Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Antimicrob Agents Chemother ; 67(7): e0009023, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37278639

RESUMEN

Mycobacterium abscessus infections are difficult to treat and are often considered untreatable without tissue resection. Due to the intrinsic drug-resistant nature of the bacteria, combination therapy of three or more antibiotics is recommended. A major challenge in treating M. abscessus infections is the absence of a universal combination therapy with satisfying clinical success rates, leaving clinicians to treat infections using antibiotics lacking efficacy data. We systematically measured drug combinations in M. abscessus to establish a resource of drug interaction data and identify patterns of synergy to help design optimized combination therapies. We measured 191 pairwise drug combination effects among 22 antibacterials and identified 71 synergistic pairs, 54 antagonistic pairs, and 66 potentiator-antibiotic pairs. We found that commonly used drug combinations in the clinic, such as azithromycin and amikacin, are antagonistic in the lab reference strain ATCC 19977, whereas novel combinations, such as azithromycin and rifampicin, are synergistic. Another challenge in developing universally effective multidrug therapies for M. abscessus is the significant variation in drug response between isolates. We measured drug interactions in a focused set of 36 drug pairs across a small panel of clinical isolates with rough and smooth morphotypes. We observed strain-dependent drug interactions that cannot be predicted from single-drug susceptibility profiles or known drug mechanisms of action. Our study demonstrates the immense potential to identify synergistic drug combinations in the vast drug combination space and emphasizes the importance of strain-specific combination measurements for designing improved therapeutic interventions.


Asunto(s)
Infecciones por Mycobacterium no Tuberculosas , Mycobacterium abscessus , Humanos , Azitromicina/farmacología , Azitromicina/uso terapéutico , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Amicacina/farmacología , Amicacina/uso terapéutico , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Infecciones por Mycobacterium no Tuberculosas/microbiología , Interacciones Farmacológicas , Pruebas de Sensibilidad Microbiana
2.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G227-G237, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32597706

RESUMEN

Antibiotic treatment is a standard therapy for Clostridioides difficile infection, but dysbiosis of the gut microbiota due to antibiotic exposure is also a major risk factor for the disease. Following an initial episode of C. difficile infection, a relentless cycle of recurrence can occur, where persistent treatment-related dysbiosis predisposes the patient to subsequent relapse. This study uses a longitudinal study design to compare the effects of a narrow-spectrum (ridinilazole) or broad-spectrum antibiotic (vancomycin) on intestinal bile acid profiles and their associations with gut bacteria over the course of C. difficile infection treatment. At the end of treatment (day 10), subjects receiving vancomycin showed a nearly 100-fold increase in the ratio of conjugated to secondary bile acids in their stool compared with baseline, whereas subjects receiving ridinilazole maintained this ratio near baseline levels. Correlation analysis detected significant positive associations between secondary bile acids and several Bacteroidales and Clostridiales families. These families were depleted in the vancomycin group but preserved at near-baseline abundance in the ridinilazole group. Enterobacteriaceae, which expanded to a greater extent in the vancomycin group, correlated negatively and positively with secondary and conjugated primary bile acids, respectively. Bile acid ratios at the end of treatment were significantly different between those who recurred and those who did not. These results indicate that a narrow-spectrum antibiotic maintains an intestinal bile acid profile associated with a lowered risk of recurrence.NEW & NOTEWORTHY This is the first study to demonstrate in humans the relationships between Clostridioides difficile antibiotic treatment choice and bile acid metabolism both during therapy and after treatment cessation. The results show a microbiota- and metabolome-preserving property of a novel narrow-spectrum agent that correlates with the agent's favorable sustained clinical response rates compared with broad-spectrum antibiotic treatment.


Asunto(s)
Antibacterianos/farmacología , Bencimidazoles/farmacología , Ácidos y Sales Biliares/química , Clostridiales/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Piridinas/farmacología , Ácidos y Sales Biliares/metabolismo , Heces/química , Microbioma Gastrointestinal/fisiología , Humanos
3.
J Antimicrob Chemother ; 73(8): 2078-2084, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29718329

RESUMEN

Objectives: We evaluated the antimicrobial susceptibility and ribotypes of Clostridium difficile isolates from participants in a Phase 2 study of ridinilazole, a novel targeted-spectrum agent for treatment of C. difficile infection. Methods: Participants received ridinilazole (200 mg twice daily) or vancomycin (125 mg four times daily) for 10 days (ClinicalTrials.gov: NCT02092935). The MICs of ridinilazole and comparators for C. difficile isolates from stool samples were determined by agar dilution. Toxin gene profiling was performed by multiplex PCR and ribotype identification by capillary electrophoresis. Results: Eighty-nine isolates were recovered from 88/100 participants (one participant had two strains at baseline). The median colony count (cfu/g stool) was 1.9 × 104 (range: 2.5 × 102-7.0 × 106). Twelve participants (three received ridinilazole and nine received vancomycin) experienced recurrence, confirmed by immunoassays for free toxin in stool samples. The ribotype of eight out of nine isolates obtained at recurrence matched those of the initial isolates. All isolates, including those obtained at recurrence, were susceptible to ridinilazole within the expected range [median (range) MIC: 0.12 (0.06-0.5) mg/L]. The median (range) vancomycin MIC was 1 (0.5-4.0) mg/L. At baseline, 13.6% and 13.3% of samples in the ridinilazole and vancomycin groups were positive for VRE, increasing to 23.7% and 29.7% by day 40, respectively. Common ribotypes included 014-20 (14 isolates), 027 (13), 106 (7), 002 (7), 078-126 (4), 001 (4), 087 (3) and 198 (3). Toxin gene profiling of nearly all baseline isolates (98.9%) revealed a binary toxin gene (cdtA/cdtB) prevalence of 35%. Conclusions: Ridinilazole potently inhibited recovered C. difficile isolates. Recurrence was not associated with altered susceptibility.


Asunto(s)
Bencimidazoles/farmacología , Clostridioides difficile/clasificación , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/tratamiento farmacológico , Piridinas/farmacología , Vancomicina/farmacología , Antibacterianos/farmacología , Método Doble Ciego , Genes Bacterianos , Humanos , Pruebas de Sensibilidad Microbiana , Ribotipificación
4.
Anaerobe ; 51: 68-72, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29709869

RESUMEN

Extraintestinal infection with Clostridium difficile has been reported but remains uncommon. Treatment of this unusual complication is complex given the limitations of current therapeutic options. Here we report a novel case of recurrent extraintestinal C. difficile infection that occurred following fecal microbiota transplantation. Using whole genome sequencing, we confirmed recrudescence rather than reinfection was responsible. The patient ultimately responded to prolonged, targeted antimicrobial therapy informed by susceptibility testing.


Asunto(s)
Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/epidemiología , Trasplante de Microbiota Fecal/efectos adversos , Antibacterianos/uso terapéutico , Clostridioides difficile/clasificación , Clostridioides difficile/genética , Humanos , Masculino , Persona de Mediana Edad , Recurrencia , Resultado del Tratamiento , Secuenciación Completa del Genoma
5.
Infect Immun ; 84(1): 138-48, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26483408

RESUMEN

Infection with enterohemorrhagic Escherichia coli (EHEC) can result in severe disease, including hemorrhagic colitis and the hemolytic uremic syndrome. Shiga toxins (Stx) are the key EHEC virulence determinant contributing to severe disease. Despite inhibiting protein synthesis, Shiga toxins paradoxically induce the expression of proinflammatory cytokines from various cell types in vitro, including intestinal epithelial cells (IECs). This effect is mediated in large part by the ribotoxic stress response (RSR). The Shiga toxin-induced RSR is known to involve the activation of the stress-activated protein kinases (SAPKs) p38 and JNK. In some cell types, Stx also can induce the classical mitogen-activated protein kinases (MAPKs) or ERK1/2, but the mechanism(s) by which this activation occurs is unknown. In this study, we investigated the mechanism by which Stx activates ERK1/2s in IECs and the contribution of ERK1/2 activation to interleukin-8 (IL-8) expression. We demonstrate that Stx1 activates ERK1/2 in a biphasic manner: the first phase occurs in response to StxB1 subunit, while the second phase requires StxA1 subunit activity. We show that the A subunit-dependent ERK1/2 activation is mediated through ZAK-dependent signaling, and inhibition of ERK1/2 activation via the MEK1/2 inhibitors U0126 and PD98059 results in decreased Stx1-mediated IL-8 mRNA. Finally, we demonstrate that ERK1/2 are activated in vivo in the colon of Stx2-intoxicated infant rabbits, a model in which Stx2 induces a primarily neutrophilic inflammatory response. Together, our data support a role for ERK1/2 activation in the development of Stx-mediated intestinal inflammation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Butadienos/farmacología , Línea Celular , Escherichia coli Enterohemorrágica/patogenicidad , Activación Enzimática , Células Epiteliales/inmunología , Infecciones por Escherichia coli/microbiología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/genética , Células HEK293 , Síndrome Hemolítico-Urémico/microbiología , Humanos , Inflamación/inmunología , Interleucina-8/biosíntesis , Interleucina-8/genética , Mucosa Intestinal/inmunología , Quinasas Quinasa Quinasa PAM , Nitrilos/farmacología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Conejos
6.
Infect Immun ; 83(1): 28-38, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312954

RESUMEN

Shiga toxins (Stx) are a family of cytotoxic proteins that can cause hemolytic-uremic syndrome (HUS), a thrombotic microangiopathy, following infections by Shiga toxin-producing Escherichia coli (STEC). Renal failure is a key feature of HUS and a major cause of childhood renal failure worldwide. There are currently no specific therapies for STEC-associated HUS, and the mechanism of Stx-induced renal injury is not well understood primarily due to a lack of fully representative animal models and an inability to monitor disease progression on a molecular or cellular level in humans at early stages. Three-dimensional (3D) tissue models have been shown to be more in vivo-like in their phenotype and physiology than 2D cultures for numerous disease models, including cancer and polycystic kidney disease. It is unknown whether exposure of a 3D renal tissue model to Stx will yield a more in vivo-like response than 2D cell culture. In this study, we characterized Stx2-mediated cytotoxicity in a bioengineered 3D human renal tissue model previously shown to be a predictor of drug-induced nephrotoxicity and compared its response to Stx2 exposure in 2D cell culture. Our results demonstrate that although many mechanistic aspects of cytotoxicity were similar between 3D and 2D, treatment of the 3D tissues with Stx resulted in an elevated secretion of the kidney injury marker 1 (Kim-1) and the cytokine interleukin-8 compared to the 2D cell cultures. This study represents the first application of 3D tissues for the study of Stx-mediated kidney injury.


Asunto(s)
Riñón/efectos de los fármacos , Organoides/efectos de los fármacos , Toxina Shiga II/toxicidad , Receptor Celular 1 del Virus de la Hepatitis A , Humanos , Glicoproteínas de Membrana/análisis , Modelos Biológicos , Técnicas de Cultivo de Órganos , Receptores Virales/análisis
7.
Antimicrob Agents Chemother ; 59(8): 4593-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26014940

RESUMEN

Vancomycin-resistant enterococci (VRE) are endemic in health care settings. These organisms colonize the gastrointestinal tract and can lead to infection which is associated with increased mortality. There is no treatment for VRE colonization. We conducted a randomized, double-blind, placebo-controlled clinical trial to examine the safety and efficacy of administration of the probiotic Lactobacillus rhamnosus GG (LGG) for the reduction or elimination of intestinal colonization by VRE. Colonized adults were randomized to receive LGG or placebo for 14 days. Quantitative stool cultures for LGG and VRE were collected at baseline and days 7, 14, 21, 28, and 56. Day 14 stool samples from some subjects were analyzed by quantitative PCR (qPCR) for LGG. Patients were closely monitored for adverse events. Eleven subjects, of whom 5 received LGG and 6 received placebo, were analyzed. No differences in VRE colony counts were seen at any time points between groups. No decline in colony counts was seen over time in subjects who received LGG. LGG was detected by PCR in all samples tested from subjects who received LGG but was only isolated in culture from 2 of 5 subjects in the LGG group. No treatment-related adverse events were seen. We demonstrated that LGG could be administered safely to patients with comorbidities and is recoverable in some patients' stool cultures. Concomitant administration of antibiotics may have resulted in an inability to recover viable organisms from stool samples, but LGG DNA could still be detected by qPCR. LGG administration did not affect VRE colonization in this study. (This study was registered at Clinicaltrials.gov under registration no. NCT00756262.).


Asunto(s)
Tracto Gastrointestinal/microbiología , Lacticaseibacillus rhamnosus/crecimiento & desarrollo , Probióticos/administración & dosificación , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Vancomicina/farmacología , Anciano , Anciano de 80 o más Años , Recuento de Colonia Microbiana , Comorbilidad , Método Doble Ciego , Heces/microbiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
9.
Curr Top Microbiol Immunol ; 357: 41-65, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22057792

RESUMEN

Shiga toxins and ricin are potent inhibitors of protein synthesis. In addition to causing inhibition of protein synthesis, these toxins activate proinflammatory signaling cascades that may contribute to the severe diseases associated with toxin exposure. Treatment of cells with Shiga toxins and ricin have been shown to activate a number of signaling pathways including those associated with the ribotoxic stress response, Nuclear factor kappa B activation, inflammasome activation, the unfolded protein response, mTOR signaling, hemostasis, and retrograde trafficking. In this chapter, we review our current understanding of these signaling pathways as they pertain to intoxication by Shiga toxins and ricin.


Asunto(s)
Ricina/farmacología , Toxinas Shiga/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Proteínas Portadoras/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , FN-kappa B/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Transporte de Proteínas , Toxinas Shiga/metabolismo , Estrés Fisiológico
10.
Infect Immun ; 80(8): 2858-67, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22689816

RESUMEN

Shiga-toxigenic Escherichia coli (STEC) O113:H21 strains that lack the locus of enterocyte effacement (LEE) efficiently invade eukaryotic cells in vitro, unlike LEE-positive O157:H7 strains. We used a fliC deletion mutant of the O113:H21 STEC strain 98NK2 (98NK2ΔfliC) to show that invasion of colonic epithelial (HCT-8) cells is heavily dependent on production of flagellin, even though adherence to the cells was actually enhanced in the mutant. Flagellin binds and signals through Toll-like receptor 5 (TLR5), but there was no evidence that either TLR5, the adaptor protein myeloid differentiation primary response gene 88 (MyD88), or the serine kinase interleukin-1 receptor-associated kinase (IRAK) were required for invasion of HCT-8 cells by strain 98NK2, as judged by transfection, RNA knockdown, or inhibitor studies. However, pretreatment of cells with anti-asialo-GM1 significantly decreased 98NK2 invasion (by 40.8%), while neuraminidase treatment (which cleaves terminal sialic acid residues, thus converting GM1 into asialo-GM1) significantly increased invasion (by 70.7%). Pretreatment of HCT-8 cells with either the cholesterol-depleting agent methyl-ß-cyclodextrin (MßCD) or the tyrosine kinase inhibitor genistein significantly decreased invasion by 98NK2, indicating a potential role for lipid rafts in the invasion mechanism. Confocal microscopy also showed invading 98NK2 colocalized with lipid raft markers caveolin-1 and GM1. Interestingly, anti-asialo-GM1, neuraminidase, MßCD, and genistein have similar effects on the vestigial level of STEC invasion seen for STEC strain 98NK2ΔfliC, indicating that lipid rafts mediate a common step in flagellin-dependent and flagellin-independent cellular invasion.


Asunto(s)
Colon/citología , Células Epiteliales/microbiología , Flagelina/metabolismo , Microdominios de Membrana/fisiología , Escherichia coli Shiga-Toxigénica/metabolismo , Línea Celular Tumoral , Células Epiteliales/citología , Flagelina/genética , Gangliósido G(M1)/genética , Gangliósido G(M1)/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Escherichia coli Shiga-Toxigénica/clasificación , Escherichia coli Shiga-Toxigénica/genética , Receptor Toll-Like 5/metabolismo
11.
Nature ; 443(7111): 548-52, 2006 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-17024087

RESUMEN

AB5 toxins are produced by pathogenic bacteria and consist of enzymatic A subunits that corrupt essential eukaryotic cell functions, and pentameric B subunits that mediate uptake into the target cell. AB5 toxins include the Shiga, cholera and pertussis toxins and a recently discovered fourth family, subtilase cytotoxin, which is produced by certain Shiga toxigenic strains of Escherichia coli. Here we show that the extreme cytotoxicity of this toxin for eukaryotic cells is due to a specific single-site cleavage of the essential endoplasmic reticulum chaperone BiP/GRP78. The A subunit is a subtilase-like serine protease; structural studies revealed an unusually deep active-site cleft, which accounts for its exquisite substrate specificity. A single amino-acid substitution in the BiP target site prevented cleavage, and co-expression of this resistant protein protected transfected cells against the toxin. BiP is a master regulator of endoplasmic reticulum function, and its cleavage by subtilase cytotoxin represents a previously unknown trigger for cell death.


Asunto(s)
Toxinas Bacterianas/metabolismo , Citotoxinas/metabolismo , Retículo Endoplásmico/metabolismo , Escherichia coli/enzimología , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/antagonistas & inhibidores , Chaperonas Moleculares/metabolismo , Subtilisina/metabolismo , Animales , Sitios de Unión , Bovinos , Chlorocebus aethiops , Chaperón BiP del Retículo Endoplásmico , Escherichia coli/patogenicidad , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Ratones , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutagénesis , Especificidad por Sustrato , Células Vero
12.
Curr Opin Microbiol ; 65: 183-190, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34929548

RESUMEN

The formation of attaching and effacing (A/E) lesions on intestinal epithelium, combined with Shiga toxin production, are hallmarks of enterohemorrhagic Escherichia coli (EHEC) infection that can lead to lethal hemolytic uremic syndrome. Although an animal infection model that fully recapitulates human disease remains elusive, mice orally infected with Citrobacter rodentium(ϕStx2dact), a natural murine pathogen lysogenized with an EHEC-derived Shiga toxin 2-producing bacteriophage, develop intestinal A/E lesions and toxin-dependent systemic disease. This model has facilitated investigation of how: (A) phage gene expression and prophage induction contribute to disease and are potentially triggered by antibiotic treatment; (B) virulence gene expression is altered by microbiota and the colonic metabolomic milieu; and (C) innate immune signaling is affected by Stx. Thus, the model provides a unique tool for accessing diverse aspects of EHEC pathogenesis.


Asunto(s)
Bacteriófagos , Escherichia coli Enterohemorrágica , Infecciones por Escherichia coli , Síndrome Hemolítico-Urémico , Animales , Bacteriófagos/metabolismo , Citrobacter rodentium/genética , Citrobacter rodentium/metabolismo , Modelos Animales de Enfermedad , Escherichia coli Enterohemorrágica/metabolismo , Femenino , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/metabolismo , Síndrome Hemolítico-Urémico/patología , Humanos , Mucosa Intestinal/metabolismo , Masculino , Ratones
13.
J Infect Dis ; 202(9): 1415-23, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20874089

RESUMEN

Subtilase cytotoxin (SubAB), produced by certain virulent Shiga toxigenic Escherichia coli strains, causes hemolytic uremic syndrome-like pathology in mice, including extensive microvascular thrombosis. SubAB acts by specifically cleaving the essential endoplasmic reticulum chaperone binding immunoglobulin protein (BiP). BiP has been reported to inhibit the activation of tissue factor (TF), the major initiator of extrinsic coagulation. We hypothesized that the apparent prothrombotic effect of SubAB in vivo may involve the stimulation of TF­dependent procoagulant activity. TF­dependent procoagulant activity, TF messenger RNA (mRNA) levels, and BiP cleavage were therefore examined in human macrophage cells and primary human umbilical vein endothelial cells exposed to SubAB. In both types of cells, SubAB significantly increased TF­dependent procoagulant activity, induced TF mRNA expression, and mediated BiP cleavage. No effects were seen when cells were treated with a nonproteolytic mutant toxin, SubAA272B. Our results suggest that the procoagulant effect of SubAB may be dependent on both the up­regulation of TF expression and the activation of TF by means of BiP cleavage.


Asunto(s)
Proteínas de Escherichia coli/toxicidad , Proteínas de Choque Térmico/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Subtilisinas/toxicidad , Tromboplastina/biosíntesis , Animales , Coagulación Sanguínea/efectos de los fármacos , Células Cultivadas , Chaperón BiP del Retículo Endoplásmico , Células Endoteliales/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Perfilación de la Expresión Génica , Humanos , Macrófagos/efectos de los fármacos , Ratones , Subtilisinas/metabolismo , Trombosis/inducido químicamente
14.
Infect Immun ; 78(7): 2984-94, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20439475

RESUMEN

Shiga toxins expressed in the intestinal lumen during infection with Shiga-toxigenic Escherichia coli must translocate across the epithelium and enter the systemic circulation to cause systemic (pathological) effects, including hemolytic uremic syndrome. The transepithelial migration of polymorphonuclear leukocytes in response to chemokine expression by intestinal epithelial cells is thought to promote uptake of Stx from the intestinal lumen by compromising the epithelial barrier. In the present study, we investigated the hypothesis that flagellin acts in conjunction with Shiga toxin to augment this chemokine expression. We investigated the relative contributions of nuclear factor kappaB (NF-kappaB) and mitogen-activated protein kinase (MAPK) signaling to transcription and translation of interleukin-8. Using reporter gene constructs, we showed that flagellin-mediated interleukin-8 gene transcription is heavily dependent on both NF-kappaB and extracellular signal-regulated kinase 1 and 2 (ERK-1/2) activation. In contrast, inhibition of p38 has no detectable effect on interleukin-8 gene transcription, even though flagellin-mediated activation of host p38 is critical for maximal interleukin-8 protein expression. Inhibition of MAPK-interacting kinase 1 suggests that p38 signaling affects the posttranscriptional regulation of interleukin-8 protein expression induced by flagellin. Cotreatment with Stx2 and flagellin results in a synergistic upregulation of c-Jun N-terminal protein kinases (JNKs), p38 activation, and a superinduction of interleukin-8 mRNA. This synergism was also evident at the protein level, with increased interleukin-8 protein detectable following cotreatment with flagellin and Stx2. We propose that flagellin, in conjunction with Shiga toxin, synergistically upregulates stress-activated protein kinases, resulting in superinduction of interleukin-8 and, ultimately, absorption of Stx into the systemic circulation.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Flagelina/metabolismo , Interleucina-8/fisiología , Toxina Shiga II/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Línea Celular , Clonación Molecular , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Interleucina-8/biosíntesis , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/fisiología , Proteínas Quinasas Activadas por Mitógenos/fisiología , FN-kappa B/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
15.
Cell Microbiol ; 10(7): 1468-77, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18331592

RESUMEN

Shiga toxins (Stxs) and ricin initiate damage to host cells by cleaving a single adenine residue on the alpha-sarcin loop of the 28S ribosomal RNA. This molecular insult results in a cascade of intracellular events termed the ribotoxic stress response (RSR). Although Stxs and ricin have been shown to cause the RSR, the mitogen-activated protein kinase kinase kinase (MAP3K) that transduces the signal from intoxicated ribosomes to activate SAPKinases has remained elusive. We show in vitro that DHP-2 (7-[3-fluoro-4-aminophenyl-(4-(2-pyridin-2-yl-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl))]-quinoline), a zipper sterile-alpha-motif kinase (ZAK)-specific inhibitor, blocks Stx2/ricin-induced SAPKinase activation. Treatment of cells with DHP-2 also blocks Stx2/ricin-mediated upregulation of the proinflammatory cytokine interleukin-8 and results in a modest but statistically significant improvement in cell viability following Stx2/ricin treatment. Finally we show that siRNA directed against the N-terminus of ZAK diminishes Stx2/Ricin-induced SAPKinase activation. Together, these data demonstrate that a ZAK isoform(s) is the MAP3Kinase that transduces the RSR. Therefore, ZAKalpha and/or beta isoforms may act as potential therapeutic target(s) for treating Stx/ricin-associated illnesses. Furthermore, a small molecule inhibitor like DHP-2 may prove valuable in preventing the Stx/ricin-induced proinflammatory and/or apoptotic effects that are thought to contribute to pathogenesis by Stx-producing Escherichia coli and ricin.


Asunto(s)
Citocinas/metabolismo , Regulación de la Expresión Génica , Proteínas Quinasas/metabolismo , Ricina/metabolismo , Toxina Shiga II/metabolismo , Anciano , Animales , Línea Celular , Niño , Chlorocebus aethiops , Citocinas/genética , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Interleucina-8/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas/genética , Pirazoles/metabolismo , Quinolinas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Escherichia coli Shiga-Toxigénica/metabolismo , Escherichia coli Shiga-Toxigénica/patogenicidad , Células Vero , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Cell Microbiol ; 10(3): 795-806, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18042253

RESUMEN

Subtilase cytotoxin (SubAB) is the prototype of a new family of AB5 cytotoxins produced by Shiga toxigenic Escherichia coli. Its cytotoxic activity is due to its capacity to enter cells and specifically cleave the endoplasmic reticulum (ER) chaperone BiP. However, its trafficking within target cells has not been investigated previously. In Vero cells, fluorescence colocalization with subcellular markers established that SubAB is trafficked from the cell surface to the ER via a retrograde pathway similar, but not identical, to those of Shiga toxin (Stx) and cholera toxin (Ctx), with their pathways converging at the Golgi. The clathrin inhibitor phenylarsine oxide prevented SubAB entry and BiP cleavage in SubAB-treated Vero, HeLa and N2A cells, while cholesterol depletion did not, demonstrating that, unlike either Stx or Ctx, SubAB internalization is exclusively clathrin-dependent.


Asunto(s)
Clatrina/metabolismo , Retículo Endoplásmico/enzimología , Proteínas de Escherichia coli/metabolismo , Proteínas de Choque Térmico/antagonistas & inhibidores , Chaperonas Moleculares/antagonistas & inhibidores , Escherichia coli Shiga-Toxigénica/fisiología , Subtilisinas/metabolismo , Animales , Arsenicales/farmacología , Chlorocebus aethiops , Clatrina/antagonistas & inhibidores , Retículo Endoplásmico/química , Chaperón BiP del Retículo Endoplásmico , Inhibidores Enzimáticos/farmacología , Aparato de Golgi/química , Células HeLa , Humanos , Microscopía Fluorescente , Células Vero
17.
Cell Microbiol ; 10(9): 1775-86, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18433465

RESUMEN

Subtilase cytotoxin (SubAB) is the prototype of a new family of AB(5) cytotoxins produced by Shiga toxigenic Escherichia coli. Its cytotoxic activity is due to its capacity to enter cells and specifically cleave the essential endoplasmic reticulum (ER) chaperone BiP (GRP78). In the present study, we have examined its capacity to trigger the three ER stress-signalling pathways in Vero cells. Activation of PKR-like ER kinase was demonstrated by phosphorylation of eIF2alpha, which occurred within 30 min of toxin treatment, and correlated with inhibition of global protein synthesis. Activation of inositol-requiring enzyme 1 was demonstrated by splicing of X-box-binding protein 1 mRNA, while activating transcription factor 6 activation was demonstrated by depletion of the 90 kDa uncleaved form, and appearance of the 50 kDa cleaved form. The rapidity with which ER stress-signalling responses are triggered by exposure of cells to SubAB is consistent with the hypothesis that cleavage by the toxin causes BiP to dissociate from the signalling molecules.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Escherichia coli Shiga-Toxigénica/metabolismo , Subtilisinas/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Chlorocebus aethiops , Transducción de Señal , Células Vero
18.
PLoS One ; 13(8): e0199810, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30071046

RESUMEN

Ridinilazole, a novel targeted antibacterial being developed for the treatment of C. difficile infection (CDI) and prevention of recurrence, was shown in a recent Phase 2 study to be superior to vancomycin with regard to the primary efficacy measure, sustained clinical response (SCR), with the superiority being driven primarily by marked reductions in the rates of CDI recurrence within 30 days. Tolerability of ridinilazole was comparable to that of vancomycin. The current nested cohort study compared the effects of ridinilazole and vancomycin on fecal microbiota during and after treatment among participants in the Phase 2 study. Changes in the microbiota were assessed using qPCR and high-throughput sequencing on participants' stools collected at multiple time-points (baseline [Day 1], Day 5, end-of-treatment [EOT; Day 10], Day 25, end-of-study [EOS; Day 40], and at CDI recurrence). qPCR analyses showed profound losses of Bacteroides, C. coccoides, C. leptum, and Prevotella groups at EOT with vancomycin treatment, while ridinilazole-treated participants had a modest decrease in C. leptum group levels at EOT, with levels recovering by Day 25. Vancomycin-treated participants had a significant increase in the Enterobacteriaceae group, with this increase persisting beyond EOT. At EOT, alpha diversity decreased with both antibiotics, though to a significantly lesser extent with ridinilazole (p <0.0001). Beta diversity analysis showed a significantly larger weighted Unifrac distance from baseline-to-EOT with vancomycin. Taxonomically, ridinilazole had a markedly narrower impact, with modest reductions in relative abundance in Firmicutes taxa. Microbiota composition returned to baseline sooner with ridinilazole than with vancomycin. Vancomycin treatment resulted in microbiome-wide changes, with significant reductions in relative abundances of Firmicutes, Bacteroidetes, Actinobacteria, and a profound increase in abundance of Proteobacteria. These findings demonstrate that ridinilazole is significantly less disruptive to microbiota than vancomycin, which may contribute to the reduced CDI recurrence observed in the Phase 2 study.


Asunto(s)
Antibacterianos/uso terapéutico , Bencimidazoles/uso terapéutico , Piridinas/uso terapéutico , Vancomicina/uso terapéutico , Actinobacteria/efectos de los fármacos , Actinobacteria/aislamiento & purificación , Antibacterianos/farmacología , Bencimidazoles/farmacología , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/genética , Clostridioides difficile/aislamiento & purificación , Infecciones por Clostridium/tratamiento farmacológico , Infecciones por Clostridium/microbiología , Estudios de Cohortes , ADN Bacteriano/química , ADN Bacteriano/aislamiento & purificación , ADN Bacteriano/metabolismo , Análisis Discriminante , Heces/microbiología , Firmicutes/efectos de los fármacos , Firmicutes/aislamiento & purificación , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Análisis de Componente Principal , Proteobacteria/efectos de los fármacos , Proteobacteria/aislamiento & purificación , Piridinas/farmacología , Vancomicina/farmacología
19.
Clin Infect Dis ; 43(12): 1587-95, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17109294

RESUMEN

In 1982, hemorrhagic colitis and hemolytic-uremic syndrome were linked to infection with Escherichia coli O157:H7, a serotype now classified as Shiga toxin-producing E. coli (STEC). Thereafter, hemorrhagic colitis and hemolytic-uremic syndrome associated with non-O157 STEC serogroups were reported, with the frequency of non-O157 STEC illness rivaling that of O157:H7 in certain geographic regions. In the United States, non-O157 E. coli may account for up to 20%-50% of all STEC infections. A high index of suspicion, paired with options to test for non-O157 STEC infection, are necessary for early recognition and appropriate treatment of these infections. Supportive care without the use of antibiotics is currently considered to be optimal treatment for all STEC infections. This commentary provides a perspective on the non-O157 STEC as human pathogens, how and when the clinician should approach the diagnosis of these organisms, and the challenges ahead.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Escherichia coli O157/patogenicidad , Virulencia , Técnicas de Tipificación Bacteriana , Colitis/etiología , Colitis/microbiología , Infecciones por Escherichia coli/complicaciones , Escherichia coli O157/clasificación , Síndrome Hemolítico-Urémico/etiología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Serotipificación , Toxinas Shiga
20.
Toxins (Basel) ; 8(9)2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27598200

RESUMEN

Ricin activates the proinflammatory ribotoxic stress response through the mitogen activated protein 3 kinase (MAP3K) ZAK, resulting in activation of mitogen activated protein kinases (MAPKs) p38 and JNK1/2. We had a novel zak-/- mouse generated to study the role of ZAK signaling in vivo during ricin intoxication. To characterize this murine strain, we intoxicated zak-/- and zak+/+ bone marrow-derived murine macrophages with ricin, measured p38 and JNK1/2 activation by Western blot, and measured zak, c-jun, and cxcl-1 expression by qRT-PCR. To determine whether zak-/- mice differed from wild-type mice in their in vivo response to ricin, we performed oral ricin intoxication experiments with zak+/+ and zak-/- mice, using blinded histopathology scoring of duodenal tissue sections to determine differences in tissue damage. Unlike macrophages derived from zak+/+ mice, those derived from the novel zak-/- strain fail to activate p38 and JNK1/2 and have decreased c-jun and cxcl-1 expression following ricin intoxication. Furthermore, compared with zak+/+ mice, zak-/- mice have decreased duodenal damage following in vivo ricin challenge. zak-/- mice demonstrate a distinct ribotoxic stress-associated phenotype in response to ricin and therefore provide a new animal model for in vivo studies of ZAK signaling.


Asunto(s)
Duodeno/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/deficiencia , Macrófagos/efectos de los fármacos , Ricina/toxicidad , Estrés Fisiológico/efectos de los fármacos , Animales , Células Cultivadas , Quimiocina CXCL1/metabolismo , Duodeno/enzimología , Duodeno/patología , Activación Enzimática , Genotipo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Macrófagos/enzimología , Macrófagos/patología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA