas Bacterianas/genética , Esporas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo
4.
BMC Bioinformatics ; 23(1): 319, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35931960

RESUMEN

BACKGROUND: Visceral Leishmaniasis (VL) is a fatal vector-borne parasitic disorder occurring mainly in tropical and subtropical regions. VL falls under the category of neglected tropical diseases with growing drug resistance and lacking a licensed vaccine. Conventional vaccine synthesis techniques are often very laborious and challenging. With the advancement of bioinformatics and its application in immunology, it is now more convenient to design multi-epitope vaccines comprising predicted immuno-dominant epitopes of multiple antigenic proteins. We have chosen four antigenic proteins of Leishmania donovani and identified their T-cell and B-cell epitopes, utilizing those for in-silico chimeric vaccine designing. The various physicochemical characteristics of the vaccine have been explored and the tertiary structure of the chimeric construct is predicted to perform docking studies and molecular dynamics simulations. RESULTS: The vaccine construct is generated by joining the epitopes with specific linkers. The predicted tertiary structure of the vaccine has been found to be valid and docking studies reveal the construct shows a high affinity towards the TLR-4 receptor. Population coverage analysis shows the vaccine can be effective on the majority of the world population. In-silico immune simulation studies confirms the vaccine to raise a pro-inflammatory response with the proliferation of activated T and B cells. In-silico codon optimization and cloning of the vaccine nucleic acid sequence have also been achieved in the pET28a vector. CONCLUSION: The above bioinformatics data support that the construct may act as a potential vaccine. Further wet lab synthesis of the vaccine and in vivo works has to be undertaken in animal model to confirm vaccine potency.


Asunto(s)
Leishmania donovani , Leishmaniasis Visceral , Biología Computacional/métodos , Epítopos de Linfocito B , Epítopos de Linfocito T/química , Humanos , Leishmaniasis Visceral/prevención & control , Simulación del Acoplamiento Molecular , Vacunas de Subunidad/química
5.
Biochemistry ; 61(20): 2188-2197, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36166360

RESUMEN

The receptor binding domain(s) (RBD) of spike (S) proteins of SARS-CoV-1 and SARS-CoV-2 (severe acute respiratory syndrome coronavirus) undergoes closed to open transition to engage with host ACE2 receptors. In this study, using multi atomistic (equilibrium) and targeted (non-equilibrium) molecular dynamics simulations, we have compared energetics of RBD opening pathways in full-length (modeled from cryo-EM structures) S proteins of SARS-CoV-1 and SARS-CoV-2. Our data indicate that amino acid variations at the RBD interaction interface can culminate into distinct free energy landscapes of RBD opening in these S proteins. We further report that mutations in the S protein of SARS-CoV-2 variants of concern can reduce the protein-protein interaction affinity of RBD(s) with its neighboring domains and could favor its opening to access ACE2 receptors. The findings can also aid in predicting the impact of future mutations on the rate of S protein opening for rapid host receptor scanning.


Asunto(s)
SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , Aminoácidos/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Sitios de Unión , COVID-19/genética , Mutación , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/química
6.
J Cell Biochem ; 123(7): 1171-1182, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35527389

RESUMEN

The emergence of multiple drug-resistant "super gonorrhoea" complicates the management and treatment of Neisseria gonorrhoeae infections due to the progressive accumulation of mutations in the biological targets of frontline antimicrobials. Continuous evaluation and reporting of newer molecular targets and their inhibitors are necessary. Here, we present l-asparaginase of N. gonorrhoeae (NgA) as a new molecular target based on structure-based high-throughput screening, molecular dynamics(MD) simulations, and validation by biophysical, biochemical, and cell viability assays. We observed that the NgA is evolutionarily conserved in both the drug-resistant and susceptible strains of N. gonorrhoeae, indicating its importance in the growth and survival of the pathogen. Three Food and Drug Administration-approved drugs, pemirolast, thalidomide, and decitabine, were identified as potential inhibitors of NgA using high-throughput screening. The binding energies of the drugs with NgA were -20.14, -19.67, and -16.47 kcal/mol, respectively, compared to -6.82 ± 1.46 for enzyme-substrate l-Asn, as obtained through MD simulations. Subsequently, fluorescence quenching and differential scanning calorimetry experiments validated the in silico data. The observance of inhibition of NgA activity at micromolar drug concentrations further strengthened our findings. Conclusive evidence came from the cell viability assays where these drugs were found to impede the growth of N. gonorrhoeae culture effectively. Thus, our study establishes l-asparaginase as a new molecular target against gonococcal infections. From this study, we propose that targeting of NgA can be explored to control N. gonorrhoeae infections in combination therapy.


Asunto(s)
Antiinfecciosos , Gonorrea , Antibacterianos/farmacología , Antiinfecciosos/farmacología , Asparaginasa/farmacología , Farmacorresistencia Bacteriana/genética , Gonorrea/tratamiento farmacológico , Gonorrea/prevención & control , Humanos , Pruebas de Sensibilidad Microbiana , Neisseria gonorrhoeae/genética
7.
Biochem J ; 476(12): 1817-1841, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31138770

RESUMEN

Compelling evidence implicates self-assembly of amyloid-ß (Aß1-42) peptides into soluble oligomers and fibrils as a major underlying event in Alzheimer's disease (AD) pathogenesis. Herein, we employed amyloid-degrading keratinase (kerA) enzyme as a key Aß1-42-binding scaffold to identify five keratinase-guided peptides (KgPs) capable of interacting with and altering amyloidogenic conversion of Aß1-42 The KgPs showed micromolar affinities with Aß1-42 and abolished its sigmoidal amyloidogenic transition, resulting in abrogation of fibrillogenesis. Comprehensive assessment using dynamic light scattering (DLS), atomic force microscopy (AFM) and Fourier-transform infrared (FTIR) spectroscopy showed that KgPs induced the formation of off-pathway oligomers comparatively larger than the native Aß1-42 oligomers but with a significantly reduced cross-ß signature. These off-pathway oligomers exhibited low immunoreactivity against oligomer-specific (A11) and fibril-specific (OC) antibodies and rescued neuronal cells from Aß1-42 oligomer toxicity as well as neuronal apoptosis. Structural analysis using molecular docking and molecular dynamics (MD) simulations showed two preferred KgP binding sites (Lys16-Phe20 and Leu28-Val39) on the NMR ensembles of monomeric and fibrillar Aß1-42, indicating an interruption of crucial hydrophobic and aromatic interactions. Overall, our results demonstrate a new approach for designing potential anti-amyloid molecules that could pave way for developing effective therapeutics against AD and other amyloid diseases.


Asunto(s)
Péptidos beta-Amiloides , Apoptosis , Bacillus licheniformis/enzimología , Proteínas Bacterianas/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Neuronas/metabolismo , Fragmentos de Péptidos , Péptido Hidrolasas/química , Agregado de Proteínas , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Línea Celular Tumoral , Humanos , Neuronas/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo
8.
J Cell Biochem ; 120(1): 143-154, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30230585

RESUMEN

Multidrug-resistant Mycobacterium tuberculosis (Mtb) has emerged as a major health challenge, necessitating the search for new molecular targets. A secretory amidohydrolase, l-asparaginase of Mtb (MtA), originally implicated in nitrogen assimilation and neutralization of acidic microenvironment inside human alveolar macrophages, has been proposed as a crucial metabolic enzyme. To investigate whether this enzyme could serve as a potential drug target, it was studied for structural details and active site-specific inhibitors were tested on cultured Mycobacterial strain. The structural details of MtA obtained through comparative modeling and molecular dynamics simulations provided insights about the orchestration of an alternate reaction mechanism at the active site. This was contrary to the critical Tyr flipping mechanism reported in other asparaginases. We report the novel finding of Tyr to Val replacement in catalytic triad I along with the structural reorganization of a ß-hairpin loop upon substrate binding in MtA active site. Further, 5 MtA-specific, active-site-based inhibitors were obtained by following a rigorous differential screening protocol. When tested on Mycobacterium culture, 3 of these, M3 (ZINC 4740895), M26 (ZINC 33535), and doxorubicin showed promising results with inhibitory concentrations (IC 50 ) of 431, 100, and 56 µM, respectively. Based on our findings and considering stark differences with human asparaginase, we project MtA as a promising molecular target against which the selected inhibitors may be used to counteract Mtb infection effectively.


Asunto(s)
Antituberculosos/química , Antituberculosos/farmacología , Asparaginasa/antagonistas & inhibidores , Asparaginasa/química , Doxorrubicina/química , Doxorrubicina/farmacología , Mycobacterium tuberculosis/enzimología , Secuencia de Aminoácidos , Dominio Catalítico , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Estabilidad de Medicamentos , Humanos , Enlace de Hidrógeno , Concentración 50 Inhibidora , Microscopía de Fuerza Atómica , Simulación de Dinámica Molecular , Terapia Molecular Dirigida , Conformación Proteica en Hélice alfa , Estructura Terciaria de Proteína , Interfaz Usuario-Computador
9.
Biochemistry ; 57(16): 2359-2372, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29637772

RESUMEN

Gelsolin is an actin-severing protein that attains an open functional conformation in the presence of Ca2+ or low pH. Mutations (D187N/Y) in the second domain of gelsolin trigger the proteolytic pathway producing amyloidogenic fragments that form the pathological hallmark of gelsolin amyloidosis and lattice corneal dystrophy type 2 (LCD2). Here, we show that the D187N mutant gelsolin in a Ca2+ depleted, low pH-activated, open conformation could assemble into amyloidogenic oligomers without necessarily undergoing the specific proteolytic step. Although both wild-type (WT) and mutant proteins exhibit closely overlapping globular shapes at physiological conditions, the latter exhibits subjugated actin depolymerization, loss of thermodynamic stability, and folding cooperativity. Mutant gelsolin displayed aberrant conformational unwinding and formed structural conformers with high associative properties at low pH conditions. A SAXS intensity profile and Guinier analysis of these conformers showed the formation of unusual, higher order aggregates. Extended incubation at low pH resulted in the formation of thioflavin T and Congo red positive, ß-sheet rich aggregates with a fibrillar, amyloid-like morphology visible under electron and atomic force microscopy. Mass spectrometric analysis of disaggregated end-stage fibrils displayed peptide fragments encompassing the entire protein sequence, indicating the involvement of full length mutant gelsolin in fibril formation. Atomistic and REMD simulations indicated a larger increase in solvent accessibility and loss of fold architecture in mutant gelsolin at low pH as compared to WT gelsolin. Our findings support the existence of a secondary oligomerization-dependent aggregation pathway associated with gelsolin amyloidosis and can pave the way for better therapeutic strategies.


Asunto(s)
Proteínas Amiloidogénicas/genética , Gelsolina/genética , Proteínas Mutantes/genética , Conformación Proteica , Secuencia de Aminoácidos/genética , Amiloide/química , Amiloide/genética , Proteínas Amiloidogénicas/química , Gelsolina/química , Humanos , Microscopía de Fuerza Atómica , Proteínas Mutantes/química , Mutación , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Agregación Patológica de Proteínas/genética , Estabilidad Proteica , Proteolisis , Difracción de Rayos X
10.
J Biol Chem ; 292(40): 16440-16462, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28821620

RESUMEN

Cardiac hypertrophy and associated heart fibrosis remain a major cause of death worldwide. Phytochemicals have gained attention as alternative therapeutics for managing cardiovascular diseases. These include the extract from the plant Terminalia arjuna, which is a popular cardioprotectant and may prevent or slow progression of pathological hypertrophy to heart failure. Here, we investigated the mode of action of a principal bioactive T. arjuna compound, arjunolic acid (AA), in ameliorating hemodynamic load-induced cardiac fibrosis and identified its intracellular target. Our data revealed that AA significantly represses collagen expression and improves cardiac function during hypertrophy. We found that AA binds to and stabilizes the ligand-binding domain of peroxisome proliferator-activated receptor α (PPARα) and increases its expression during cardiac hypertrophy. PPARα knockdown during AA treatment in hypertrophy samples, including angiotensin II-treated adult cardiac fibroblasts and renal artery-ligated rat heart, suggests that AA-driven cardioprotection primarily arises from PPARα agonism. Moreover, AA-induced PPARα up-regulation leads to repression of TGF-ß signaling, specifically by inhibiting TGF-ß-activated kinase1 (TAK1) phosphorylation. We observed that PPARα directly interacts with TAK1, predominantly via PPARα N-terminal transactivation domain (AF-1) thereby masking the TAK1 kinase domain. The AA-induced PPARα-bound TAK1 level thereby shows inverse correlation with the phosphorylation level of TAK1 and subsequent reduction in p38 MAPK and NF-κBp65 activation, ultimately culminating in amelioration of excess collagen synthesis in cardiac hypertrophy. In conclusion, our findings unravel the mechanism of AA action in regressing hypertrophy-associated cardiac fibrosis by assigning a role of AA as a PPARα agonist that inactivates non-canonical TGF-ß signaling.


Asunto(s)
Cardiomegalia/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Miocardio/metabolismo , PPAR alfa/agonistas , Factor de Crecimiento Transformador beta/metabolismo , Triterpenos/farmacología , Animales , Cardiomegalia/patología , Colágeno/biosíntesis , Fibrosis , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Miocardio/patología , Ratas , Ratas Wistar , Factor de Transcripción ReIA/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
J Mol Recognit ; 31(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28961341

RESUMEN

The heparin-protein interaction plays a vital role in numerous physiological and pathological processes. Not only is the binding mechanism of these interactions poorly understood, studies concerning their therapeutic targeting are also limited. Here, we have studied the interaction of the heparin interacting peptide (HIP) from Tat (which plays important role in HIV infections) with heparin. Isothermal titration calorimetry binding exhibits distinct biphasic isotherm with two different affinities in the HIP-heparin complex formation. Overall, the binding was mainly driven by the nonionic interactions with a small contribution from ionic interactions. The stoichiometric analysis suggested that the minimal site for a single HIP molecule is a chain of 4 to 5 saccharide molecules, also supported by docking studies. The investigation was also focused on exploiting the possibility of using a small molecule as an inhibitor of the HIP-heparin complex. Quinacrine, because of its ability to mimic the HIP interactions with heparin, was shown to successfully modulate the HIP-heparin interactions. This result demonstrates the feasibility of inhibiting the disease relevant heparin-protein interactions by a small molecule, which could be an effective strategy for the development of future therapeutic agents.


Asunto(s)
Heparina/química , Fragmentos de Péptidos/química , Quinacrina/química , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química , Sitios de Unión , Simulación del Acoplamiento Molecular , Unión Proteica , Termodinámica
12.
Biochem J ; 474(1): 123-147, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803245

RESUMEN

Misfolding and aggregation of cellular prion protein is associated with a large array of neurological disorders commonly called the transmissible spongiform encephalopathies. Designing inhibitors against prions has remained a daunting task owing to limited information about mechanism(s) of their pathogenic self-assembly. Here, we explore the anti-prion properties of a combinatorial library of bispidine-based peptidomimetics (BPMs) that conjugate amino acids with hydrophobic and aromatic side chains. Keeping the bispidine unit unaltered, a series of structurally diverse BPMs were synthesized and tested for their prion-modulating properties. Administration of Leu- and Trp-BPMs delayed and completely inhibited the amyloidogenic conversion of human prion protein (HuPrP), respectively. We found that each BPM induced the HuPrP to form unique oligomeric nanostructures differing in their biophysical properties, cellular toxicities and response to conformation-specific antibodies. While Leu-BPMs were found to stabilize the oligomers, Trp-BPMs effected transient oligomerization, resulting in the formation of non-toxic, non-fibrillar aggregates. Yet another aromatic residue, Phe, however, accelerated the aggregation process in HuPrP. Molecular insights obtained through MD (molecular dynamics) simulations suggested that each BPM differently engages a conserved Tyr 169 residue at the α2-ß2 loop of HuPrP and affects the stability of α2 and α3 helices. Our results demonstrate that this new class of molecules having chemical scaffolds conjugating hydrophobic/aromatic residues could effectively modulate prion aggregation and toxicity.


Asunto(s)
Amiloide/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Nanoestructuras/química , Peptidomiméticos/química , Priones/química , Agregado de Proteínas , Anticuerpos/química , Humanos , Biblioteca de Péptidos , Estructura Secundaria de Proteína
13.
Biochim Biophys Acta ; 1864(7): 794-804, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27045222

RESUMEN

Certain amino acid stretches are considered 'critical' to trigger amyloidogenesis in a protein. Synthetic peptides corresponding to these stretches are often used as experimental mimics for studying the amyloidogenesis of their parent protein. Here we provide evidence that such simple extrapolation is misleading. We scrutinized each step of amyloid progression of full length bovine carbonic anhydrase (BCA) and compared it with the amyloidogenic process of its critical peptide stretch 201-227 (PepB). We found that under similar solution conditions amyloidogenesis of BCA followed surface-catalyzed secondary nucleation, whereas, that of PepB followed classical nucleation-dependent pathway. AFM images showed that while BCA formed short, thick and branched fibrils, PepB formed thin, long and unbranched fibrils. Structural information obtained by ATR-FTIR spectroscopy suggested parallel arrangement of intermolecular ß-sheet in BCA amyloids in contrast to PepB amyloids which arranged into antiparallel ß sheets. Amyloids formed by BCA were unable to seed the fibrillation of PepB and vice versa. Even the intermediates formed during lag phase revealed contrasting FTIR and far UV CD signature, hydrophobicity, morphology and cell cytotoxicity. Thus, we propose that sequences other than critical amyloidogenic stretches may significantly influence the initiation, polymerization and final fibrillar morphology of amyloid forming protein. The results have been discussed in light of primary sequence mediated amyloid polymorphism and its importance in the rational design of amyloid nanomaterials possessing desired physico-chemical properties.


Asunto(s)
Amiloide/química , Anhidrasas Carbónicas/química , Secuencia de Aminoácidos , Amiloide/ultraestructura , Animales , Anhidrasas Carbónicas/ultraestructura , Bovinos , Microscopía de Fuerza Atómica , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Análisis de Secuencia de Proteína , Espectroscopía Infrarroja por Transformada de Fourier
14.
Biochim Biophys Acta Proteins Proteom ; 1865(5): 589-603, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28238838

RESUMEN

Parkinson's disease is characterized by the presence of insoluble and neurotoxic aggregates (amyloid fibrils) of an intrinsically disordered protein α-synuclein. In this study we have examined the effects of four naturally occurring polyphenols in combination with ß-cyclodextrin (ß-CD) on the aggregation of α-synuclein in the presence of macromolecular crowding agents. Our results reveal that even at sub-stoichiometric concentrations of the individual components, the polyphenol-ß-CD combination(s) not only inhibited the aggregation of the proteins but was also effective in disaggregating preformed fibrils. Curcumin was found to be the most efficient, followed by baicalein with (-)-epigallocatechin gallate and resveratrol coming in next, the latter two exhibiting very similar effects. Our results suggest that the efficiency of curcumin results from a balanced composition of the phenolic OH groups, benzene rings and flexibility. The latter ensures proper positioning of the functional groups to maximize the underlying interactions with both the monomeric form of α-synuclein and its aggregates. The uniqueness of ß-CD was reinforced by the observation that none of the other cyclodextrin variants [α-CD and HP-ß-CD] used was as effective, in spite of these possessing better water solubility. Moreover, the fact that the combinations remained effective under conditions of macromolecular crowding suggests that these have the potential to be developed into viable drug compositions in the near future. MTT assays on cell viability independently confirmed this hypothesis wherein these combinations (and the polyphenols alone too) appreciably impeded the toxicity of the prefibrillar α-synuclein aggregates on the mouse neuroblastoma cell lines (N2a cells).


Asunto(s)
Proteínas Amiloidogénicas/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Polifenoles/administración & dosificación , alfa-Sinucleína/metabolismo , beta-Ciclodextrinas/administración & dosificación , Amiloide/química , Amiloide/efectos de los fármacos , Amiloide/metabolismo , Proteínas Amiloidogénicas/química , Animales , Catequina/análogos & derivados , Catequina/química , Línea Celular , Supervivencia Celular , Dicroismo Circular , Curcumina/administración & dosificación , Curcumina/química , Humanos , Ratones , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Polifenoles/química , Agregación Patológica de Proteínas/tratamiento farmacológico , Agregación Patológica de Proteínas/patología , alfa-Sinucleína/química , beta-Ciclodextrinas/química
15.
Arch Biochem Biophys ; 622: 36-46, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28461187

RESUMEN

In obligate dimeric proteins of hyperthermophilic origin the question whether the native dimer is obtained by association of folded monomers or through concomitant folding and assembly of subunits has intrigued researchers. To find an answer we studied the folding of a dimeric enzyme l-asparaginase from Pyrococcus furiosus (PfA) for which we reported earlier that it unfolds cooperatively without populating folded monomeric intermediates. However, in the present study we report the finding of a folded monomeric intermediate of PfA under acidic condition. This monomer, although inactive, displayed secondary and tertiary structural features identical to the native protein and re-assembled to active dimeric form upon reversal of pH. The monomer is conformationally flexible and thermodynamically and kinetically less stable than the native dimer. Interestingly, when incubated at 60 °C the folded monomer, with exposed ANS-binding hydrophobic surfaces, spontaneously converted to amyloid fibrils. On the basis of our data we propose that PfA directly assembles into a multimeric form perhaps as an evolutionary adaptation to avoid accumulation of aggregation prone monomeric intermediates.


Asunto(s)
Amiloide/metabolismo , Asparaginasa/metabolismo , Pliegue de Proteína , Pyrococcus furiosus/enzimología , Amiloide/química , Asparaginasa/química , Estabilidad de Enzimas , Cinética , Modelos Moleculares , Multimerización de Proteína , Pyrococcus furiosus/química , Pyrococcus furiosus/metabolismo , Termodinámica
16.
Biochim Biophys Acta ; 1854(5): 426-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25666897

RESUMEN

Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease that affects motor neurons. Unfortunately, effective therapeutics against this disease is still not available. Almost 20% of familial ALS (fALS) is suggested to be associated with pathological deposition of superoxide dismutase (SOD1). Evidences suggest that SOD1-containing pathological inclusions in ALS exhibit amyloid like properties. An effective strategy to combat ALS may be to inhibit amyloid formation of SOD1 using small molecules. In the present study, we observed the fibrillation of one of the premature forms of SOD1 (SOD1 with reduced disulfide) in the presence of curcumin. Using ThT binding assay, AFM, TEM images and FTIR, we demonstrate that curcumin inhibits the DTT-induced fibrillation of SOD1 and favors the formation of smaller and disordered aggregates of SOD1. The enhancement in curcumin fluorescence on the addition of oligomers and pre-fibrillar aggregates of SOD1 suggests binding of these species to curcumin. Docking studies indicate that putative binding site of curcumin may be the amyloidogenic regions of SOD1. Further, there is a significant increase in SOD1 mediated toxicity in the regime of pre-fibrillar and fibrillar aggregates which is not evident in curcumin containing samples. All these data suggest that curcumin reduces toxicity by binding to the amyloidogenic regions of the species on the aggregation pathway and blocking the formation of the toxic species. Nanoparticles of curcumin with higher aqueous solubility show similar aggregation control as that of curcumin bulk. This suggests a potential role for curcumin in the treatment of ALS.


Asunto(s)
Amiloide/efectos de los fármacos , Amiloide/toxicidad , Curcumina/farmacocinética , Agregado de Proteínas/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Amiloide/química , Amiloide/metabolismo , Células Cultivadas , Curcumina/química , Citoprotección/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Unión Proteica , Multimerización de Proteína/efectos de los fármacos , Superóxido Dismutasa/química , Superóxido Dismutasa-1
17.
Extremophiles ; 19(3): 681-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25862541

RESUMEN

Here, we report the folding and assembly of a Pyrococcus furiosus-derived protein, L-asparaginase (PfA). PfA functions as a homodimer, with each monomer made of distinct N- and C-terminal domains. The purified individual domains as well as single Trp mutant of each domain were subjected to chemical denaturation/renaturation and probed by combination of spectroscopic, chromatographic, quenching and scattering techniques. We found that the N-domain acts like a folding scaffold and assists the folding of remaining polypeptide. The domains displayed sequential folding with the N-domain having higher thermodynamic stability. We report that the extreme thermal stability of PfA is due to the presence of high intersubunit associative forces supported by extensive H-bonding and ionic interactions network. Our results proved that folding cooperativity in a thermophilic, multisubunit protein is dictated by concomitant folding and association of constituent domains directly into a native quaternary structure. This report gives an account of the factors responsible for folding and stability of a therapeutically and industrially important protein.


Asunto(s)
Asparaginasa/química , Proteínas Bacterianas/química , Pliegue de Proteína , Multimerización de Proteína , Pyrococcus furiosus/enzimología , Secuencia de Aminoácidos , Estabilidad de Enzimas , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Subunidades de Proteína/química
18.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 12): 3187-97, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25478837

RESUMEN

Covalent linkers bridging the domains of multidomain proteins are considered to be crucial for assembly and function. In this report, an exception in which the linker of a two-domain dimeric L-asparaginase from Pyrococcus furiosus (PfA) was found to be dispensable is presented. Domains of this enzyme assembled without the linker into a conjoined tetrameric form that exhibited higher activity than the parent enzyme. The global shape and quaternary structure of the conjoined PfA were also similar to the wild-type PfA, as observed by their solution scattering profiles and X-ray crystallographic data. Comparison of the crystal structures of substrate-bound and unbound enzymes revealed an altogether new active-site composition and mechanism of action. Thus, conjoined PfA is presented as a unique enzyme obtained through noncovalent, linker-less assembly of constituent domains that is stable enough to function efficiently at elevated temperatures.


Asunto(s)
Asparaginasa/química , Pyrococcus furiosus/enzimología , Secuencia de Aminoácidos , Asparaginasa/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Modelos Moleculares , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Terciaria de Proteína , Pyrococcus furiosus/química , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Difracción de Rayos X
19.
ACS Infect Dis ; 10(6): 1990-2001, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38815059

RESUMEN

Conserved molecular signatures in multidrug-resistant Salmonella typhi can serve as novel therapeutic targets for mitigation of infection. In this regard, we present the S. typhi cell division activator protein (StCAP) as a conserved target across S. typhi variants. From in silico and fluorimetric assessments, we found that StCAP is a DNA-binding protein. Replacement of the identified DNA-interacting residue Arg34 of StCAP with Ala34 showed a dramatic (15-fold) increase in Kd value compared to the wild type (Kd 546 nm) as well as a decrease in thermal stability (10 °C shift). Out of the two screened molecules against the DNA-binding pocket of StCAP, eltrombopag, and nilotinib, the former displayed better binding. Eltrombopag inhibited the stand-alone S. typhi culture with an IC50 of 38 µM. The effect was much more pronounced on THP-1-derived macrophages (T1Mac) infected with S. typhi where colony formation was severely hindered with IC50 reduced further to 10 µM. Apoptotic protease activating factor1 (Apaf1), a key molecule for intrinsic apoptosis, was identified as an StCAP-interacting partner by pull-down assay against T1Mac. Further, StCAP-transfected T1Mac showed a significant increase in LC3 II (autophagy marker) expression and downregulation of caspase 3 protein. From these experiments, we conclude that StCAP provides a crucial survival advantage to S. typhi during infection, thereby making it a potent alternative therapeutic target.


Asunto(s)
Proteínas Bacterianas , Salmonella typhi , Salmonella typhi/efectos de los fármacos , Salmonella typhi/genética , Salmonella typhi/patogenicidad , Humanos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Apoptosis/efectos de los fármacos , Macrófagos/microbiología , Macrófagos/efectos de los fármacos , Células THP-1 , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Autofagia/efectos de los fármacos , Fiebre Tifoidea/microbiología , División Celular/efectos de los fármacos
20.
FASEB J ; 26(3): 1161-71, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22166247

RESUMEN

Thermophilic l-asparaginases display high stability and activity at elevated temperatures. However, they are of limited use in leukemia therapy because of their low substrate affinity and reduced activity under physiological conditions. In an attempt to combine stability with activity at physiological conditions, 3 active-site mutants of Pyrococcus furiosus l-asparaginase (PfA) were developed. The mutants, specifically K274E, showed improved enzymatic properties at physiological conditions as compared to the wild type. All variants were thermodynamically stable and resistant to proteolytic digestion. None of the enzymes displayed glutaminase activity, a highly desirable therapeutic property. All variants showed higher and significant killing of human cell lines HL60, MCF7, and K562 as compared to the Escherichia coli l-asparaginase. Our study revealed that increased substrate accessibility through the active site loop plays a major role in determining activity. A new mechanistic insight has been proposed based on molecular dynamics simulated structures, where dynamic flipping of a critical Tyr residue is responsible for the activity of thermophilic l-asparaginases. Our study not only resulted in development of PfA mutants with combination of desirable properties but also gave a mechanistic insight about their activity.


Asunto(s)
Proteínas Arqueales/genética , Asparaginasa/genética , Dominio Catalítico/genética , Mutación , Secuencia de Aminoácidos , Antineoplásicos/química , Antineoplásicos/farmacología , Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Asparaginasa/metabolismo , Asparaginasa/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dicroismo Circular , Relación Dosis-Respuesta a Droga , Estabilidad de Enzimas , Células HL-60 , Calor , Humanos , Células K562 , Cinética , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Pyrococcus furiosus/enzimología , Pyrococcus furiosus/genética , Homología de Secuencia de Aminoácido , Espectrometría de Fluorescencia , Especificidad por Sustrato , Termodinámica , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA