Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(50): E4904-12, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24191014

RESUMEN

Pathogenic microorganisms and toxins have evolved a variety of mechanisms to gain access to the host-cell cytosol and thereby exert virulent effects upon the host. One common mechanism of cellular entry requires trafficking to an acidified endosome, which promotes translocation across the host membrane. To identify small-molecule inhibitors that block this process, a library of 30,000 small molecules was screened for inhibitors of anthrax lethal toxin. Here we report that 4-bromobenzaldehyde N-(2,6-dimethylphenyl)semicarbazone, the most active compound identified in the screen, inhibits intoxication by lethal toxin and blocks the entry of multiple other acid-dependent bacterial toxins and viruses into mammalian cells. This compound, which we named EGA, also delays lysosomal targeting and degradation of the EGF receptor, indicating that it targets host-membrane trafficking. In contrast, EGA does not block endosomal recycling of transferrin, retrograde trafficking of ricin, phagolysosomal trafficking, or phagosome permeabilization by Franciscella tularensis. Furthermore, EGA does not neutralize acidic organelles, demonstrating that its mechanism of action is distinct from pH-raising agents such as ammonium chloride and bafilomycin A1. EGA is a powerful tool for the study of membrane trafficking and represents a class of host-targeted compounds for therapeutic development to treat infectious disease.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Endosomas/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Semicarbazonas/farmacología , Internalización del Virus/efectos de los fármacos , Aminas , Animales , Transporte Biológico/fisiología , Caspasa 1/metabolismo , Cromatografía Liquida , Endosomas/fisiología , Citometría de Flujo , Células HeLa , Humanos , Macrófagos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Estructura Molecular , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Semicarbazonas/química , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
2.
Artículo en Inglés | MEDLINE | ID: mdl-38669694

RESUMEN

There is a high unmet need for early detection approaches for diffuse gastric cancer (DGC). We examined whether the stool proteome of mouse models of GC or individuals with hereditary diffuse GC (HDGC) have utility as biomarkers for early detection. Proteomic mass spectrometry of stool from a genetically engineered mouse model driven by oncogenic KrasG12D and loss of p53 and Cdh1 in gastric parietal cells (known as TCON mice) identified differentially abundant proteins compared to littermate controls. Immunoblot assays validated a panel of proteins including actinin alpha 4 (ACTN4), N-acylsphingosine amidohydrolase 2 (ASAH2), dipeptidyl peptidase 4 (DPP4), and valosin-containing protein (VCP) as enriched in TCON stool compared to littermate control stool. Immunofluorescence analysis of these proteins in TCON stomach sections revealed increased protein expression as compared to littermate controls. Proteomic mass spectrometry of stool obtained from HDGC patients with CDH1 mutations identified increased expression of ASAH2, DPP4, VCP, lactotransferrin (LTF), and tropomyosin-2 (TPM2) relative to stool from healthy sex and age-matched donors. Chemical inhibition of ASAH2 using C6-urea ceramide was toxic to GC cell lines and patient derived-GC organoids. This toxicity was reversed by adding downstream products of the S1P synthesis pathway, suggesting a dependency on ASAH2 activity in GC. An exploratory analysis of the HDGC stool microbiome identified features which correlated with patient tumors. Here we provide evidence supporting the potential of analyzing stool biomarkers for the early detection of DGC.

3.
ACS Infect Dis ; 7(8): 2176-2191, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34218660

RESUMEN

Anthrax is caused by Bacillus anthracis and can result in nearly 100% mortality due in part to anthrax toxin. Antimalarial amodiaquine (AQ) acts as a host-oriented inhibitor of anthrax toxin endocytosis. Here, we determined the pharmacokinetics and safety of AQ in mice, rabbits, and humans as well as the efficacy in the fly, mouse, and rabbit models of anthrax infection. In the therapeutic-intervention studies, AQ nearly doubled the survival of mice infected subcutaneously with a B. anthracis dose lethal to 60% of the animals (LD60). In rabbits challenged with 200 LD50 of aerosolized B. anthracis, AQ as a monotherapy delayed death, doubled the survival rate of infected animals that received a suboptimal amount of antibacterial levofloxacin, and reduced bacteremia and toxemia in tissues. Surprisingly, the anthrax efficacy of AQ relies on an additional host macrophage-directed antibacterial mechanism, which was validated in the toxin-independent Drosophila model of Bacillus infection. Lastly, a systematic literature review of the safety and pharmacokinetics of AQ in humans from over 2 000 published articles revealed that AQ is likely safe when taken as prescribed, and its pharmacokinetics predicts anthrax efficacy in humans. Our results support the future examination of AQ as adjunctive therapy for the prophylactic anthrax treatment.


Asunto(s)
Carbunco , Bacillus anthracis , Amodiaquina , Animales , Carbunco/tratamiento farmacológico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Levofloxacino , Ratones , Conejos , Revisiones Sistemáticas como Asunto
4.
ACS Infect Dis ; 4(12): 1746-1754, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30354048

RESUMEN

Inflammasomes activate caspase-1 in response to molecular signals from pathogens and other dangerous stimuli as a part of the innate immune response. A previous study discovered a small-molecule, 4-fluoro- N'-[1-(2-pyridinyl)ethylidene]benzohydrazide, which we named DN1, that reduces the cytotoxicity of anthrax lethal toxin (LT). We determined that DN1 protected cells irrespectively of LT concentration and reduced the pathogenicity of an additional bacterial exotoxin and several viruses. Using the LT cytotoxicity pathway, we show that DN1 does not prevent LT internalization and catalytic activity or caspase-1 activation. Moreover, DN1 does not affect the proteolytic activity of host cathepsin B, which facilitates the cytoplasmic entry of toxins. PubChem Bioactivities lists two G protein-coupled receptors (GPCR), type-1 angiotensin II receptor and apelin receptor, as targets of DN1. The inhibition of phosphatidylinositol 3-kinase, phospholipase C, and protein kinase B, which are downstream of GPCR signaling, synergized with DN1 in protecting cells from LT. We hypothesize that DN1-mediated antagonism of GPCRs modulates signal transduction pathways to induce a cellular state that reduces LT-induced pyroptosis downstream of caspase-1 activation. DN1 also reduced the susceptibility of Drosophila melanogaster to toxin-associated bacterial infections. Future experiments will aim to further characterize how DN1 modulates signal transduction pathways to inhibit pyroptotic cell death in LT-sensitive macrophages. DN1 represents a novel chemical probe to investigate host cellular mechanisms that mediate cell death in response to pathogenic agents.


Asunto(s)
Carbunco/fisiopatología , Antibacterianos/farmacología , Antígenos Bacterianos/toxicidad , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/toxicidad , Muerte Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Carbunco/tratamiento farmacológico , Carbunco/metabolismo , Carbunco/microbiología , Antibacterianos/química , Bacillus anthracis/genética , Bacillus anthracis/crecimiento & desarrollo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/antagonistas & inhibidores , Caspasa 1/genética , Caspasa 1/metabolismo , Catepsina B/genética , Catepsina B/metabolismo , Drosophila melanogaster , Femenino , Interacciones Huésped-Patógeno , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Bibliotecas de Moléculas Pequeñas/química
5.
ACS Infect Dis ; 4(8): 1235-1245, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-29749721

RESUMEN

Exploiting the host endocytic trafficking pathway is a common mechanism by which bacterial exotoxins gain entry to exert virulent effects upon the host cells. A previous study identified a small-molecule, 1-(2,6-dimethyl-1-piperidinyl)-3-[(2-isopropyl-5-methylcyclohexyl)oxy]-2-propanol, that blocks the process of anthrax lethal toxin (LT) cytotoxicity. Here, we report the characterization of the bioactivity of this compound, which we named RC1. We found that RC1 protected host cells independently of LT concentration and also blocked intoxication by other bacterial exotoxins, suggesting that the target of the compound is a host factor. Using the anthrax LT intoxication pathway as a reference, we show that while anthrax toxin is able to bind to cells and establish an endosomal pore in the presence of the drug, the toxin is unable to translocate into the cytosol. We demonstrate that RC1 does not inhibit the toxin directly but rather reduces the enzymatic activity of host cathepsin B that mediates the escape of toxins into the cytoplasm from late endosomes. We demonstrate that the pathogenicity of Human cytomegalovirus and Herpes simplex virus 1, which relies on cathepsin B protease activity, is reduced by RC1. This study reveals the potential of RC1 as a broad-spectrum host-oriented therapy against several aggressive and deadly pathogens.


Asunto(s)
Antídotos/farmacología , Antivirales/farmacología , Catepsina B/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Piperidinas/farmacología , Animales , Toxinas Bacterianas/antagonistas & inhibidores , Línea Celular , Citomegalovirus/efectos de los fármacos , Citomegalovirus/crecimiento & desarrollo , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/crecimiento & desarrollo , Humanos , Ratones
6.
Sci Rep ; 5: 13476, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26310922

RESUMEN

A longstanding and still-increasing threat to the effective treatment of infectious diseases is resistance to antimicrobial countermeasures. Potentially, the targeting of host proteins and pathways essential for the detrimental effects of pathogens offers an approach that may discover broad-spectrum anti-pathogen countermeasures and circumvent the effects of pathogen mutations leading to resistance. Here we report implementation of a strategy for discovering broad-spectrum host-oriented therapies against multiple pathogenic agents by multiplex screening of drugs for protection against the detrimental effects of multiple pathogens, identification of host cell pathways inhibited by the drug, and screening for effects of the agent on other pathogens exploiting the same pathway. We show that a clinically used antimalarial drug, Amodiaquine, discovered by this strategy, protects host cells against infection by multiple toxins and viruses by inhibiting host cathepsin B. Our results reveal the practicality of discovering broadly acting anti-pathogen countermeasures that target host proteins exploited by pathogens.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Virus/efectos de los fármacos , Amodiaquina/química , Amodiaquina/farmacología , Animales , Catepsina B/metabolismo , Muerte Celular/efectos de los fármacos , Citosol/efectos de los fármacos , Citosol/metabolismo , Aprobación de Drogas , Ebolavirus/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Células HeLa , Humanos , Metaboloma/efectos de los fármacos , Ratones , Modelos Biológicos , Células RAW 264.7 , Estados Unidos , United States Food and Drug Administration
7.
ACS Med Chem Lett ; 5(4): 363-7, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24900841

RESUMEN

EGA, 1, prevents the entry of multiple viruses and bacterial toxins into mammalian cells by inhibiting vesicular trafficking. The cellular target of 1 is unknown, and a structure-activity relationship study was conducted in order to develop a strategy for target identification. A compound with midnanomolar potency was identified (2), and three photoaffinity labels were synthesized (3-5). For this series, the expected photochemistry of the phenyl azide moiety is a more important factor than the IC50 of the photoprobe in obtaining a successful photolabeling event. While 3 was the most effective reversible inhibitor of the series, it provided no protection to cells against anthrax lethal toxin (LT) following UV irradiation. Conversely, 5, which possessed weak bioactivity in the standard assay, conferred robust irreversible protection vs LT to cells upon UV photolysis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA