Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(6): e1011425, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37289815

RESUMEN

Cryptosporidium hominis is a serious cause of childhood diarrhea in developing countries. The development of therapeutics is impeded by major technical roadblocks including lack of cryopreservation and simple culturing methods. This impacts the availability of optimized/standardized singular sources of infectious parasite oocysts for research and human challenge studies. The human C. hominis TU502 isolate is currently propagated in gnotobiotic piglets in only one laboratory, which limits access to oocysts. Streamlined cryopreservation could enable creation of a biobank to serve as an oocyst source for research and distribution to other investigators requiring C. hominis. Here, we report cryopreservation of C. hominis TU502 oocysts by vitrification using specially designed specimen containers scaled to 100 µL volume. Thawed oocysts exhibit ~70% viability with robust excystation and 100% infection rate in gnotobiotic piglets. The availability of optimized/standardized sources of oocysts may streamline drug and vaccine evaluation by enabling wider access to biological specimens.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Animales , Humanos , Porcinos , Criptosporidiosis/parasitología , Vitrificación , Oocistos , Criopreservación
2.
PLoS Pathog ; 18(9): e1010713, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36107831

RESUMEN

Enteric microbial pathogens, including Escherichia coli, Shigella and Cryptosporidium species, take a particularly heavy toll in low-income countries and are highly associated with infant mortality. We describe here a means to display anti-infective agents on the surface of a probiotic bacterium. Because of their stability and versatility, VHHs, the variable domains of camelid heavy-chain-only antibodies, have potential as components of novel agents to treat or prevent enteric infectious disease. We isolated and characterized VHHs targeting several enteropathogenic E. coli (EPEC) virulence factors: flagellin (Fla), which is required for bacterial motility and promotes colonization; both intimin and the translocated intimin receptor (Tir), which together play key roles in attachment to enterocytes; and E. coli secreted protein A (EspA), an essential component of the type III secretion system (T3SS) that is required for virulence. Several VHHs that recognize Fla, intimin, or Tir blocked function in vitro. The probiotic strain E. coli Nissle 1917 (EcN) produces on the bacterial surface curli fibers, which are the major proteinaceous component of E. coli biofilms. A subset of Fla-, intimin-, or Tir-binding VHHs, as well as VHHs that recognize either a T3SS of another important bacterial pathogen (Shigella flexneri), a soluble bacterial toxin (Shiga toxin or Clostridioides difficile toxin TcdA), or a major surface antigen of an important eukaryotic pathogen (Cryptosporidium parvum) were fused to CsgA, the major curli fiber subunit. Scanning electron micrographs indicated CsgA-VHH fusions were assembled into curli fibers on the EcN surface, and Congo Red binding indicated that these recombinant curli fibers were produced at high levels. Ectopic production of these VHHs conferred on EcN the cognate binding activity and, in the case of anti-Shiga toxin, was neutralizing. Taken together, these results demonstrate the potential of the curli-based pathogen sequestration strategy described herein and contribute to the development of novel VHH-based gut therapeutics.


Asunto(s)
Toxinas Bacterianas , Criptosporidiosis , Cryptosporidium , Escherichia coli Enteropatógena , Probióticos , Anticuerpos de Dominio Único , Humanos , Antígenos de Superficie , Rojo Congo , Flagelina , Sistemas de Secreción Tipo III , Factores de Virulencia/genética
3.
Antimicrob Agents Chemother ; 67(4): e0142522, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-36920244

RESUMEN

Recent advances on the development of bumped kinase inhibitors for treatment of cryptosporidiosis have focused on the 5-aminopyrazole-4-carboxamide scaffold, due to analogs that have less hERG inhibition, superior efficacy, and strong in vitro safety profiles. Three compounds, BKI-1770, -1841, and -1708, showed strong efficacy in C. parvum infected mice. Both BKI-1770 and BKI-1841 had efficacy in the C. parvum newborn calf model, reducing diarrhea and oocyst excretion. However, both compounds caused hyperflexion of the limbs seen as dropped pasterns. Toxicity experiments in rats and calves dosed with BKI-1770 showed enlargement of the epiphyseal growth plate at doses only slightly higher than the efficacious dose. Mice were used as a screen to check for bone toxicity, by changes to the tibia epiphyseal growth plate, or neurological causes, by use of a locomotor activity box. These results showed neurological effects from both BKI-1770 and BKI-1841 and bone toxicity in mice from BKI-1770, indicating one or both effects may be contributing to toxicity. However, BKI-1708 remains a viable treatment candidate for further evaluation as it showed no signs of bone toxicity or neurological effects in mice.


Asunto(s)
Antineoplásicos , Antiprotozoarios , Criptosporidiosis , Cryptosporidium parvum , Animales , Bovinos , Ratones , Ratas , Criptosporidiosis/tratamiento farmacológico , Antiprotozoarios/farmacología , Antineoplásicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Oocistos
4.
Infect Immun ; 90(7): e0012722, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35735982

RESUMEN

Cryptosporidiosis was shown a decade ago to be a major contributor to morbidity and mortality of diarrheal disease in children in low-income countries. A serious obstacle to develop and evaluate immunogens and vaccines to control this disease is the lack of well-characterized immunocompetent rodent models. Here, we optimized and compared two mouse models for the evaluation of vaccines: the Cryptosporidium tyzzeri model, which is convenient for screening large numbers of potential mixtures of immunogens, and the Cryptosporidium parvum-infected mouse pretreated with interferon gamma-neutralizing monoclonal antibody.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Animales , Criptosporidiosis/prevención & control , Diarrea , Modelos Animales de Enfermedad , Ratones
5.
Vet Pathol ; 59(4): 648-660, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35521761

RESUMEN

There is a need to standardize pathologic endpoints in animal models of SARS-CoV-2 infection to help benchmark study quality, improve cross-institutional comparison of data, and assess therapeutic efficacy so that potential drugs and vaccines for SARS-CoV-2 can rapidly advance. The Syrian hamster model is a tractable small animal model for COVID-19 that models clinical disease in humans. Using the hamster model, the authors used traditional pathologic assessment with quantitative image analysis to assess disease outcomes in hamsters administered polyclonal immune sera from previously challenged rhesus macaques. The authors then used quantitative image analysis to assess pathologic endpoints across studies performed at different institutions using different tissue processing protocols. The authors detail pathological features of SARS-CoV-2 infection longitudinally and use immunohistochemistry to quantify myeloid cells and T lymphocyte infiltrates during SARS-CoV-2 infection. High-dose immune sera protected hamsters from weight loss and diminished viral replication in tissues and reduced lung lesions. Cumulative pathology scoring correlated with weight loss and was robust in distinguishing IgG efficacy. In formalin-infused lungs, quantitative measurement of percent area affected also correlated with weight loss but was less robust in non-formalin-infused lungs. Longitudinal immunohistochemical assessment of interstitial macrophage infiltrates showed that peak infiltration corresponded to weight loss, yet quantitative assessment of macrophage, neutrophil, and CD3+ T lymphocyte numbers did not distinguish IgG treatment effects. Here, the authors show that quantitative image analysis was a useful adjunct tool for assessing SARS-CoV-2 treatment outcomes in the hamster model.


Asunto(s)
COVID-19 , Enfermedades de los Roedores , Animales , COVID-19/veterinaria , Vacunas contra la COVID-19 , Cricetinae , Modelos Animales de Enfermedad , Humanos , Sueros Inmunes , Inmunoglobulina G , Pulmón/patología , Macaca mulatta , Mesocricetus , Enfermedades de los Roedores/patología , SARS-CoV-2 , Pérdida de Peso
6.
J Infect Dis ; 221(2): 276-284, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31495879

RESUMEN

Nosocomial infections with Clostridium difficile are on the rise in the Unites States, attributed to emergence of antibiotic-resistant and hypervirulent strains associated with greater likelihood of recurrent infections. In addition to antibiotics, treatment with Merck anti-toxin B (TcdB) antibody bezlotoxumab is reported to reduce recurrent infections. However, treatment with anti-toxin A (TcdA) antibody actotoxumab was associated with dramatically increased disease severity and mortality rates in humans and gnotobiotic piglets. Using isogenic mutants of C. difficile strain NAPI/BI/027 deficient in TcdA (A-B+) or TcdB (A+B-), and the wild type, we investigated how and why treatment of infected animals with anti-TcdA dramatically increased disease severity. Contrary to the hypothesis, among piglets treated with anti-TcdA, those with A+B- infection were disease free, in contrast to the disease enhancement seen in those with wild-type or A-B+ infection. It seems that the lack of TcdA, through either deletion or neutralization with anti-TcdA, reduces a competitive pressure, allowing TcdB to freely exert its profound effect, leading to increased mucosal injury and disease severity.


Asunto(s)
Antibacterianos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos ampliamente neutralizantes/administración & dosificación , Infecciones por Clostridium/tratamiento farmacológico , Animales , Antibacterianos/farmacología , Clostridioides difficile/efectos de los fármacos , Colon Descendente/patología , Vida Libre de Gérmenes/efectos de los fármacos , Humanos , Porcinos
7.
J Infect Dis ; 220(2): 285-293, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-30893435

RESUMEN

BACKGROUND: Cryptosporidiosis, an enteric protozoon, causes substantial morbidity and mortality associated with diarrhea in children <2 years old in low- to middle-income countries. There is no vaccine and treatments are inadequate. A piperazine-based compound, MMV665917, has in vitro and in vivo efficacy against Cryptosporidium parvum. In this study, we evaluated the efficacy of MMV665917 in gnotobiotic piglets experimentally infected with Cryptosporidium hominis, the species responsible for >75% of diarrhea reported in these children. METHODS: Gnotobiotic piglets were orally challenged with C hominis oocysts, and oral treatment with MMV665917 was commenced 3 days after challenge. Oocyst excretion and diarrhea severity were observed daily, and mucosal colonization and lesions were recorded after necropsy. RESULTS: MMV665917 significantly reduced fecal oocyst excretion, parasite colonization and damage to the intestinal mucosa, and peak diarrheal symptoms, compared with infected untreated controls. A dose of 20 mg/kg twice daily for 7 days was more effective than 10 mg/kg. There were no signs of organ toxicity at either dose, but 20 mg/kg was associated with slightly elevated blood cholesterol and monocytes at euthanasia. CONCLUSIONS: These results demonstrate the effectiveness of this drug against C hominis. Piperazine-derivative MMV665917 may potentially be used to treat human cryptosporidiosis; however, further investigations are required.


Asunto(s)
Criptosporidiosis/tratamiento farmacológico , Cryptosporidium parvum/efectos de los fármacos , Diarrea/tratamiento farmacológico , Piperazinas/farmacología , Animales , Criptosporidiosis/parasitología , Diarrea/parasitología , Modelos Animales de Enfermedad , Mucosa Intestinal/parasitología , Monocitos/parasitología , Oocistos/efectos de los fármacos , Porcinos
8.
J Biol Chem ; 292(40): 16677-16687, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28842484

RESUMEN

Numerous Gram-negative pathogens infect eukaryotes and use the type III secretion system (T3SS) to deliver effector proteins into host cells. One important T3SS feature is an extracellular needle with an associated tip complex responsible for assembly of a pore-forming translocon in the host cell membrane. Shigella spp. cause shigellosis, also called bacillary dysentery, and invade colonic epithelial cells via the T3SS. The tip complex of Shigella flexneri contains invasion plasmid antigen D (IpaD), which initially regulates secretion and provides a physical platform for the translocon pore. The tip complex represents a promising therapeutic target for many important T3SS-containing pathogens. Here, in an effort to further elucidate its function, we created a panel of single-VH domain antibodies (VHHs) that recognize distinct epitopes within IpaD. These VHHs recognized the in situ tip complex and modulated the infectious properties of Shigella Moreover, structural elucidation of several IpaD-VHH complexes provided critical insights into tip complex formation and function. Of note, one VHH heterodimer could reduce Shigella hemolytic activity by >80%. Our observations along with previous findings support the hypothesis that the hydrophobic translocator (IpaB in Shigella) likely binds to a region within the tip protein that is structurally conserved across all T3SS-possessing pathogens, suggesting potential therapeutic avenues for managing infections by these pathogens.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Sistemas de Secreción Bacterianos/inmunología , Epítopos/inmunología , Shigella flexneri/inmunología , Anticuerpos de Cadena Única/inmunología , Animales , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Sistemas de Secreción Bacterianos/genética , Camélidos del Nuevo Mundo , Evolución Molecular Dirigida , Epítopos/genética , Shigella flexneri/genética
9.
Infect Immun ; 86(11)2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150259

RESUMEN

The symptoms of Clostridium difficile infection (CDI) are attributed largely to two C. difficile toxins, TcdA and TcdB. Significant efforts have been devoted to developing vaccines targeting both toxins through parenteral immunization routes. However, C. difficile is an enteric pathogen, and mucosal/oral immunization would be particularly useful to protect the host against CDI, considering that the gut is the main site of disease onset and progression. Moreover, vaccines directed only against toxins do not target the cells and spores that transmit the disease. Previously, we constructed a chimeric vaccine candidate, mTcd138, comprised of the glucosyltransferase and cysteine proteinase domains of TcdB and the receptor binding domain of TcdA. In this study, to develop an oral vaccine that can target both C. difficile toxins and colonization/adhesion factors, we expressed mTcd138 in a nontoxigenic C. difficile (NTCD) strain, resulting in strain NTCD_mTcd138. Oral immunization with spores of NTCD_mTcd138 provided mice full protection against infection with a hypervirulent C. difficile strain, UK6 (ribotype 027). The protective strength and efficacy of NTCD_mTcd138 were further evaluated in the acute CDI hamster model. Oral immunization with spores of NTCD_mTcd138 also provided hamsters significant protection against infection with 2 × 104 UK6 spores, a dose 200-fold higher than the lethal dose of UK6 in hamsters. These results imply that the genetically modified, nontoxigenic C. difficile strain expressing mTcd138 may represent a novel mucosal vaccine candidate against CDI.


Asunto(s)
Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Clostridioides difficile/inmunología , Infecciones por Clostridium/prevención & control , Enterotoxinas/inmunología , Administración Oral , Animales , Proteínas Bacterianas/genética , Toxinas Bacterianas/genética , Vacunas Bacterianas/genética , Clostridioides difficile/genética , Infecciones por Clostridium/inmunología , Cricetinae , Modelos Animales de Enfermedad , Enterotoxinas/genética , Ratones , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
10.
Artículo en Inglés | MEDLINE | ID: mdl-29661877

RESUMEN

Recent reports highlighting the global significance of cryptosporidiosis among children have renewed efforts to develop control measures. We evaluated the efficacy of bumped kinase inhibitor (BKI) 1369 in the gnotobiotic piglet model of acute diarrhea caused by Cryptosporidium hominis, the species responsible for most human cases. Five-day treatment with BKI 1369 reduced signs of disease early during treatment compared to those of untreated animals. Piglets treated with BKI 1369 exhibited significant reductions of oocyst excretion, mucosal colonization by C. hominis, and mucosal lesions, which resulted in considerable symptomatic improvement. BKI 1369 reduced the parasite burden and disease severity in the gnotobiotic pig model. Together these data suggest that a BKI-mediated therapeutic may be an effective treatment against cryptosporidiosis.


Asunto(s)
Antiprotozoarios/uso terapéutico , Criptosporidiosis/tratamiento farmacológico , Cryptosporidium/efectos de los fármacos , Diarrea/tratamiento farmacológico , Piperidinas/uso terapéutico , Pirimidinas/uso terapéutico , Quinolinas/uso terapéutico , Enfermedad Aguda , Animales , Animales Recién Nacidos , Criptosporidiosis/parasitología , Diarrea/parasitología , Modelos Animales de Enfermedad , Vida Libre de Gérmenes , Oocistos/metabolismo , Carga de Parásitos , Porcinos
11.
Anaerobe ; 41: 58-67, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27262694

RESUMEN

The gut microbiome, composed of the microflora that inhabit the gastrointestinal tract and their genomes, make up a complex ecosystem that can be disrupted by antibiotic use. The ensuing dysbiosis is conducive to the emergence of opportunistic pathogens such as Clostridium difficile. A novel approach to protect the microbiome from antibiotic-mediated dysbiosis is the use of beta-lactamase enzymes to degrade residual antibiotics in the gastrointestinal tract before the microflora are harmed. Here we present the preclinical development and early clinical studies of the beta-lactamase enzymes, P3A, currently referred to as SYN-004, and its precursor, P1A. Both P1A and SYN-004 were designed as orally-delivered, non-systemically available therapeutics for use with intravenous beta-lactam antibiotics. SYN-004 was engineered from P1A, a beta-lactamase isolated from Bacillus licheniformis, to broaden its antibiotic degradation profile. SYN-004 efficiently hydrolyses penicillins and cephalosporins, the most widely used IV beta-lactam antibiotics. In animal studies, SYN-004 degraded ceftriaxone in the GI tract of dogs and protected the microbiome of pigs from ceftriaxone-induced changes. Phase I clinical studies demonstrated SYN-004 safety and tolerability. Phase 2 studies are in progress to assess the utility of SYN-004 for the prevention of antibiotic-associated diarrhea and Clostridium difficile disease.


Asunto(s)
Antibacterianos/farmacología , Enterocolitis Seudomembranosa/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Proteínas Recombinantes/farmacología , beta-Lactamasas/farmacología , Animales , Antibacterianos/química , Antibacterianos/uso terapéutico , Clostridioides difficile/efectos de los fármacos , Perros , Farmacorresistencia Bacteriana , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Cinética , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapéutico , Sus scrofa , beta-Lactamasas/química , beta-Lactamasas/uso terapéutico
12.
Clin Microbiol Rev ; 27(3): 575-86, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24982322

RESUMEN

The protozoan parasite Cryptosporidium infects all major vertebrate groups and causes significant diarrhea in humans, with a spectrum of diseases ranging from asymptomatic to life-threatening. Children and immunodeficient individuals are disproportionately affected, especially in developing countries, where cryptosporidiosis contributes substantially to morbidity and mortality in preschool-age children. Despite the enormous disease burden from cryptosporidiosis, no antiprotozoal agent or vaccine exists for effective treatment or prevention. Cryptosporidiosis involving the respiratory tract has been described for avian species and mammals, including immunocompromised humans. Recent evidence indicates that respiratory cryptosporidiosis may occur commonly in immunocompetent children with cryptosporidial diarrhea and unexplained cough. Findings from animal models, human case reports, and a few epidemiological studies suggest that Cryptosporidium may be transmitted via respiratory secretions, in addition to the more recognized fecal-oral route. It is postulated that transmission of Cryptosporidium oocysts may occur by inhalation of aerosolized droplets or by contact with fomites contaminated by coughing. Delineating the role of the respiratory tract in disease transmission may provide necessary evidence to establish further guidelines for prevention of cryptosporidiosis.


Asunto(s)
Criptosporidiosis/parasitología , Criptosporidiosis/transmisión , Cryptosporidium/fisiología , Animales , Criptosporidiosis/diagnóstico , Criptosporidiosis/terapia , Cryptosporidium/clasificación , Humanos , Inhalación
13.
J Infect Dis ; 211(8): 1334-41, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25381448

RESUMEN

BACKGROUND: Clostridium difficile is a primary cause of antibiotic-associated diarrhea that typically develops when gut microbiota is altered. Conventional treatment for C. difficile infection (CDI) is additional antimicrobial administration, which further disrupts normal intestinal microbiota, often resulting in poor treatment outcomes. METHODS: A pregnant dairy cow was repeatedly immunized with recombinant mutants of toxins A and B produced by C. difficile, and the resultant hyperimmune bovine colostrum (HBC) was evaluated for therapeutic efficacy in gnotobiotic piglets with diarrhea due to CDI. Control piglets received nonimmune colostrum. To determine the impact of HBC on gut microbiota, 1 of 2 groups of piglets transplanted with normal human gut microbiota was treated with HBC. RESULTS: Nonimmune colostrum-treated piglets developed moderate to severe diarrhea and colitis. In contrast, HBC-treated piglets had mild or no diarrhea and mild or no colitis. Lyophilization had no detectable impact on HBC efficacy. HBC had no discernible effect on the composition of normal human gut microbiota in the porcine intestinal tract. CONCLUSIONS: HBC provides an oral, cost-effective, and safe alternative to antibiotic therapy for CDI. By preserving intestinal microbiota, HBC may be more efficacious than antibiotics. Additional studies are warranted to establish HBC as a viable immunotherapeutic agent for human use against CDI.


Asunto(s)
Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/terapia , Calostro/inmunología , Anciano , Animales , Antibacterianos/inmunología , Bovinos , Colitis/inmunología , Colitis/microbiología , Colitis/terapia , Diarrea/inmunología , Diarrea/microbiología , Femenino , Humanos , Factores Inmunológicos/inmunología , Enfermedades Intestinales/inmunología , Intestinos/inmunología , Intestinos/microbiología , Masculino , Persona de Mediana Edad , Embarazo , Porcinos
14.
Infect Immun ; 83(1): 286-91, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25368111

RESUMEN

Hemolytic-uremic syndrome (HUS), caused by Shiga toxin (Stx)-producing Escherichia coli (STEC), remains untreatable. Production of human monoclonal antibodies against Stx, which are highly effective in preventing Stx sequelae in animal models, is languishing due to cost and logistics. We reported previously that the production and evaluation of a camelid heavy-chain-only VH domain (VHH)-based neutralizing agent (VNA) targeting Stx1 and Stx2 (VNA-Stx) protected mice from Stx1 and Stx2 intoxication. Here we report that a single intramuscular (i.m.) injection of a nonreplicating adenovirus (Ad) vector carrying a secretory transgene of VNA-Stx (Ad/VNA-Stx) protected mice challenged with Stx2 and protected gnotobiotic piglets infected with STEC from fatal systemic intoxication. One i.m. dose of Ad/VNA-Stx prevented fatal central nervous system (CNS) symptoms in 9 of 10 animals when it was given to piglets 24 h after bacterial challenge and in 5 of 9 animals when it was given 48 h after bacterial challenge, just prior to the onset of CNS symptoms. All 6 placebo animals died or were euthanized with severe CNS symptoms. Ad/VNA-Stx treatment had no impact on diarrhea. In conclusion, Ad/VNA-Stx treatment is effective in protecting piglets from fatal Stx2-mediated CNS complications following STEC challenge. With a low production cost and further development, this could presumably be an effective treatment for patients with HUS and/or individuals at high risk of developing HUS due to exposure to STEC.


Asunto(s)
Adenovirus Humanos/genética , Anticuerpos Neutralizantes/uso terapéutico , Infecciones por Escherichia coli/tratamiento farmacológico , Escherichia coli O157/inmunología , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Toxina Shiga I/antagonistas & inhibidores , Toxina Shiga II/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/genética , Modelos Animales de Enfermedad , Portadores de Fármacos/administración & dosificación , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Escherichia coli O157/genética , Femenino , Vectores Genéticos , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/microbiología , Inyecciones Intramusculares , Ratones , Toxina Shiga I/inmunología , Toxina Shiga II/inmunología , Análisis de Supervivencia , Porcinos , Factores de Tiempo
15.
Antimicrob Agents Chemother ; 59(7): 4199-205, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25941230

RESUMEN

The increasing incidence and severity of infection by Clostridium difficile have stimulated attempts to develop new antimicrobial therapies. We report here the relative abilities of two antibiotics (metronidazole and vancomycin) in current use for treating C. difficile infection and of a third antimicrobial, surotomycin, to kill C. difficile cells at various stages of development and to inhibit the production of the toxin proteins that are the major virulence factors. The results indicate that none of the drugs affects the viability of spores at 8× MIC or 80× MIC and that all of the drugs kill exponential-phase cells when provided at 8× MIC. In contrast, none of the drugs killed stationary-phase cells or inhibited toxin production when provided at 8× MIC and neither vancomycin nor metronidazole killed stationary-phase cells when provided at 80× MIC. Surotomycin, on the other hand, did kill stationary-phase cells when provided at 80× MIC but did so without inducing lysis.


Asunto(s)
Antibacterianos/farmacología , Toxinas Bacterianas/biosíntesis , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/metabolismo , Enterotoxinas/biosíntesis , Lipopéptidos/farmacología , Péptidos Cíclicos/farmacología , Toxinas Bacterianas/genética , Pared Celular/efectos de los fármacos , Clostridioides difficile/genética , Enterotoxinas/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Metronidazol/farmacología , Pruebas de Sensibilidad Microbiana , Mutación/genética , Esporas Bacterianas/efectos de los fármacos , Vancomicina/farmacología , Factores de Virulencia/metabolismo
16.
Antimicrob Agents Chemother ; 58(12): 7560-4, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25267665

RESUMEN

The efficacy of oral tigecycline treatment (2 mg/kg of body weight for 7 days) of Clostridium difficile infection (CDI) was evaluated in the gnotobiotic pig model, and its effect on human gut microflora transplanted into the gnotobiotic pig was determined. Tigecycline oral treatment improved survival, clinical signs, and lesion severity and markedly decreased concentrations of Firmicutes but did not promote CDI. Our data showed that oral tigecycline treatment has a potential beneficial effect on the treatment of CDI.


Asunto(s)
Antibacterianos/farmacología , Clostridioides difficile/efectos de los fármacos , Enterocolitis Seudomembranosa/tratamiento farmacológico , Vida Libre de Gérmenes , Microbiota/efectos de los fármacos , Minociclina/análogos & derivados , Administración Oral , Animales , Clostridioides difficile/crecimiento & desarrollo , Esquema de Medicación , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/patología , Fluoroquinolonas/farmacología , Humanos , Interleucina-8/antagonistas & inhibidores , Interleucina-8/biosíntesis , Interleucina-8/metabolismo , Minociclina/farmacología , Pirimidinonas/farmacología , Porcinos , Tigeciclina , Vancomicina/farmacología
17.
J Infect Dis ; 207(2): 323-30, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23125448

RESUMEN

BACKGROUND: A dramatic increase in morbidity and mortality from Clostridium difficile infection (CDI) due to the recent emergence of virulent, antibiotic-resistant strains has led to a search for alternatives to antibiotics, including vaccines and immune-based therapy that target the 2 key toxins-TcdA and TcdB. METHODS: We investigated the efficacy of specific human monoclonal antibodies (HuMab) and alpaca polyclonal antibodies against each toxin separately and in combination in the gnotobiotic piglet model of CDI. Additionally, the HuMab and polyclonal antibodies were exploited to investigate the precise contribution of each toxin to systemic and/or gastrointestinal (GI) tract disease. RESULTS: Our results indicate that TcdB is an important virulence factor associated with GI and systemic pathology. Administration of anti-TcdB antibody alone or with anti-TcdA protected 100% of piglets from development of systemic CDI and minimized GI lesions. Conversely, 100% of piglets administered only anti-TcdA developed severe GI and systemic disease, with 67%-83% fatality, faring worse than placebo-treated control animals. CONCLUSIONS: These results highlight the importance of TcdB in the pathogenesis of CDI and the effectiveness of TcdB-specific antibody in treating CDI. However, the results raise new questions regarding the nature of TcdA interaction with therapeutic antibodies.


Asunto(s)
Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales Humanizados/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/prevención & control , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/prevención & control , Enterotoxinas/inmunología , Animales , Anticuerpos Antibacterianos/administración & dosificación , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Vida Libre de Gérmenes , Humanos , Porcinos
18.
Gut Microbes ; 16(1): 2297897, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38189373

RESUMEN

Cryptosporidiosis is a major cause of severe diarrheal disease in infants from resource poor settings. The majority of infections are caused by the human-specific pathogen C. hominis and absence of in vitro growth platforms has limited our understanding of host-pathogen interactions and development of effective treatments. To address this problem, we developed a stem cell-derived culture system for C. hominis using human enterocytes differentiated under air-liquid interface (ALI) conditions. Human ALI cultures supported robust growth and complete development of C. hominis in vitro including all life cycle stages. Cryptosporidium infection induced a strong interferon response from enterocytes, possibly driven, in part, by an endogenous dsRNA virus in the parasite. Prior infection with Cryptosporidium induced type III IFN secretion and consequently blunted infection with Rotavirus, including live attenuated vaccine strains. The development of hALI provides a platform for further studies on human-specific pathogens, including clinically important coinfections that may alter vaccine efficacy.


Asunto(s)
Criptosporidiosis , Cryptosporidium , Microbioma Gastrointestinal , Rotavirus , Lactante , Humanos , Interferón lambda , Células Epiteliales , Zea mays
19.
Infect Immun ; 81(12): 4592-603, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24082082

RESUMEN

Shiga toxin-producing Escherichia coli (STEC) is a major cause of severe food-borne disease worldwide, and two Shiga toxins, Stx1 and Stx2, are primarily responsible for the serious disease consequence, hemolytic-uremic syndrome (HUS). Here we report identification of a panel of heavy-chain-only antibody (Ab) V(H) (VHH) domains that neutralize Stx1 and/or Stx2 in cell-based assays. VHH heterodimer toxin-neutralizing agents containing two linked Stx1-neutralizing VHHs or two Stx2-neutralizing VHHs were generally much more potent at Stx neutralization than a pool of the two-component monomers tested in cell-based assays and in vivo mouse models. We recently reported that clearance of toxins can be promoted by coadministering a VHH-based toxin-neutralizing agent with an antitag monoclonal antibody (MAb), called the "effector Ab," that indirectly decorates each toxin molecule with four Ab molecules. Decoration occurs because the Ab binds to a common epitopic tag present at two sites on each of the two VHH heterodimer molecules that bind to each toxin molecule. Here we show that coadministration of effector Ab substantially improved the efficacy of Stx toxin-neutralizing agents to prevent death or kidney damage in mice following challenge with Stx1 or Stx2. A single toxin-neutralizing agent consisting of a double-tagged VHH heterotrimer--one Stx1-specific VHH, one Stx2-specific VHH, and one Stx1/Stx2 cross-specific VHH--was effective in preventing all symptoms of intoxication from Stx1 and Stx2 when coadministered with effector Ab. Overall, the availability of simple, defined, recombinant proteins that provide cost-effective protection against HUS opens up new therapeutic approaches to managing disease.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Toxina Shiga I/inmunología , Toxina Shiga II/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Escherichia coli Enteropatógena/inmunología , Escherichia coli Enteropatógena/metabolismo , Femenino , Ratones , Datos de Secuencia Molecular , Toxina Shiga I/metabolismo , Toxina Shiga II/metabolismo , Escherichia coli Shiga-Toxigénica/inmunología , Escherichia coli Shiga-Toxigénica/metabolismo
20.
Antimicrob Agents Chemother ; 57(8): 4039-41, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23689716

RESUMEN

The novel antibiotic MBX-500, dosed at 100, 200, or 400 mg/kg twice daily for 7 days, was evaluated for the treatment of Clostridium difficile infection (CDI) in the gnotobiotic pig model. MBX-500 increased survival at all doses and at high doses improved clinical signs and reduced lesion severity, similar to vancomycin. Our results show that MBX-500 is an effective antibiotic for the treatment of diarrhea associated with CDI and prevents severe systemic disease.


Asunto(s)
Antibacterianos/farmacología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/tratamiento farmacológico , Diarrea/tratamiento farmacológico , Fluoroquinolonas/farmacología , Pirimidinonas/farmacología , Animales , Colon/microbiología , Colon/patología , Diarrea/microbiología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Vida Libre de Gérmenes , Estimación de Kaplan-Meier , Índice de Severidad de la Enfermedad , Porcinos , Resultado del Tratamiento , Vancomicina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA