Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(25): e2304128120, 2023 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-37307464

RESUMEN

Acquired ribosomal RNA (rRNA) methylation has emerged as a significant mechanism of aminoglycoside resistance in pathogenic bacterial infections. Modification of a single nucleotide in the ribosome decoding center by the aminoglycoside-resistance 16S rRNA (m7G1405) methyltransferases effectively blocks the action of all 4,6-deoxystreptamine ring-containing aminoglycosides, including the latest generation of drugs. To define the molecular basis of 30S subunit recognition and G1405 modification by these enzymes, we used a S-adenosyl-L-methionine analog to trap the complex in a postcatalytic state to enable determination of a global 3.0 Å cryo-electron microscopy structure of the m7G1405 methyltransferase RmtC bound to the mature Escherichia coli 30S ribosomal subunit. This structure, together with functional analyses of RmtC variants, identifies the RmtC N-terminal domain as critical for recognition and docking of the enzyme on a conserved 16S rRNA tertiary surface adjacent to G1405 in 16S rRNA helix 44 (h44). To access the G1405 N7 position for modification, a collection of residues across one surface of RmtC, including a loop that undergoes a disorder-to order transition upon 30S subunit binding, induces significant distortion of h44. This distortion flips G1405 into the enzyme active site where it is positioned for modification by two almost universally conserved RmtC residues. These studies expand our understanding of ribosome recognition by rRNA modification enzymes and present a more complete structural basis for future development of strategies to inhibit m7G1405 modification to resensitize bacterial pathogens to aminoglycosides.


Asunto(s)
Aminoglicósidos , Antibacterianos , ARN Ribosómico 16S , Microscopía por Crioelectrón , Metiltransferasas , ARN Ribosómico , Escherichia coli
2.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35017296

RESUMEN

The 2'-5'-oligoadenylate synthetases (OAS) are innate immune sensors of cytosolic double-stranded RNA (dsRNA) that play a critical role in limiting viral infection. How these proteins are able to avoid aberrant activation by cellular RNAs is not fully understood, but adenosine-to-inosine (A-to-I) editing has been proposed to limit accumulation of endogenous RNAs that might otherwise cause stimulation of the OAS/RNase L pathway. Here, we aim to uncover whether and how such sequence modifications can restrict the ability of short, defined dsRNAs to activate the single-domain form of OAS, OAS1. Unexpectedly, we find that all tested inosine-containing dsRNAs have an increased capacity to activate OAS1, whether in a destabilizing (I•U) or standard Watson-Crick-like base pairing (I-C) context. Additional variants with strongly destabilizing A•C mismatches or stabilizing G-C pairs also exhibit increased capacity to activate OAS1, eliminating helical stability as a factor in the relative ability of the dsRNAs to activate OAS1. Using thermal difference spectra and molecular dynamics simulations, we identify both increased helical dynamics and specific local changes in helical structure as important factors in the capacity of short dsRNAs to activate OAS1. These helical features may facilitate more ready adoption of the distorted OAS1-bound conformation or stabilize important structures to predispose the dsRNA for optimal binding and activation of OAS1. These studies thus reveal the molecular basis for the greater capacity of some short dsRNAs to activate OAS1 in a sequence-independent manner.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/química , 2',5'-Oligoadenilato Sintetasa/metabolismo , Disparidad de Par Base , ARN Bicatenario/metabolismo , Secuencia de Bases , Endorribonucleasas/metabolismo , Activación Enzimática , Humanos , Inosina/metabolismo , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Edición de ARN , Estabilidad del ARN , Relación Estructura-Actividad , Temperatura
3.
Proc Natl Acad Sci U S A ; 119(14): e2120352119, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35357969

RESUMEN

Changes in bacterial ribosomal RNA (rRNA) methylation status can alter the activity of diverse groups of ribosome-targeting antibiotics. These modifications are typically incorporated by a single methyltransferase that acts on one nucleotide target and rRNA methylation directly prevents drug binding, thereby conferring drug resistance. Loss of intrinsic methylation can also result in antibiotic resistance. For example, Mycobacterium tuberculosis becomes sensitized to tuberactinomycin antibiotics, such as capreomycin and viomycin, due to the action of the intrinsic methyltransferase TlyA. TlyA is unique among antibiotic resistance-associated methyltransferases as it has dual 16S and 23S rRNA substrate specificity and can incorporate cytidine-2'-O-methylations within two structurally distinct contexts. Here, we report the structure of a mycobacterial 50S subunit-TlyA complex trapped in a postcatalytic state with a S-adenosyl-L-methionine analog using single-particle cryogenic electron microscopy. Together with complementary functional analyses, this structure reveals critical roles in 23S rRNA substrate recognition for conserved residues across an interaction surface that spans both TlyA domains. These interactions position the TlyA active site over the target nucleotide C2144, which is flipped from 23S Helix 69 in a process stabilized by stacking of TlyA residue Phe157 on the adjacent A2143. Base flipping may thus be a common strategy among rRNA methyltransferase enzymes, even in cases where the target site is accessible without such structural reorganization. Finally, functional studies with 30S subunit suggest that the same TlyA interaction surface is employed to recognize this second substrate, but with distinct dependencies on essential conserved residues.


Asunto(s)
Proteínas Bacterianas , Metiltransferasas , Mycobacterium tuberculosis , Subunidades Ribosómicas Grandes Bacterianas , Proteínas Bacterianas/química , Dominio Catalítico , Farmacorresistencia Bacteriana/genética , Metiltransferasas/química , Mycobacterium tuberculosis/enzimología , Mycobacterium tuberculosis/genética , Conformación Proteica en Hélice alfa , ARN Ribosómico 16S/química , ARN Ribosómico 23S/química , Subunidades Ribosómicas Grandes Bacterianas/química
4.
J Biol Chem ; 298(10): 102393, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988649

RESUMEN

The SpoU-TrmD (SPOUT) methyltransferase superfamily was designated when structural similarity was identified between the transfer RNA-modifying enzymes TrmH (SpoU) and TrmD. SPOUT methyltransferases are found in all domains of life and predominantly modify transfer RNA or ribosomal RNA substrates, though one instance of an enzyme with a protein substrate has been reported. Modifications placed by SPOUT methyltransferases play diverse roles in regulating cellular processes such as ensuring translational fidelity, altering RNA stability, and conferring bacterial resistance to antibiotics. This large collection of S-adenosyl-L-methionine-dependent methyltransferases is defined by a unique α/ß fold with a deep trefoil knot in their catalytic (SPOUT) domain. Herein, we describe current knowledge of SPOUT enzyme structure, domain architecture, and key elements of catalytic function, including S-adenosyl-L-methionine co-substrate binding, beginning with a new sequence alignment that divides the SPOUT methyltransferase superfamily into four major clades. Finally, a major focus of this review will be on our growing understanding of how these diverse enzymes accomplish the molecular feat of specific substrate recognition and modification, as highlighted by recent advances in our knowledge of protein-RNA complex structures and the discovery of the dependence of one SPOUT methyltransferase on metal ion binding for catalysis. Considering the broad biological roles of RNA modifications, developing a deeper understanding of the process of substrate recognition by the SPOUT enzymes will be critical for defining many facets of fundamental RNA biology with implications for human disease.


Asunto(s)
Metiltransferasas , ARNt Metiltransferasas , Humanos , Metiltransferasas/química , Metiltransferasas/metabolismo , Modelos Moleculares , ARN de Transferencia/metabolismo , S-Adenosilmetionina/metabolismo , Especificidad por Sustrato , ARNt Metiltransferasas/química , ARNt Metiltransferasas/metabolismo
5.
J Mol Evol ; 91(2): 225-235, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36869271

RESUMEN

Chlorophyllide a oxygenase (CAO) is responsible for converting chlorophyll a to chlorophyll b in a two-step oxygenation reaction. CAO belongs to the family of Rieske-mononuclear iron oxygenases. Although the structure and reaction mechanism of other Rieske monooxygenases have been described, a member of plant Rieske non-heme iron-dependent monooxygenase has not been structurally characterized. The enzymes in this family usually form a trimeric structure and electrons are transferred between the non-heme iron site and the Rieske center of the adjoining subunits. CAO is supposed to form a similar structural arrangement. However, in Mamiellales such as Micromonas and Ostreococcus, CAO is encoded by two genes where non-heme iron site and Rieske cluster localize on the distinct polypeptides. It is not clear if they can form a similar structural organization to achieve the enzymatic activity. In this study, the tertiary structures of CAO from the model plant Arabidopsis thaliana and the Prasinophyte Micromonas pusilla were predicted by deep learning-based methods, followed by energy minimization and subsequent stereochemical quality assessment of the predicted models. Furthermore, the chlorophyll a binding cavity and the interaction of ferredoxin, which is the electron donor, on the surface of Micromonas CAO were predicted. The electron transfer pathway was predicted in Micromonas CAO and the overall structure of the CAO active site was conserved even though it forms a heterodimeric complex. The structures presented in this study will serve as a basis for understanding the reaction mechanism and regulation of the plant monooxygenase family to which CAO belongs.


Asunto(s)
Arabidopsis , Clorofilidas , Chlorophyta , Clorofilidas/metabolismo , Clorofila A/metabolismo , Oxigenasas/genética , Oxigenasas/química , Oxigenasas/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Oxigenasas de Función Mixta/metabolismo , Plantas , Chlorophyta/metabolismo , Hierro/metabolismo
6.
J Chem Inf Model ; 61(9): 4442-4454, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34516120

RESUMEN

The emergence of new viral infections and drug-resistant bacteria urgently necessitates expedient therapeutic development. Repurposing and redesign of existing drugs against different targets are one potential way in which to accelerate this process. Suramin was initially developed as a successful antiparasitic drug but has also shown promising antiviral and antibacterial activities. However, due to its high conformational flexibility and negative charge, suramin is considered quite promiscuous toward positively charged sites within nucleic acid binding proteins. Although some suramin analogs have been developed against specific targets, only limited structure-activity relationship studies were performed, and virtual screening has yet to be used to identify more specific inhibitor(s) based on its scaffold. Using available structures, we investigated suramin's target diversity, confirming that suramin preferentially binds to protein pockets that are both positively charged and enriched in aromatic or leucine residues. Further, suramin's high conformational flexibility allows adaptation to structurally diverse binding surfaces. From this platform, we developed a framework for structure- and docking-guided elaboration of suramin analog scaffolds using virtual screening of suramin and heparin analogs against a panel of diverse therapeutically relevant viral and bacterial protein targets. Use of this new framework to design potentially specific suramin analogs is exemplified using the SARS-CoV-2 RNA-dependent RNA polymerase and nucleocapsid protein, identifying leads that might inhibit a wide range of coronaviruses. The approach presented here establishes a computational framework for designing suramin analogs against different bacterial and viral targets and repurposing existing drugs for more specific inhibitory activity.


Asunto(s)
COVID-19 , Suramina , Antibacterianos/farmacología , Antivirales/farmacología , Humanos , ARN Viral , SARS-CoV-2 , Suramina/farmacología
7.
J Bacteriol ; 202(20)2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32747430

RESUMEN

Pseudomonas aeruginosa isolates from chronic lung infections often overproduce alginate, giving rise to the mucoid phenotype. Isolation of mucoid strains from chronic lung infections correlates with a poor patient outcome. The most common mutation that causes the mucoid phenotype is called mucA22 and results in a truncated form of the anti-sigma factor MucA that is continuously subjected to proteolysis. When a functional MucA is absent, the cognate sigma factor, AlgT, is no longer sequestered and continuously transcribes the alginate biosynthesis operon, leading to alginate overproduction. In this work, we report that in the absence of wild-type MucA, providing exogenous AlgT is toxic. This is intriguing, since mucoid strains endogenously possess high levels of AlgT. Furthermore, we show that suppressors of toxic AlgT production have mutations in mucP, a protease involved in MucA degradation, and provide the first atomistic model of MucP. Based on our findings, we speculate that mutations in mucP stabilize the truncated form of MucA22, rendering it functional and therefore able to reduce toxicity by properly sequestering AlgT.IMPORTANCEPseudomonas aeruginosa is an opportunistic bacterial pathogen capable of causing chronic lung infections. Phenotypes important for the long-term persistence and adaption to this unique lung ecosystem are largely regulated by the AlgT sigma factor. Chronic infection isolates often contain mutations in the anti-sigma factor mucA, resulting in uncontrolled AlgT and continuous production of alginate in addition to the expression of ∼300 additional genes. Here, we report that in the absence of wild-type MucA, AlgT overproduction is lethal and that suppressors of toxic AlgT production have mutations in the MucA protease, MucP. Since AlgT contributes to the establishment of chronic infections, understanding how AlgT is regulated will provide vital information on how P. aeruginosa is capable of causing long-term infections.


Asunto(s)
Alginatos/metabolismo , Proteínas Bacterianas/genética , Pseudomonas aeruginosa/genética , Factor sigma/genética , Regulación Bacteriana de la Expresión Génica , Mutación , Operón/genética , Fenotipo , Proteolisis , Pseudomonas aeruginosa/aislamiento & purificación
8.
J Biol Chem ; 294(52): 20109-20121, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31753919

RESUMEN

The opportunistic bacterial pathogen Pseudomonas aeruginosa is a leading cause of serious infections in individuals with cystic fibrosis, compromised immune systems, or severe burns. P. aeruginosa adhesion to host epithelial cells is enhanced by surface-exposed translation elongation factor EF-Tu carrying a Lys-5 trimethylation, incorporated by the methyltransferase EftM. Thus, the EF-Tu modification by EftM may represent a target to prevent P. aeruginosa infections in vulnerable individuals. Here, we extend our understanding of EftM activity by defining the molecular mechanism by which it recognizes EF-Tu. Acting on the observation that EftM can bind to EF-Tu lacking its N-terminal peptide (encompassing the Lys-5 target site), we generated an EftM homology model and used it in protein/protein docking studies to predict EftM/EF-Tu interactions. Using site-directed mutagenesis of residues in both proteins, coupled with binding and methyltransferase activity assays, we experimentally validated the predicted protein/protein interface. We also show that EftM cannot methylate the isolated N-terminal EF-Tu peptide and that binding-induced conformational changes in EftM are likely needed to enable placement of the first 5-6 amino acids of EF-Tu into a conserved peptide-binding channel in EftM. In this channel, a group of residues that are highly conserved in EftM proteins position the N-terminal sequence to facilitate Lys-5 modification. Our findings reveal that EftM employs molecular strategies for substrate recognition common among both class I (Rossmann fold) and class II (SET domain) methyltransferases and pave the way for studies seeking a deeper understanding of EftM's mechanism of action on EF-Tu.


Asunto(s)
Proteínas Bacterianas/metabolismo , Metiltransferasas/metabolismo , Pseudomonas aeruginosa/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Evolución Molecular , Metiltransferasas/química , Metiltransferasas/genética , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Factor Tu de Elongación Peptídica/química , Factor Tu de Elongación Peptídica/genética , Factor Tu de Elongación Peptídica/metabolismo , Unión Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Alineación de Secuencia , Especificidad por Sustrato
9.
J Biol Chem ; 294(46): 17642-17653, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31594862

RESUMEN

Methylation of the small ribosome subunit rRNA in the ribosomal decoding center results in exceptionally high-level aminoglycoside resistance in bacteria. Enzymes that methylate 16S rRNA on N7 of nucleotide G1405 (m7G1405) have been identified in both aminoglycoside-producing and clinically drug-resistant pathogenic bacteria. Using a fluorescence polarization 30S-binding assay and a new crystal structure of the methyltransferase RmtC at 3.14 Å resolution, here we report a structure-guided functional study of 30S substrate recognition by the aminoglycoside resistance-associated 16S rRNA (m7G1405) methyltransferases. We found that the binding site for these enzymes in the 30S subunit directly overlaps with that of a second family of aminoglycoside resistance-associated 16S rRNA (m1A1408) methyltransferases, suggesting that both groups of enzymes may exploit the same conserved rRNA tertiary surface for docking to the 30S. Within RmtC, we defined an N-terminal domain surface, comprising basic residues from both the N1 and N2 subdomains, that directly contributes to 30S-binding affinity. In contrast, additional residues lining a contiguous adjacent surface on the C-terminal domain were critical for 16S rRNA modification but did not directly contribute to the binding affinity. The results from our experiments define the critical features of m7G1405 methyltransferase-substrate recognition and distinguish at least two distinct, functionally critical contributions of the tested enzyme residues: 30S-binding affinity and stabilizing a binding-induced 16S rRNA conformation necessary for G1405 modification. Our study sets the scene for future high-resolution structural studies of the 30S-methyltransferase complex and for potential exploitation of unique aspects of substrate recognition in future therapeutic strategies.


Asunto(s)
Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana , Metiltransferasas/metabolismo , Proteus mirabilis/enzimología , Subunidades Ribosómicas Pequeñas Bacterianas/metabolismo , Aminoglicósidos/metabolismo , Aminoglicósidos/farmacología , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/química , Cristalografía por Rayos X , Humanos , Metiltransferasas/química , Modelos Moleculares , Conformación Proteica , Infecciones por Proteus/tratamiento farmacológico , Infecciones por Proteus/microbiología , Proteus mirabilis/efectos de los fármacos , Proteus mirabilis/metabolismo , ARN Ribosómico 16S/metabolismo , Subunidades Ribosómicas Pequeñas Bacterianas/química , Especificidad por Sustrato
10.
Artículo en Inglés | MEDLINE | ID: mdl-32457110

RESUMEN

Resistance-nodulation-division (RND) efflux pumps are important contributors to bacterial antibiotic resistance. In this study, we combined evolutionary sequence analyses, computational structural modeling, and ligand docking to develop a framework that can explain the known antibiotic substrate selectivity differences between two Pseudomonas aeruginosa RND transporters, MexY and MexB. For efficient efflux, antibiotic substrates must possess a "Goldilocks affinity": binding strong enough to allow interaction with transporter but not so tight as to impede movement through the pump.


Asunto(s)
Antibacterianos , Pseudomonas aeruginosa , Antibacterianos/farmacología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Farmacorresistencia Bacteriana , Proteínas de Transporte de Membrana/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
11.
Mol Phylogenet Evol ; 107: 356-366, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27894997

RESUMEN

Nucleoid-associated proteins (NAPs) are chromosome-organizing factors, which affect the transcriptional landscape of a bacterial cell. HU is an NAP, which binds to DNA with a broad specificity while homologous IHF (Integration Host Factor), binds DNA with moderately higher specificity. Specificity and differential binding affinity of HU/IHF proteins towards their target binding sites play a crucial role in their regulatory dynamics. Decades of biochemical and genomic studies have been carried out for HU and IHF like proteins. Yet, questions related to their DNA binding specificity, and differential ability to bend DNA thus affecting the binding site length remained unanswered. In addition, the problem has not been investigated from an evolutionary perspective. Our phylogenetic analysis revealed three major clades belonging to HU, IHFα and IHFß like proteins with reference to E. coli. We carried out a comparative analysis of three-dimensional structures of HU/IHF proteins to gain insight into the structural basis of clade division. The present study revealed three major features which contribute to differential DNA binding specificity of HU/IHF proteins, (I) conformational restriction of DNA binding residues due to salt-bridge formation, (II) the enrichment of alanine in the DNA binding site increasing conformational space of flexible side chains in its vicinity and (III) nature of DNA binding residue (Arg to Lys bias in different clades) which interacts differentially to DNA bases. We observed an extended electropositive surface at the DNA draping site for IHF clade proteins compared to HU, which stabilizes the DNA bend. Differences in the dimer stabilization strategies between HU and IHF were also observed. Our analysis reveals a comprehensive evolutionary picture, which rationalizes the origin of multi-specificity of HU/IHF proteins using sequence and structure-based determinants, which could also be applied to understand differences in binding specificities of other nucleic acid binding proteins.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , ADN Bacteriano/metabolismo , Evolución Molecular , Factores de Integración del Huésped/química , Factores de Integración del Huésped/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Secuencia de Bases , Sitios de Unión , Secuencia Conservada , Escherichia coli/genética , Filogenia , Unión Proteica , Multimerización de Proteína , Estabilidad Proteica , Sales (Química)/química
12.
Langmuir ; 31(50): 13600-8, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26597225

RESUMEN

A study on self-assembly of anisotropically substituted penta-aryl fullerenes in water has been reported. The penta-phenol-substituted amphiphilic fullerene derivative [C60Ph5(OH)5] exhibited self-assembled vesicular nanostructures in water under the experimental conditions. The size of the vesicles was observed to depend upon the kinetics of self-assembly and could be varied from ∼300 to ∼70 nm. Our mechanistic study indicated that the self-assembly of C60Ph5(OH)5 is driven by extensive intermolecular as well as water-mediated hydrogen bonding along with fullerene-fullerene hydrophobic interaction in water. The cumulative effect of these interactions is responsible for the stability of vesicular structures even on the removal of solvent. The substitution of phenol with anisole resulted in different packing and interaction of the fullerene derivative, as indicated in the molecular dynamics studies, thus resulting in different self-assembled nanostructures. The hollow vesicles were further encapsulated with a hydrophobic conjugated polymer and water-soluble dye as guest molecules. Such confinement of π-conjugated polymers in fullerene has significance in bulk heterojunction devices for efficient exciton diffusion.

13.
J Biomol Struct Dyn ; 41(13): 6236-6248, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35916030

RESUMEN

Defensins are antimicrobial peptides consisting of intramolecular disulphide bonds in a complex folded arrangement of two or three antiparallel ß-sheets with or without an α-helical structure. They are produced by a vast range of organisms being constitutively expressed or induced in various tissues against different stimuli like infection, injury or other inflammatory factors. Two classes of invertebrate defensin exist, namely CS-αß and big defensin, the latter being predominantly present in molluscs. Intriguingly, an invertebrate big defensin gene has been hypothesized as the most probable ancestor of vertebrate ß-defensins. Here, conserved residues were identified for both big defensin and ß-defensin. In silico mutation on conserved amino acid positions of the ß-defensin-like domain of big defensin from Crassostrea gigas was carried out to understand the effects of mutation on the structure and function of the protein. R64A and E71A have been identified as deleterious as well as destabilizing for the protein. Changes in amino acid network and aggregation propensity were also observed upon mutating these two charged residues. 100 ns molecular dynamics simulations of wild-type, R64A and E71A structures revealed significant conformational changes in the case of mutants. Furthermore, molecular docking highlighted the significance of R64 in ligand interaction. In conclusion, these results provide the first in-depth understanding of the structural and functional importance imparted by two conserved charged residues in the C-terminal region of big defensin. It also enhances the existing knowledge about this antimicrobial peptide for application in therapeutics and other aspects of protein engineering.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Ostreidae , beta-Defensinas , Animales , beta-Defensinas/genética , beta-Defensinas/química , Secuencia de Aminoácidos , Defensinas/química , Simulación del Acoplamiento Molecular , Aminoácidos , Ostreidae/metabolismo
14.
bioRxiv ; 2023 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-37425949

RESUMEN

The Resistance-Nodulation-Division (RND) efflux pump superfamily is pervasive among Gram-negative pathogens and contributes extensively to clinical antibiotic resistance. The opportunistic pathogen Pseudomonas aeruginosa contains 12 RND-type efflux systems, with four contributing to resistance including MexXY-OprM which is uniquely able to export aminoglycosides. At the site of initial substrate recognition, small molecule probes of the inner membrane transporter (e.g., MexY) have potential as important functional tools to understand substrate selectivity and a foundation for developing adjuvant efflux pump inhibitors (EPIs). Here, we optimized the scaffold of berberine, a known but weak MexY EPI, using an in-silico high-throughput screen to identify di-berberine conjugates with enhanced synergistic action with aminoglycosides. Further, docking and molecular dynamics simulations of di-berberine conjugates reveal unique contact residues and thus sensitivities of MexY from distinct P. aeruginosa strains. This work thereby reveals di-berberine conjugates to be useful probes of MexY transporter function and potential leads for EPI development.

15.
RSC Med Chem ; 14(4): 624-643, 2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37122541

RESUMEN

The rise of multidrug-resistant bacterial infections is a cause of global concern. There is an urgent need to both revitalize antibacterial agents that are ineffective due to resistance while concurrently developing new antibiotics with novel targets and mechanisms of action. Pathogen associated resistance-conferring ribosomal RNA (rRNA) methyltransferases are a growing threat that, as a group, collectively render a total of seven clinically-relevant ribosome-targeting antibiotic classes ineffective. Increasing frequency of identification and their growing prevalence relative to other resistance mechanisms suggests that these resistance determinants are rapidly spreading among human pathogens and could contribute significantly to the increased likelihood of a post-antibiotic era. Herein, with a view toward stimulating future studies to counter the effects of these rRNA methyltransferases, we summarize their prevalence, the fitness cost(s) to bacteria of their acquisition and expression, and current efforts toward targeting clinically relevant enzymes of this class.

16.
bioRxiv ; 2023 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-36993224

RESUMEN

Acquired ribosomal RNA (rRNA) methylation has emerged as a significant mechanism of aminoglycoside resistance in pathogenic bacterial infections. Modification of a single nucleotide in the ribosome decoding center by the aminoglycoside-resistance 16S rRNA (m 7 G1405) methyltransferases effectively blocks the action of all 4,6-deoxystreptamine ring-containing aminoglycosides, including the latest generation of drugs. To define the molecular basis of 30S subunit recognition and G1405 modification by these enzymes, we used a S-adenosyl-L-methionine (SAM) analog to trap the complex in a post-catalytic state to enable determination of an overall 3.0 Å cryo-electron microscopy structure of the m 7 G1405 methyltransferase RmtC bound to the mature Escherichia coli 30S ribosomal subunit. This structure, together with functional analyses of RmtC variants, identifies the RmtC N-terminal domain as critical for recognition and docking of the enzyme on a conserved 16S rRNA tertiary surface adjacent to G1405 in 16S rRNA helix 44 (h44). To access the G1405 N7 position for modification, a collection of residues across one surface of RmtC, including a loop that undergoes a disorder to order transition upon 30S subunit binding, induces significant distortion of h44. This distortion flips G1405 into the enzyme active site where it is positioned for modification by two almost universally conserved RmtC residues. These studies expand our understanding of ribosome recognition by rRNA modification enzymes and present a more complete structural basis for future development of strategies to inhibit m 7 G1405 modification to re-sensitize bacterial pathogens to aminoglycosides.

17.
Indian J Otolaryngol Head Neck Surg ; 74(1): 45-49, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33102186

RESUMEN

Introduction: COVID 19 made a serious impact on many aspects of everyday life. The world saw a paradigm shift in the education system favouring online learning during the constrains of pandemic. Methodology: To assess the attitude of the students towards online learning in subject of ENT, we conducted an observational study among 170 third year MBBS undergraduate students of our institute attending online classes through the student portal of our university website. Results: Our survey revealed students favoured online learning to sustain their academic interest and development during this pandemic. Yet, they perceived many challenges during online learning like lack of face-to-face interactions, lack of socialization, distraction by social media, technology related issues etc. Students also opted for a combined approach of learning in the post pandemic period. Conclusion: This article reflects the challenges faced during online learning and added the innovative methods that can be included to overcome the obstacles of online learning. During this period of COVID, one must embrace the alternative to classroom learning to keep up with one's academic development and can consider an integrated approach of learning after the pandemic.

18.
J Biosci ; 472022.
Artículo en Inglés | MEDLINE | ID: mdl-36222137

RESUMEN

Leucine-rich repeats (LRRs) - the protein-protein and protein-ligand interaction motif of proteins participating in a plethora of functions in plants, vertebrates, invertebrates, and prokaryotes - are a fascinating piece of conserved yet versatile structural motif. In toll-like receptors (TLRs), this domain forms the extracellular part that is preceded by an intracellular toll/interleukin-1 receptor (TIR) domain. The extracellular part is crucial for recognizing a structurally diverse set of viral, bacterial, fungal, and parasite-derived components, while the TIR domain is recruited for activation of downstream signaling following recognition. The distinct ability of the paralogs TLR1 and TLR6 to dimerize with TLR2 and recognize different ligands intrigued and motivated us to exchange the dimerizing and ligand-binding residues between TLR1/6 and note the effect on dimer formation and ligand binding. The appreciable sequence modification brought about no significant alteration in the native scaffold of the motif, as revealed from the comparison of simulations with wild-type dimers. Moreover, docking of the exchanged ligands to the variant proteins supported favorable binding. Thus, the structural stability and the functional plasticity offered by the motif might be the reason for its extensive use across cellular functions and life forms, a feature crucial for coevolution and the knowledge essential for therapeutics.


Asunto(s)
Receptor Toll-Like 1 , Receptor Toll-Like 6 , Animales , Leucina/genética , Ligandos , Receptores de Interleucina-1 , Receptor Toll-Like 1/metabolismo , Receptor Toll-Like 2/química , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 6/metabolismo , Receptores Toll-Like/química , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
19.
Indian J Otolaryngol Head Neck Surg ; 74(Suppl 3): 5003-5007, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36742784

RESUMEN

Obstructive sleep apnea (OSA) has been linked to and is associated with increased cardiovascular and cerebrovascular morbidity. Ongoing inflammatory responses play an important role in this association. Systemic inflammation is important in pathophysiology of OSA and its comorbidity. In this study, we aimed to evaluate the role of neutrophil-to-lymphocyte ratio (NLR) in OSA patients and comparing with other well-known inflammatory marker, C-reactive protein (CRP) along with thyroid-stimulating hormone(TSH) and body-mass index(BMI). We conducted a retrospective analysis of 162 patients with OSA and divided them into 2 categories based on apnea-hypopnea index (AHI) (< 30 and > = 30), and recorded their leukocyte profiles, sex, age and body mass index. 80 matched healthy controls were taken. Patients were excluded if they had underlying cancer, chronic inflammatory disease, any systemic infection, uncontrolled hypertension and diabetes mellitus, a known acute coronary syndrome, valvular heart disease, renal or hepatic dysfunction. We found that N/L Ratio in severe OSA patients was significantly higher compared with mild and moderate OSA patients and healthy controls (p < 0.001). CRP levels were not different in all OSA stages (p = 0.595). We noted a significant difference in mean BMI of the two groups. In the wake of increase in prevalence of OSA in a developing country like India coupled with inadequate proportion of sleep labs, NLR is an inexpensive, easy to obtain, widely available marker of inflammation that might in combination with other markers assist in identifying patients with severe OSA.

20.
Protein Sci ; 31(10): e4430, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36173179

RESUMEN

Chlorophyll degradation plays a myriad of physiological roles in photosynthetic organisms, including acclimation to light environment and nutrient remobilization during senescence. Mg extraction from chlorophyll a is the first and committed step of the chlorophyll degradation pathway. This reaction is catalyzed by the Mg-dechelatase enzyme encoded by Stay-Green (SGR). The reaction mechanism of SGR protein remains elusive since metal ion extraction from organic molecules is not a common enzymatic reaction. Additionally, experimentally derived structural information about SGR or its homologs has not yet been reported. In this study, the crystal structure of the SGR homolog from Anaerolineae bacterium was determined using the molecular replacement method at 1.85 Å resolution. Our previous study showed that three residues-H32, D34, and D62 are essential for the catalytic activity of the enzyme. Biochemical analysis involving mutants of D34 residue further strengthened its importance in the functioning of the dechelatase. Docking simulation also revealed the interaction between the D34 side chain and central Mg ion of chlorophyll a. Structural analysis showed the arrangement of D34/H32/D62 in the form of a catalytic triad that is generally found in hydrolases. The probable reaction mechanism suggests that deprotonated D34 side chain coordinates and destabilizes Mg, resulting in Mg extraction. Besides, H32 possibly acts as a general base catalyst and D62 facilitates H32 to be a better proton acceptor. Taken together, the reaction mechanism of SGR partially mirrors the one observed in hydrolases.


Asunto(s)
Chloroflexi , Chloroflexi/metabolismo , Clorofila , Clorofila A , Enzimas , Hidrolasas , Protones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA