Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Anal Biochem ; 666: 115047, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36682579

RESUMO

Due to the emergence of multidrug resistant pathogens, it is imperative to identify new targets for antibiotic drug discovery. The S-adenosylhomocysteine (SAH) nucleosidase enzyme is a promising target for antimicrobial drug development due to its critical functions in multiple bacterial processes including recycling of toxic byproducts of S-adenosylmethionine (SAM)-mediated reactions and producing the precursor of the universal quorum sensing signal, autoinducer-2 (AI-2). Riboswitches are structured RNA elements typically used by bacteria to precisely monitor and respond to changes in essential bacterial processes, including metabolism. Natural riboswitches fused to a reporter gene can be exploited to detect changes in metabolism or in physiological signaling. We performed a high-throughput screen (HTS) using an SAH-riboswitch controlled ß-galactosidase reporter gene in Escherichia coli to discover small molecules that inhibit SAH recycling. We demonstrate that the assay strategy using SAH riboswitches to detect the effects of SAH nucleosidase inhibitors can quickly identify compounds that penetrate the barriers of Gram-negative bacterial cells and perturb pathways involving SAH.


Assuntos
Riboswitch , S-Adenosilmetionina/metabolismo , RNA/genética , Bactérias/genética , N-Glicosil Hidrolases/genética , N-Glicosil Hidrolases/metabolismo
2.
ACS Med Chem Lett ; 11(10): 1843-1847, 2020 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-33062162

RESUMO

Human Macrophage Migration Inhibitory Factor (MIF) is a trimeric cytokine implicated in a number of inflammatory and autoimmune diseases and cancer. We previously reported that the dye p425 (Chicago Sky Blue), which bound MIF at the interface of two MIF trimers covering the tautomerase and allosteric pockets, revealed a unique strategy to block MIF's pro-inflammatory activities. Structural liabilities, including the large size, precluded p425 as a medicinal chemistry lead for drug development. We report here a rational design strategy linking only the fragment of p425 that binds over the tautomerase pocket to the core of ibudilast, a known MIF allosteric site-specific inhibitor. The chimeric compound, termed L2-4048, was shown by X-ray crystallography to bind at the allosteric and tautomerase sites as anticipated. L2-4048 retained target binding and blocked MIF's tautomerase CD74 receptor binding, and pro-inflammatory activities. Our studies lay the foundation for the design and synthesis of smaller and more drug-like compounds that retain the MIF inhibitory properties of this chimera.

3.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 7): 1922-33, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25004969

RESUMO

Clostridium difficile, a Gram-positive, spore-forming anaerobic bacterium, is the leading cause of infectious diarrhea among hospitalized patients. C. difficile is frequently associated with antibiotic treatment, and causes diseases ranging from antibiotic-associated diarrhea to life-threatening pseudomembranous colitis. The severity of C. difficile infections is exacerbated by the emergence of hypervirulent and multidrug-resistant strains, which are difficult to treat and are often associated with increased mortality rates. Alanine racemase (Alr) is a pyridoxal-5'-phosphate (PLP)-dependent enzyme that catalyzes the reversible racemization of L- and D-alanine. Since D-alanine is an essential component of the bacterial cell-wall peptidoglycan, and there are no known Alr homologs in humans, this enzyme is being tested as an antibiotic target. Cycloserine is an antibiotic that inhibits Alr. In this study, the catalytic properties and crystal structures of recombinant Alr from the virulent and multidrug-resistant C. difficile strain 630 are presented. Three crystal structures of C. difficile Alr (CdAlr), corresponding to the complex with PLP, the complex with cycloserine and a K271T mutant form of the enzyme with bound PLP, are presented. The structures are prototypical Alr homodimers with two active sites in which the cofactor PLP and cycloserine are localized. Kinetic analyses reveal that the K271T mutant CdAlr has the highest catalytic constants reported to date for any Alr. Additional studies are needed to identify the basis for the high catalytic activity. The structural and activity data presented are first steps towards using CdAlr for the development of structure-based therapeutics for C. difficile infections.


Assuntos
Alanina Racemase/química , Clostridioides difficile/enzimologia , Farmacorresistência Bacteriana Múltipla , Sequência de Aminoácidos , Cromatografia em Gel , Clostridioides difficile/efeitos dos fármacos , Cristalografia por Raios X , Dimerização , Dados de Sequência Molecular , Conformação Proteica , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos
4.
Biochem Pharmacol ; 86(2): 222-30, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23680030

RESUMO

The genus Mycobacterium includes non-pathogenic species such as M. smegmatis, and pathogenic species such as M. tuberculosis, the causative agent of tuberculosis (TB). Treatment of TB requires a lengthy regimen of several antibiotics, whose effectiveness has been compromised by the emergence of resistant strains. New antibiotics that can shorten the treatment course and those that have not been compromised by bacterial resistance are needed. In this study, we report that thiadiazolidinones, a relatively little-studied heterocyclic class, inhibit the activity of mycobacterial alanine racemase, an essential enzyme that converts l-alanine to d-alanine for peptidoglycan synthesis. Twelve members of the thiadiazolidinone family were evaluated for inhibition of M. tuberculosis and M. smegmatis alanine racemase activity and bacterial growth. Thiadiazolidinones inhibited M. tuberculosis and M. smegmatis alanine racemases to different extents with 50% inhibitory concentrations (IC50) ranging from <0.03 to 28µM and 23 to >150µM, respectively. The compounds also inhibited the growth of these bacteria, including multidrug resistant strains of M. tuberculosis. The minimal inhibitory concentrations (MIC) for drug-susceptible M. tuberculosis and M. smegmatis ranged from 6.25µg/ml to 100µg/ml, and from 1.56 to 6.25µg/ml for drug-resistant M. tuberculosis. The in vitro activities of thiadiazolidinones suggest that this family of compounds might represent starting points for medicinal chemistry efforts aimed at developing novel antimycobacterial agents.


Assuntos
Alanina Racemase/antagonistas & inibidores , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Tiadiazóis/farmacologia , Alanina Racemase/química , Alanina Racemase/metabolismo , Sequência de Aminoácidos , Catálise , Dados de Sequência Molecular , Mycobacterium smegmatis/enzimologia , Mycobacterium tuberculosis/enzimologia , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
5.
J Biol Chem ; 287(36): 30653-63, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22782901

RESUMO

Macrophage migration inhibitory factor (MIF) is a catalytic cytokine and an upstream mediator of the inflammatory pathway. MIF has broad regulatory properties, dysregulation of which has been implicated in the pathology of multiple immunological diseases. Inhibition of MIF activity with small molecules has proven beneficial in a number of disease models. Known small molecule MIF inhibitors typically bind in the tautomerase site of the MIF trimer, often covalently modifying the catalytic proline. Allosteric MIF inhibitors, particularly those that associate with the protein by noncovalent interactions, could reveal novel ways to block MIF activity for therapeutic benefit and serve as chemical probes to elucidate the structural basis for the diverse regulatory properties of MIF. In this study, we report the identification and functional characterization of a novel allosteric MIF inhibitor. Identified from a high throughput screening effort, this sulfonated azo compound termed p425 strongly inhibited the ability of MIF to tautomerize 4-hydroxyphenyl pyruvate. Furthermore, p425 blocked the interaction of MIF with its receptor, CD74, and interfered with the pro-inflammatory activities of the cytokine. Structural studies revealed a unique mode of binding for p425, with a single molecule of the inhibitor occupying the interface of two MIF trimers. The inhibitor binds MIF mainly on the protein surface through hydrophobic interactions that are stabilized by hydrogen bonding with four highly specific residues from three different monomers. The mode of p425 binding reveals a unique way to block the activity of the cytokine for potential therapeutic benefit in MIF-associated diseases.


Assuntos
Antígenos de Diferenciação de Linfócitos B/metabolismo , Compostos Azo , Fibroblastos/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Oxirredutases Intramoleculares , Fatores Inibidores da Migração de Macrófagos , Azul Tripano/química , Azul Tripano/farmacologia , Regulação Alostérica/efeitos dos fármacos , Antígenos de Diferenciação de Linfócitos B/química , Compostos Azo/química , Compostos Azo/farmacologia , Células Cultivadas , Fibroblastos/citologia , Antígenos de Histocompatibilidade Classe II/química , Humanos , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/química , Fatores Inibidores da Migração de Macrófagos/metabolismo , Ligação Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína
6.
Biochem Pharmacol ; 83(3): 368-77, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22146584

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) is a human pathogen and a major cause of hospital-acquired infections. New antibacterial agents that have not been compromised by bacterial resistance are needed to treat MRSA-related infections. We chose the S. aureus cell wall synthesis enzyme, alanine racemase (Alr) as the target for a high-throughput screening effort to obtain novel enzyme inhibitors, which inhibit bacterial growth. Among the 'hits' identified was a thiadiazolidinone with chemical properties attractive for lead development. This study evaluated the mode of action, antimicrobial activities, and mammalian cell cytotoxicity of the thiadiazolidinone family in order to assess its potential for development as a therapeutic agent against MRSA. The thiadiazolidones inhibited Alr activity with 50% inhibitory concentrations (IC50) ranging from 0.36 to 6.4 µM, and they appear to inhibit the enzyme irreversibly. The series inhibited the growth of S. aureus, including MRSA strains, with minimal inhibitory concentrations (MICs) ranging from 6.25 to 100 µg/ml. The antimicrobial activity showed selectivity against Gram-positive bacteria and fungi, but not Gram-negative bacteria. The series inhibited human HeLa cell proliferation. Lead development centering on the thiadiazolidinone series would require additional medicinal chemistry efforts to enhance the antibacterial activity and minimize mammalian cell toxicity.


Assuntos
Alanina Racemase/antagonistas & inibidores , Antibacterianos/síntese química , Antibacterianos/farmacologia , Staphylococcus aureus Resistente à Meticilina/enzimologia , Tiadiazóis/química , Tiadiazóis/farmacologia , Alanina Racemase/metabolismo , Antibacterianos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Células HeLa , Humanos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Tiadiazóis/classificação
7.
PLoS One ; 6(5): e20374, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21637807

RESUMO

BACKGROUND: In an effort to discover new drugs to treat tuberculosis (TB) we chose alanine racemase as the target of our drug discovery efforts. In Mycobacterium tuberculosis, the causative agent of TB, alanine racemase plays an essential role in cell wall synthesis as it racemizes L-alanine into D-alanine, a key building block in the biosynthesis of peptidoglycan. Good antimicrobial effects have been achieved by inhibition of this enzyme with suicide substrates, but the clinical utility of this class of inhibitors is limited due to their lack of target specificity and toxicity. Therefore, inhibitors that are not substrate analogs and that act through different mechanisms of enzyme inhibition are necessary for therapeutic development for this drug target. METHODOLOGY/PRINCIPAL FINDINGS: To obtain non-substrate alanine racemase inhibitors, we developed a high-throughput screening platform and screened 53,000 small molecule compounds for enzyme-specific inhibitors. We examined the 'hits' for structural novelty, antimicrobial activity against M. tuberculosis, general cellular cytotoxicity, and mechanism of enzyme inhibition. We identified seventeen novel non-substrate alanine racemase inhibitors that are structurally different than any currently known enzyme inhibitors. Seven of these are active against M. tuberculosis and minimally cytotoxic against mammalian cells. CONCLUSIONS/SIGNIFICANCE: This study highlights the feasibility of obtaining novel alanine racemase inhibitor lead compounds by high-throughput screening for development of new anti-TB agents.


Assuntos
Alanina Racemase/antagonistas & inibidores , Anti-Infecciosos/farmacologia , Inibidores Enzimáticos/classificação , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , Alanina Desidrogenase/metabolismo , Alanina Racemase/química , Alanina Racemase/metabolismo , Alanina Racemase/farmacologia , Anti-Infecciosos/análise , Anti-Infecciosos/química , Anti-Infecciosos/classificação , Morte Celular/efeitos dos fármacos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/química , Células HeLa , Humanos , Concentração Inibidora 50 , Cinética , Espectrometria de Massas , Testes de Sensibilidade Microbiana , Especificidade por Substrato/efeitos dos fármacos
8.
J Immunol ; 183(1): 650-60, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19535627

RESUMO

West Nile virus is an emerging pathogen that can cause fatal neurological disease. A recombinant human mAb, mAb11, has been described as a candidate for the prevention and treatment of West Nile disease. Using a yeast surface display epitope mapping assay and neutralization escape mutant, we show that mAb11 recognizes the fusion loop, at the distal end of domain II of the West Nile virus envelope protein. Ab mAb11 cross-reacts with all four dengue viruses and provides protection against dengue (serotypes 2 and 4) viruses. In contrast to the parental West Nile virus, a neutralization escape variant failed to cause lethal encephalitis (at higher infectious doses) or induce the inflammatory responses associated with blood-brain barrier permeability in mice, suggesting an important role for the fusion loop in viral pathogenesis. Our data demonstrate that an intact West Nile virus fusion loop is critical for virulence, and that human mAb11 targeting this region is efficacious against West Nile virus infection. These experiments define the molecular determinant on the envelope protein recognized by mAb11 and demonstrate the importance of this region in causing West Nile encephalitis.


Assuntos
Anticorpos Monoclonais/metabolismo , Sítios de Ligação de Anticorpos , Peptídeos/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/patogenicidade , Animais , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular , Reações Cruzadas , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptídeos/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Febre do Nilo Ocidental/terapia , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/imunologia
9.
Antiviral Res ; 82(3): 166-8, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19135091

RESUMO

West Nile virus (WNV) is an emerging human pathogen for which specific antiviral therapy has not been developed. The therapeutic potential of RNA interference (RNAi) as a sequence-specific inhibitor of WNV has been well demonstrated. Although multiple siRNA targets have been identified within the genomic coding region, targets within the untranslated regions (UTR), which encode cis-acting regulatory elements, remain relatively unknown. In WNV and other flaviviruses, UTRs are located at the genomic termini. These regions form complex secondary structures, which pose difficulty when designing effective siRNA targets. In this study, we report the identification of siRNA targets in the WNV 3' UTR. These targets were selected by siRNA predictor algorithms, and synthesized as short hairpin RNA sequences from a plasmid-based expression system. Vero cells stably expressing these sequences had greatly diminished ability to support WNV replication but not the related dengue virus, demonstrating that the siRNAs were effective and suppressed WNV viral replication in a sequence-specific manner. The siRNAs developed in this study could function as potential antiviral therapeutics and as genetic tools to investigate the role of 3' UTR in WNV pathogenesis.


Assuntos
Regiões 3' não Traduzidas , Antivirais/farmacologia , RNA Interferente Pequeno/genética , RNA Viral/genética , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/genética , Animais , Sequência de Bases , Chlorocebus aethiops , Vírus da Dengue/genética , Vírus da Dengue/fisiologia , Humanos , Dados de Sequência Molecular , RNA Interferente Pequeno/farmacologia , Células Vero , Ensaio de Placa Viral , Vírus do Nilo Ocidental/fisiologia
10.
J Infect Dis ; 196(12): 1741-8, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18190253

RESUMO

The flavivirus envelope (E) protein mediates cellular attachment and fusion with host cell membranes and is recognized by virus-neutralizing antibodies. We raised antibodies against a broad range of epitopes by immunizing a horse with recombinant West Nile virus (WNV) E protein. To define epitopes recognized by protective antibodies, we selected, by affinity chromatography, immunoglobulins against immobilized linear peptides derived from parts of the E protein. Immunoglobulins binding 9 different peptides from domains I, II, and III of the E protein neutralized WNV in vitro. This indicates that multiple protective epitopes can be found in the E protein. Immunoglobulins recognizing 3 peptides derived from domains I and II of E protein protected mice against a lethal challenge with WNV. These immunoglobulins recognized the E proteins of related flaviviruses, demonstrating that antibodies targeting specific E protein epitopes could be developed for prevention and treatment of multiple flavivirus infections.


Assuntos
Anticorpos Antivirais/imunologia , Imunização Passiva/métodos , Imunoglobulinas/imunologia , Proteínas do Envelope Viral/imunologia , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/farmacologia , Afinidade de Anticorpos/imunologia , Chlorocebus aethiops , Drosophila , Epitopos/imunologia , Feminino , Imunofluorescência/métodos , Cavalos , Camundongos , Camundongos Endogâmicos C3H , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Células Vero , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA