Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 210(1): 35-46, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38754132

RESUMEN

Rationale: Pseudomonas aeruginosa infection is associated with worse outcomes in bronchiectasis. Impaired neutrophil antimicrobial responses contribute to bacterial persistence. Gremubamab is a bivalent, bispecific monoclonal antibody targeting Psl exopolysaccharide and the type 3 secretion system component PcrV. Objectives: This study evaluated the efficacy of gremubamab to enhance killing of P. aeruginosa by neutrophils from patients with bronchiectasis and to prevent P. aeruginosa-associated cytotoxicity. Methods: P. aeruginosa isolates from a global bronchiectasis cohort (n = 100) underwent whole-genome sequencing to determine target prevalence. Functional activity of gremubamab against selected isolates was tested in vitro and in vivo. Patients with bronchiectasis (n = 11) and control subjects (n = 10) were enrolled, and the effect of gremubamab in peripheral blood neutrophil opsonophagocytic killing (OPK) assays against P. aeruginosa was evaluated. Serum antibody titers to Psl and PcrV were determined (n = 30; 19 chronic P. aeruginosa infection, 11 no known P. aeruginosa infection), as was the effect of gremubamab treatment in OPK and anti-cytotoxic activity assays. Measurements and Main Results: Psl and PcrV were conserved in isolates from chronically infected patients with bronchiectasis. Seventy-three of 100 isolates had a full psl locus, and 99 of 100 contained the pcrV gene, with 20 distinct full-length PcrV protein subtypes identified. PcrV subtypes were successfully bound by gremubamab and the monoclonal antibody-mediated potent protective activity against tested isolates. Gremubamab increased bronchiectasis patient neutrophil-mediated OPK (+34.6 ± 8.1%) and phagocytosis (+70.0 ± 48.8%), similar to effects observed in neutrophils from control subjects (OPK, +30.1 ± 7.6%). No evidence of competition between gremubamab and endogenous antibodies was found, with protection against P. aeruginosa-induced cytotoxicity and enhanced OPK demonstrated with and without addition of patient serum. Conclusions: Gremubamab enhanced bronchiectasis patient neutrophil phagocytosis and killing of P. aeruginosa and reduced virulence.


Asunto(s)
Anticuerpos Biespecíficos , Bronquiectasia , Neutrófilos , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Humanos , Bronquiectasia/inmunología , Bronquiectasia/microbiología , Pseudomonas aeruginosa/inmunología , Neutrófilos/inmunología , Neutrófilos/efectos de los fármacos , Anticuerpos Biespecíficos/uso terapéutico , Anticuerpos Biespecíficos/farmacología , Femenino , Masculino , Infecciones por Pseudomonas/inmunología , Persona de Mediana Edad , Anciano , Adulto , Antígenos Bacterianos , Toxinas Bacterianas , Proteínas Citotóxicas Formadoras de Poros
2.
Artículo en Inglés | MEDLINE | ID: mdl-32152087

RESUMEN

Antibiotics revolutionized the treatment of infectious diseases; however, it is now clear that broad-spectrum antibiotics alter the composition and function of the host's microbiome. The microbiome plays a key role in human health, and its perturbation is increasingly recognized as contributing to many human diseases. Widespread broad-spectrum antibiotic use has also resulted in the emergence of multidrug-resistant pathogens, spurring the development of pathogen-specific strategies such as monoclonal antibodies (MAbs) to combat bacterial infection. Not only are pathogen-specific approaches not expected to induce resistance in nontargeted bacteria, but they are hypothesized to have minimal impact on the gut microbiome. Here, we compare the effects of antibiotics, pathogen-specific MAbs, and their controls (saline or control IgG [c-IgG]) on the gut microbiome of 7-week-old, female, C57BL/6 mice. The magnitude of change in taxonomic abundance, bacterial diversity, and bacterial metabolites, including short-chain fatty acids (SCFA) and bile acids in the fecal pellets from mice treated with pathogen-specific MAbs, was no different from that with animals treated with saline or an IgG control. Conversely, dramatic changes were observed in the relative abundance, as well as alpha and beta diversity, of the fecal microbiome and bacterial metabolites in the feces of all antibiotic-treated mice. Taken together, these results indicate that pathogen-specific MAbs do not alter the fecal microbiome like broad-spectrum antibiotics and may represent a safer, more-targeted approach to antibacterial therapy.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Ácidos y Sales Biliares/metabolismo , ADN Bacteriano/análisis , Ácidos Grasos/metabolismo , Heces/microbiología , Femenino , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , Organismos Libres de Patógenos Específicos
3.
J Biol Chem ; 293(3): 941-952, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29180448

RESUMEN

Clostridium difficile infection is the leading cause of hospital-acquired diarrhea and is mediated by the actions of two toxins, TcdA and TcdB. The toxins perturb host cell function through a multistep process of receptor binding, endocytosis, low pH-induced pore formation, and the translocation and delivery of an N-terminal glucosyltransferase domain that inactivates host GTPases. Infection studies with isogenic strains having defined toxin deletions have established TcdB as an important target for therapeutic development. Monoclonal antibodies that neutralize TcdB function have been shown to protect against C. difficile infection in animal models and reduce recurrence in humans. Here, we report the mechanism of TcdB neutralization by PA41, a humanized monoclonal antibody capable of neutralizing TcdB from a diverse array of C. difficile strains. Through a combination of structural, biochemical, and cell functional studies, involving X-ray crystallography and EM, we show that PA41 recognizes a single, highly conserved epitope on the TcdB glucosyltransferase domain and blocks productive translocation and delivery of the enzymatic cargo into the host cell. Our study reveals a unique mechanism of C. difficile toxin neutralization by a monoclonal antibody, which involves targeting a process that is conserved across the large clostridial glucosylating toxins. The PA41 antibody described here provides a valuable tool for dissecting the mechanism of toxin pore formation and translocation across the endosomal membrane.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Enterotoxinas/metabolismo , Anticuerpos Monoclonales/metabolismo , Toxinas Bacterianas/química , Células CACO-2 , Clostridioides difficile/enzimología , Cristalografía por Rayos X , Citosol/metabolismo , Enterotoxinas/química , Humanos , Concentración de Iones de Hidrógeno , Microscopía Electrónica , Rubidio/química , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/metabolismo
4.
Am J Respir Cell Mol Biol ; 59(6): 745-756, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30109945

RESUMEN

Pseudomonas aeruginosa and Klebsiella pneumoniae are two common gram-negative pathogens that are associated with bacterial pneumonia and can often be isolated from the same patient. We used a mixed-pathogen pneumonia infection model in which mice were infected with sublethal concentrations of P. aeruginosa and K. pneumoniae, resulting in significant lethality, outgrowth of both bacteria in the lung, and systemic dissemination of K. pneumoniae. Inflammation, induced by P. aeruginosa activation of Toll-like receptor 5, results in prolonged neutrophil recruitment to the lung and increased levels of neutrophil elastase in the airway, resulting in lung damage and epithelial barrier dysfunction. Live P. aeruginosa was not required to potentiate K. pneumoniae infection, and flagellin alone was sufficient to induce lethality when delivered along with Klebsiella. Prophylaxis with an anti-Toll-like receptor 5 antibody or Sivelestat, a neutrophil elastase inhibitor, reduced neutrophil influx, inflammation, and mortality. Furthermore, pathogen-specific monoclonal antibodies targeting P. aeruginosa or K. pneumoniae prevented the outgrowth of both bacteria and reduced host inflammation and lethality. These findings suggest that coinfection with P. aeruginosa may enable the outgrowth and dissemination of K. pneumoniae, and that a pathogen- or host-specific prophylactic approach targeting P. aeruginosa may prevent or limit the severity of such infections by reducing neutrophil-induced lung damage.


Asunto(s)
Coinfección/inmunología , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , Células Cultivadas , Coinfección/microbiología , Coinfección/patología , Femenino , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/patología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Neutrófilos/microbiología , Neutrófilos/patología , Neumonía/microbiología , Neumonía/patología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , Receptor Toll-Like 5/metabolismo
5.
J Biol Chem ; 292(35): 14401-14412, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28705932

RESUMEN

Clostridium difficile is a clinically significant pathogen that causes mild-to-severe (and often recurrent) colon infections. Disease symptoms stem from the activities of two large, multidomain toxins known as TcdA and TcdB. The toxins can bind, enter, and perturb host cell function through a multistep mechanism of receptor binding, endocytosis, pore formation, autoproteolysis, and glucosyltransferase-mediated modification of host substrates. Monoclonal antibodies that neutralize toxin activity provide a survival benefit in preclinical animal models and prevent recurrent infections in human clinical trials. However, the molecular mechanisms involved in these neutralizing activities are unclear. To this end, we performed structural studies on a neutralizing monoclonal antibody, PA50, a humanized mAb with both potent and broad-spectrum neutralizing activity, in complex with TcdA. Electron microscopy imaging and multiangle light-scattering analysis revealed that PA50 binds multiple sites on the TcdA C-terminal combined repetitive oligopeptides (CROPs) domain. A crystal structure of two PA50 Fabs bound to a segment of the TcdA CROPs helped define a conserved epitope that is distinct from previously identified carbohydrate-binding sites. Binding of TcdA to the host cell surface was directly blocked by either PA50 mAb or Fab and suggested that receptor blockade is the mechanism by which PA50 neutralizes TcdA. These findings highlight the importance of the CROPs C terminus in cell-surface binding and a role for neutralizing antibodies in defining structural features critical to a pathogen's mechanism of action. We conclude that PA50 protects host cells by blocking the binding of TcdA to cell surfaces.


Asunto(s)
Antibacterianos/metabolismo , Anticuerpos Neutralizantes/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/enzimología , Enterocitos/metabolismo , Enterotoxinas/metabolismo , Glucosiltransferasas/metabolismo , Modelos Moleculares , Secuencia de Aminoácidos , Antibacterianos/química , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/metabolismo , Anticuerpos Neutralizantes/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/toxicidad , Sitios de Unión de Anticuerpos , Células CACO-2 , Secuencia Conservada , Cristalografía por Rayos X , Enterocitos/efectos de los fármacos , Enterotoxinas/química , Enterotoxinas/genética , Enterotoxinas/toxicidad , Mapeo Epitopo , Glucosiltransferasas/química , Glucosiltransferasas/genética , Glucosiltransferasas/toxicidad , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/toxicidad , Secuencias Repetitivas de Aminoácido
6.
J Infect Dis ; 213(4): 640-8, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26333940

RESUMEN

BACKGROUND: The type 3 secretion protein PcrV and Psl exopolysaccharide are promising therapeutic antibody targets against Pseudomonas aeruginosa. We examined P. aeruginosa bloodstream infection (BSI) isolates for the ability to express PcrV and Psl and evaluated corresponding patient serum for active titers to these targets. METHODS: We identified 114 patients with acute P. aeruginosa BSI; 56 cases were accompanied by acute sera. Serum was evaluated for PcrV- and Psl-specific immunoglobulin G (IgG) and for cytotoxicity and opsonophagocytosis. Isolates were evaluated for susceptibility to antibiotics, expression of PcrV and Psl, and susceptibility to the anti-PcrV/Psl bispecific antibody and clinical candidate MEDI3902. RESULTS: In-hospital mortality for patients with P. aeruginosa BSI was 39%. A total of 26% of isolates were resistant to ≥3 antibiotic classes. Although PcrV and/or Psl were detected in 99% of isolates, a majority of patients lacked active titers to PcrV (100%) and Psl (98%). In addition, MEDI3902 was active against all tested isolates. CONCLUSIONS: A vast majority of P. aeruginosa BSI isolates express PcrV and Psl; however, patient sera most often lacked IgG and functionally active responses to these targets. These results suggest that therapies directed at PcrV and Psl could be a promising approach for combating P. aeruginosa bloodstream infections.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/inmunología , Bacteriemia/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , Antibacterianos/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Persona de Mediana Edad , Proteínas Opsoninas/sangre , Fagocitosis , Estudios Prospectivos , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/aislamiento & purificación
7.
Antimicrob Agents Chemother ; 58(8): 4384-91, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24841258

RESUMEN

Pseudomonas aeruginosa is a major cause of hospital-acquired infections, particularly in mechanically ventilated patients, and it is the leading cause of death in cystic fibrosis patients. A key virulence factor associated with disease severity is the P. aeruginosa type III secretion system (T3SS), which injects bacterial toxins directly into the cytoplasm of host cells. The PcrV protein, located at the tip of the T3SS injectisome complex, is required for T3SS function and is a well-validated target in animal models of immunoprophylactic strategies targeting P. aeruginosa. In an effort to identify a highly potent and protective monoclonal antibody (MAb) that inhibits the T3SS, we generated and characterized a panel of novel anti-PcrV MAbs. Interestingly, some MAbs exhibiting potent inhibition of T3SS in vitro failed to provide protection in a mouse model of P. aeruginosa infection, suggesting that effective in vivo inhibition of T3SS with anti-PcrV MAbs is epitope dependent. V2L2MD, while not the most potent MAb as assessed by in vitro cytotoxicity inhibition assays, provided strong prophylactic protection in several murine infection models and a postinfection therapeutic model. V2L2MD mediated significantly (P < 0.0001) better in vivo protection than that provided by a comparator antibody, MAb166, a well-characterized anti-PcrV MAb and the progenitor of a clinical candidate, KB001-A. The results described here support further development of a V2L2MD-containing immunotherapeutic and may suggest even greater potential than was previously recognized for the prevention and treatment of P. aeruginosa infections in high-risk populations.


Asunto(s)
Anticuerpos Antibacterianos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Antígenos Bacterianos/inmunología , Toxinas Bacterianas/inmunología , Inmunización Pasiva , Proteínas Citotóxicas Formadoras de Poros/inmunología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Monoclonales/biosíntesis , Antígenos Bacterianos/química , Sistemas de Secreción Bacterianos/inmunología , Toxinas Bacterianas/química , Pruebas Inmunológicas de Citotoxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta Inmunológica , Epítopos/química , Epítopos/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Ratones , Proteínas Citotóxicas Formadoras de Poros/química , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/mortalidad , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/inmunología , Análisis de Supervivencia
8.
Microbiol Spectr ; : e0040024, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38940542

RESUMEN

We used phage display, antibody engineering, and high-throughput assays to identify antibody-accessible targets of Klebsiella pneumoniae. We report the discovery of monoclonal antibodies (mAbs) binding to type 3 fimbrial proteins, including MrkA. We found that anti-MrkA mAbs were cross-reactive to a diverse panel of K. pneumoniae clinical isolates, representing different O-serotypes. mAbs binding to MrkA have previously been described and have been shown to provide prophylactic protection, although only modest protection when dosed therapeutically in vivo in a murine lung infection model. Here, we used a combination of binding and opsonophagocytic killing studies using a high-content imaging platform to provide a possible explanation for the modest therapeutic efficacy in vivo reported in that model. Our work shows that expression of K. pneumoniae type 3 fimbriae in in vitro culture is not homogenous within a bacterial population. Instead, sub-populations of bacteria that do, and do not, express type 3 fimbriae exist. In a high-content opsonophagocytic killing assay, we showed that MrkA-targeting antibodies initially promote killing by macrophages; however, over time, this effect is diminished. We hypothesize the reason for this is that bacteria not expressing MrkA can evade opsonophagocytosis. Our data support the fact that MrkA is a conserved, immunodominant protein that is antibody accessible on the surface of K. pneumoniae and suggest that additional studies should evaluate the potential of using anti-MrkA antibodies in different stages of K. pneumoniae infection (different sites in the body) as well as against K. pneumoniae biofilms in the body during infection and associated with medical devices.IMPORTANCEThere is an unmet, urgent need for the development of novel antimicrobial therapies for the treatment of Klebsiella pneumoniae infections. We describe the use of phage display, antibody engineering, and high-throughput assays to identify antibody-accessible targets of K. pneumoniae. We discovered monoclonal antibodies (mAbs) binding to the type 3 fimbrial protein MrkA. The anti-MrkA mAbs were found to be highly cross-reactive, binding to all K. pneumoniae strains tested from a diverse panel of clinical isolates, and were active in an opsonophagocytic killing assay at pM concentrations. MrkA is important for biofilm formation; thus, our data support further exploration of the use of anti-MrkA antibodies for preventing and/or controlling K. pneumoniae in biofilms and during infection.

9.
Apoptosis ; 17(7): 691-701, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22392482

RESUMEN

Tumor suppressor genes BRCA1 and BRCA2 function in a complex gene network that regulates homologous recombination and DNA double-strand break repair. Disruption of the BRCA-network through gene mutation, deletion, or RNAi-mediated silencing can sensitize cells to small molecule inhibitors of poly (ADP-ribose) polymerase (PARPi). Here, we demonstrate that BRCA-network disruption in the presence of PARPi leads to the selective induction and enhancement of interferon pathway and apoptotic gene expression in cultured tumor cells. In addition, we report PARPi cytotoxicity in BRCA1-deficient tumor cells is enhanced >10-fold when combined with interferon-γ. These findings establish a link between synthetic lethality of PARPi in BRCA-network disrupted cells and interferon pathway activation triggered by genetic instability.


Asunto(s)
Proteína BRCA1/genética , Redes Reguladoras de Genes/genética , Interferón gamma/metabolismo , Interferón gamma/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína BRCA1/metabolismo , Ciclo Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Células HT29 , Células HeLa , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/genética
10.
MAbs ; 14(1): 2006123, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34923908

RESUMEN

The increasing global occurrence of recalcitrant multi-drug resistant Klebsiella pneumoniae infections warrants the investigation of alternative therapy options, such as the use of monoclonal antibodies (mAbs). We used a target-agnostic phage display approach to K. pneumoniae bacteria lacking bulky, highly variable surface polysaccharides in order to isolate antibodies targeting conserved epitopes among clinically relevant strains. One antibody population contained a high proportion of unique carbohydrate binders, and biolayer interferometry revealed these antibodies bound to lipopolysaccharide (LPS). Antibodies that bound to O1 and O1/O2 LPS were identified. Antibodies were found to promote opsonophagocytic killing by human monocyte-derived macrophages and clearance of macrophage-associated bacteria when assessed using high-content imaging. One antibody, B39, was found to protect mice in a lethal model of K. pneumoniae pneumonia against both O1 and O2 strains when dosed therapeutically. High-content imaging, western blotting and fluorescence-activated cell sorting were used to determine binding to a collection of clinical K. pneumoniae O1 and O2 strains. The data suggests B39 binds to D-galactan-I and D-galactan-II of the LPS of O1 and O2 strains. Thus, we have discovered an mAb with novel binding and functional activity properties that is a promising candidate for development as a novel biotherapeutic for the treatment and prevention of K. pneumoniae infections.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Epítopos/inmunología , Infecciones por Klebsiella/inmunología , Klebsiella pneumoniae/inmunología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Animales , Farmacorresistencia Bacteriana Múltiple/genética , Farmacorresistencia Bacteriana Múltiple/inmunología , Epítopos/genética , Humanos , Infecciones por Klebsiella/genética , Klebsiella pneumoniae/genética , Lipopolisacáridos/genética , Ratones , Opsonización
11.
Microorganisms ; 9(7)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209573

RESUMEN

The gut microbiota has emerged as a key mediator of human physiology, and germ-free mice have been essential in demonstrating a role for the microbiome in disease. Preclinical models using conventional mice offer the advantage of working with a mature immune system. However, optimal protocols for fecal microbiota transplant (FMT) engraftment in conventional mice are yet to be established. Conventional BALB/c mice were randomized to receive 3-day (3d) or 3-week (3w) antibiotic (ABX) regimen in their drinking water followed by 1 or 5-daily FMTs from a human donor. Fecal samples were collected longitudinally and characterized using 16S ribosomal RNA (rRNA) sequencing. Semi-targeted metabolomic profiling of fecal samples was also done with liquid chromatography-mass spectrometry (LC-MS). Lastly, we sought to confirm our findings in BKS mice. Recovery of baseline diversity scores were greatest in the 3d groups, driven by re-emergence of mouse commensal microbiota, whereas the most resemblance to donor microbiota was seen in the 3w + 5-FMT group. Amplicon sequence variants (ASVs) that were linked to the input material (human ASVs) engrafted to a significantly greater extent when compared to mouse ASVs in the 3-week groups but not the 3-day groups. Lastly, comparison of metabolomic profiles revealed distinct functional profiles by ABX regimen. These results indicate successful model optimization and emphasize the importance of ABX duration and frequency of FMT dosing; the most stable and reliable colonization by donor ASVs was seen in the 3wk + 5-FMT group.

12.
Mol Cell Biol ; 26(10): 3853-63, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648480

RESUMEN

KIF14 is a microtubule motor protein whose elevated expression is associated with poor-prognosis breast cancer. Here we demonstrate KIF14 accumulation in mitotic cells, where it associated with developing spindle poles and spindle microtubules. Cells at later stages of mitosis were characterized by the concentration of KIF14 at the midbody. Time-lapse microscopy revealed that strong RNA interference (RNAi)-mediated silencing of KIF14 induced cytokinesis failure, causing several rounds of endoreduplication and resulting in multinucleated cells. Additionally, less efficacious KIF14-specific short interfering RNAs (siRNAs) induced multiple phenotypes, all of which resulted in acute apoptosis. Our data demonstrate the ability of siRNA-mediated silencing to generate epiallelic hypomorphs associated with KIF14 depletion. Furthermore, the link we observed between siRNA efficacy and phenotypic outcome indicates that distinct stages during cell cycle progression are disrupted by the differential modulation of KIF14 expression.


Asunto(s)
Ciclo Celular/fisiología , Citocinesis/fisiología , Silenciador del Gen , Cinesinas/metabolismo , Proteínas Oncogénicas/metabolismo , Interferencia de ARN , Adenosina Trifosfatasas/análisis , Secuencia de Aminoácidos , Apoptosis , Línea Celular , Clonación Molecular , Secuencia de Consenso , Factor de Crecimiento Epidérmico/metabolismo , Colorantes Fluorescentes , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HeLa , Humanos , Immunoblotting , Indoles , Cinesinas/química , Cinesinas/genética , Microscopía Fluorescente , Microscopía por Video , Datos de Secuencia Molecular , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Penetrancia , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética
13.
Mol Cell Biol ; 26(24): 9377-86, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17000754

RESUMEN

RNA interference technology allows the systematic genetic analysis of the molecular alterations in cancer cells and how these alterations affect response to therapies. Here we used small interfering RNA (siRNA) screens to identify genes that enhance the cytotoxicity (enhancers) of established anticancer chemotherapeutics. Hits identified in drug enhancer screens of cisplatin, gemcitabine, and paclitaxel were largely unique to the drug being tested and could be linked to the drug's mechanism of action. Hits identified by screening of a genome-scale siRNA library for cisplatin enhancers in TP53-deficient HeLa cells were significantly enriched for genes with annotated functions in DNA damage repair as well as poorly characterized genes likely having novel functions in this process. We followed up on a subset of the hits from the cisplatin enhancer screen and validated a number of enhancers whose products interact with BRCA1 and/or BRCA2. TP53(+/-) matched-pair cell lines were used to determine if knockdown of BRCA1, BRCA2, or validated hits that associate with BRCA1 and BRCA2 selectively enhances cisplatin cytotoxicity in TP53-deficient cells. Silencing of BRCA1, BRCA2, or BRCA1/2-associated genes enhanced cisplatin cytotoxicity approximately 4- to 7-fold more in TP53-deficient cells than in matched TP53 wild-type cells. Thus, tumor cells having disruptions in BRCA1/2 network genes and TP53 together are more sensitive to cisplatin than cells with either disruption alone.


Asunto(s)
Antineoplásicos/toxicidad , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA2/antagonistas & inhibidores , Cisplatino/toxicidad , Neoplasias/tratamiento farmacológico , Neoplasias/patología , ARN Interferente Pequeño/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Células HeLa , Humanos , Neoplasias/genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/biosíntesis
14.
Cell Host Microbe ; 25(4): 526-536.e4, 2019 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-30930127

RESUMEN

Bacterial biofilm infections are difficult to eradicate because of antibiotic insusceptibility and high recurrence rates. Biofilm formation by Pseudomonas aeruginosa, a leading cause of bacterial keratitis, is facilitated by the bacterial Psl exopolysaccharide and associated with heightened virulence. Using intravital microscopy, we observed that neutrophilic recruitment to corneal infections limits P. aeruginosa biofilms to the outer eye surface, preventing bacterial dissemination. Neutrophils moved to the base of forming biofilms, where they underwent neutrophil extracellular trap formation (NETosis) in response to high expression of the bacterial type-3 secretion system (T3SS). NETs formed a barrier "dead zone," confining bacteria to the external corneal environment and inhibiting bacterial dissemination into the brain. Once formed, ocular biofilms were resistant to antibiotics and neutrophil killing, advancing eye pathology. However, blocking both Psl and T3SS together with antibiotic treatment broke down the biofilm and reversed keratitis, suggesting future therapeutic strategies for this intractable infection.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Córnea/microbiología , Trampas Extracelulares/metabolismo , Meningoencefalitis/prevención & control , Neutrófilos/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , Modelos Animales de Enfermedad , Ratones , Infecciones por Pseudomonas/complicaciones , Pseudomonas aeruginosa/crecimiento & desarrollo
15.
World J Gastroenterol ; 25(33): 4904-4920, 2019 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-31543682

RESUMEN

BACKGROUND: The trans-fat containing AMLN (amylin liver non-alcoholic steatohepatitis, NASH) diet has been extensively validated in C57BL/6J mice with or without the Lepob/Lepob (ob/ob) mutation in the leptin gene for reliably inducing metabolic and liver histopathological changes recapitulating hallmarks of NASH. Due to a recent ban on trans-fats as food additive, there is a marked need for developing a new diet capable of promoting a compatible level of disease in ob/ob and C57BL/6J mice. AIM: To develop a biopsy-confirmed mouse model of NASH based on an obesogenic diet with trans-fat substituted by saturated fat. METHODS: Male ob/ob mice were fed AMLN diet or a modified AMLN diet with trans-fat (Primex shortening) substituted by equivalent amounts of palm oil [Gubra amylin NASH, (GAN) diet] for 8, 12 and 16 wk. C57BL/6J mice were fed the same diets for 28 wk. AMLN and GAN diets had similar caloric content (40% fat kcal), fructose (22%) and cholesterol (2%) level. RESULTS: The GAN diet was more obesogenic compared to the AMLN diet and impaired glucose tolerance. Biopsy-confirmed steatosis, lobular inflammation, hepatocyte ballooning, fibrotic liver lesions and hepatic transcriptome changes were similar in ob/ob mice fed the GAN or AMLN diet. C57BL/6J mice developed a mild to moderate fibrotic NASH phenotype when fed the same diets. CONCLUSION: Substitution of Primex with palm oil promotes a similar phenotype of biopsy-confirmed NASH in ob/ob and C57BL/6J mice, making GAN diet-induced obese mouse models suitable for characterizing novel NASH treatments.


Asunto(s)
Modelos Animales de Enfermedad , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/etiología , Aceite de Palma/efectos adversos , Animales , Biopsia , Dieta Alta en Grasa/efectos adversos , Humanos , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/patología , Ácidos Grasos trans/efectos adversos
16.
MAbs ; 10(1): 104-117, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28952876

RESUMEN

C5a is a potent anaphylatoxin that modulates inflammation through the C5aR1 and C5aR2 receptors. The molecular interactions between C5a-C5aR1 receptor are well defined, whereas C5a-C5aR2 receptor interactions are poorly understood. Here, we describe the generation of a human antibody, MEDI7814, that neutralizes C5a and C5adesArg binding to the C5aR1 and C5aR2 receptors, without affecting complement-mediated bacterial cell killing. Unlike other anti-C5a mAbs described, this antibody has been shown to inhibit the effects of C5a by blocking C5a binding to both C5aR1 and C5aR2 receptors. The crystal structure of the antibody in complex with human C5a reveals a discontinuous epitope of 22 amino acids. This is the first time the epitope for an antibody that blocks C5aR1 and C5aR2 receptors has been described, and this work provides a basis for molecular studies aimed at further understanding the C5a-C5aR2 receptor interaction. MEDI7814 has therapeutic potential for the treatment of acute inflammatory conditions in which both C5a receptors may mediate inflammation, such as sepsis or renal ischemia-reperfusion injury.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Afinidad de Anticuerpos , Complemento C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptores de Quimiocina/antagonistas & inhibidores , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Complemento C5a/química , Complemento C5a/inmunología , Complemento C5a/metabolismo , Mapeo Epitopo/métodos , Epítopos , Células HEK293 , Humanos , Unión Proteica , Conformación Proteica , Ingeniería de Proteínas , Receptor de Anafilatoxina C5a/química , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Receptores de Quimiocina/química , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Relación Estructura-Actividad
17.
J Clin Invest ; 127(6): 2249-2261, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28463232

RESUMEN

Pseudomonas aeruginosa is a major cause of severe infections that lead to bacteremia and high patient mortality. P. aeruginosa has evolved numerous evasion and subversion mechanisms that work in concert to overcome immune recognition and effector functions in hospitalized and immunosuppressed individuals. Here, we have used multilaser spinning-disk intravital microscopy to monitor the blood-borne stage in a murine bacteremic model of P. aeruginosa infection. P. aeruginosa adhered avidly to lung vasculature, where patrolling neutrophils and other immune cells were virtually blind to the pathogen's presence. This cloaking phenomenon was attributed to expression of Psl exopolysaccharide. Although an anti-Psl mAb activated complement and enhanced neutrophil recognition of P. aeruginosa, neutrophil-mediated clearance of the pathogen was suboptimal owing to a second subversion mechanism, namely the type 3 secretion (T3S) injectisome. Indeed, T3S prevented phagosome acidification and resisted killing inside these compartments. Antibody-mediated inhibition of the T3S protein PcrV did not enhance bacterial phagocytosis but did enhance killing of the few bacteria ingested by neutrophils. A bispecific mAb targeting both Psl and PcrV enhanced neutrophil uptake of P. aeruginosa and also greatly increased inhibition of T3S function, allowing for phagosome acidification and bacterial killing. These data highlight the need to block multiple evasion and subversion mechanisms in tandem to kill P. aeruginosa.


Asunto(s)
Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Animales , Anticuerpos Biespecíficos , Antígenos Bacterianos/inmunología , Carga Bacteriana , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Proteínas del Sistema Complemento/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Macrófagos del Hígado/microbiología , Pulmón/irrigación sanguínea , Pulmón/microbiología , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Microvasos/microbiología , Neutrófilos/inmunología , Neutrófilos/microbiología , Fagocitosis , Proteínas Citotóxicas Formadoras de Poros/inmunología , Infecciones por Pseudomonas/inmunología , Receptores Fc/metabolismo
18.
JCI Insight ; 2(9)2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28469079

RESUMEN

Initial promising results with immune sera guided early human mAb approaches against Gram-negative sepsis to an LPS neutralization mechanism, but these efforts failed in human clinical trials. Emergence of multidrug resistance has renewed interest in pathogen-specific mAbs. We utilized a pair of antibodies targeting Klebsiella pneumoniae LPS, one that both neutralizes LPS/TLR4 signaling and mediates opsonophagocytic killing (OPK) (54H7) and one that only promotes OPK (KPE33), to better understand the contribution of each mechanism to mAb protection in an acutely lethal pneumonia model. Passive immunization 24 hours prior to infection with KPE33 protected against lethal infection significantly better than 54H7, while delivery of either mAb 1 hour after infection resulted in similar levels of protection. These data suggest that early neutralization of LPS-induced signaling limits protection afforded by these mAbs. LPS neutralization prevented increases in the numbers of γδT cells, a major producer of the antimicrobial cytokine IL-17A, the contribution of which was confirmed using il17a-knockout mice. We conclude that targeting LPS for OPK without LPS signaling neutralization has potential to combat Gram-negative infection by engaging host immune defenses, rather than inhibiting beneficial innate immune pathways.

19.
Nat Commun ; 8(1): 1991, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29222409

RESUMEN

Emerging multidrug-resistant bacteria are a challenge for modern medicine, but how these pathogens are so successful is not fully understood. Robust antibacterial vaccines have prevented and reduced resistance suggesting a pivotal role for immunity in deterring antibiotic resistance. Here, we show the increased prevalence of Klebsiella pneumoniae lipopolysaccharide O2 serotype strains in all major drug resistance groups correlating with a paucity of anti-O2 antibodies in human B cell repertoires. We identify human monoclonal antibodies to O-antigens that are highly protective in mouse models of infection, even against heavily encapsulated strains. These antibodies, including a rare anti-O2 specific antibody, synergistically protect against drug-resistant strains in adjunctive therapy with meropenem, a standard-of-care antibiotic, confirming the importance of immune assistance in antibiotic therapy. These findings support an antibody-based immunotherapeutic strategy even for highly resistant K. pneumoniae infections, and underscore the effect humoral immunity has on evolving drug resistance.


Asunto(s)
Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Infecciones por Klebsiella/terapia , Klebsiella pneumoniae/fisiología , Antígenos O/inmunología , Animales , Antibacterianos/uso terapéutico , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Línea Celular , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple/genética , Farmacorresistencia Bacteriana Múltiple/inmunología , Humanos , Inmunidad Humoral , Factores Inmunológicos/uso terapéutico , Inmunoterapia/métodos , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/mortalidad , Klebsiella pneumoniae/efectos de los fármacos , Meropenem , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Serogrupo , Tasa de Supervivencia , Tienamicinas/uso terapéutico
20.
Sci Rep ; 6: 33346, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27624143

RESUMEN

Severe bacterial infection results in both uncontrolled inflammation and immune suppression in septic patients. Although there is ample evidence that complement activation provokes overwhelming pro-inflammatory responses, whether or not it plays a role in immune suppression in this case is unclear. Here, we identify that complement C5a directly participates in negative regulation of immune responses to bacteria-induced inflammation in an ex vivo model of human whole blood. Challenge of whole blood with heat-killed Pseudomonas aeruginosa induces PD-L1 expression on monocytes and the production of IL-10 and TGF-ß, which we show to be inhibited by C5a blockade. The induction of PD-L1 expression by C5a is via C5aR1but not C5aR2. Furthermore, C5a synergises with P. aeruginosa LPS in both PD-L1 expression and the production of IL-10 and TGF-ß. Mechanistically, C5a contributes to the synergy in PD-L1 expression by specifically activating Erk1/2 and JNK signaling pathways. Our study reveals a new role for C5a in directly promoting immunosuppressive responses. Therefore, aberrant production of complement C5a during bacterial infection could have broader effect on compromising host defense including the induction of immune suppression.


Asunto(s)
Antígeno B7-H1/metabolismo , Complemento C5a/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Humanos , Interleucina-10/metabolismo , Pseudomonas aeruginosa/fisiología , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA