Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Infect Dis ; 217(7): 1110-1117, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29300993

RESUMEN

Background: Cryptosporidiosis affects all human populations, but can be much more severe or life-threatening in children and individuals with weak or weakened immune systems. However, current options to treat cryptosporidiosis are limited. Methods: An in vitro phenotypic screening assay was employed to screen 1200 existing drugs for their anticryptosporidial activity and to determine the inhibitory kinetics of top hits. Selected top hits were further evaluated in mice. The action of the lead compound vorinostat on the parasite histone deacetylase (HDAC) was biochemically validated. Results: Fifteen compounds exhibited anticryptosporidial activity at nanomolar level in vitro. Among them, the histone deacetylase (HDAC) inhibitor vorinostat retained outstanding efficacy in vitro (half maximal effective concentration, EC50 = 203 nM) and in an interleukin 12 knockout mouse model (50% inhibition dose = 7.5 mg/kg). Vorinostat was effective on various parasite developmental stages and could irreversibly kill the parasite. Vorinostat was highly effective against the parasite native HDAC enzymes (half maximal inhibitory concentration, IC50 = 90.0 nM) and a recombinant Cryptosporidium parvum HDAC (the inhibitor constant, Ki = 123.0 nM). Conclusions: These findings suggest the potential for repurposing of vorinostat to treat cryptosporidiosis, and imply that the parasite HDAC can be explored for developing more selective anticryptosporidial therapeutics.


Asunto(s)
Antiprotozoarios/farmacología , Criptosporidiosis/tratamiento farmacológico , Cryptosporidium parvum/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Vorinostat/farmacología , Animales , Antiprotozoarios/uso terapéutico , Descubrimiento de Drogas , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Interleucina-12/genética , Ratones , Ratones Noqueados , Vorinostat/uso terapéutico
2.
J Eukaryot Microbiol ; 62(1): 44-50, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25155632

RESUMEN

IL-18 is known to play a key role limiting Cryptosporidium parvum infection. In this study, we show that IL-18 depletion in SCID mice significantly exacerbates C. parvum infection, whereas, treatment with recombinant IL-18 (rIL-18), significantly decreases the parasite load, as compared to controls. Increases in serum IFN-γ levels as well as the up-regulation of the antimicrobial peptides, cathelicidin antimicrobial peptide and beta defensin 3 (Defb3) were observed in the intestinal mucosa of mice treated with rIL-18. In addition, C. parvum infection significantly increased mRNA expression levels (> 50 fold) of the alpha defensins, Defa3 and 5, respectively. Interestingly, we also found a decrease in mRNA expression of IL-33 (a recently identified cytokine in the same family as IL-18) in the small intestinal tissue from mice treated with rIL-18. In comparison, the respective genes were induced by IL-18 depletion. Our findings suggest that IL-18 can mediate its protective effects via different routes such as IFN-γ induction or by directly stimulating intestinal epithelial cells to increase antimicrobial activity.


Asunto(s)
Criptosporidiosis/tratamiento farmacológico , Inmunidad Innata/efectos de los fármacos , Inmunidad Mucosa/efectos de los fármacos , Interleucina-18/farmacología , Mucosa Intestinal/efectos de los fármacos , ARN Mensajero/inmunología , Animales , Péptidos Catiónicos Antimicrobianos , Catelicidinas/agonistas , Catelicidinas/genética , Catelicidinas/inmunología , Criptosporidiosis/inmunología , Criptosporidiosis/parasitología , Cryptosporidium parvum/inmunología , Femenino , Regulación de la Expresión Génica , Interferón gamma/agonistas , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-33/antagonistas & inhibidores , Interleucina-33/genética , Interleucina-33/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Ratones , Ratones SCID , Carga de Parásitos , ARN Mensajero/agonistas , ARN Mensajero/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Transducción de Señal , alfa-Defensinas/agonistas , alfa-Defensinas/genética , alfa-Defensinas/inmunología , beta-Defensinas/agonistas , beta-Defensinas/genética , beta-Defensinas/inmunología
3.
J Infect Dis ; 209(8): 1279-87, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24273180

RESUMEN

BACKGROUND: Cryptosporidium is emerging as 1 of the 4 leading diarrheal pathogens in children in developing countries. Its infections in patients with AIDS can be fatal, whereas fully effective treatments are unavailable. The major goal of this study is to explore parasite fatty acyl-coenzyme A synthetase (ACS) as a novel drug target. METHODS: A colorimetric assay was developed to evaluate biochemical features and inhibitory kinetics of Cryptosporidium parvum ACSs using recombinant proteins. Anticryptosporidial efficacies of the ACS inhibitor triacsin C were evaluated both in vitro and in vivo. RESULTS: Cryptosporidium ACSs displayed substrate preference toward long-chain fatty acids. The activity of parasite ACSs could be specifically inhibited by triacsin C with the inhibition constant Ki in the nanomolar range. Triacsin C was highly effective against C. parvum growth in vitro (median inhibitory concentration, 136 nmol/L). Most importantly, triacsin C effectively reduced parasite oocyst production up to 88.1% with no apparent toxicity when administered to Cryptosporidium-infected interleukin 12 knockout mice at 8-15 mg/kg/d for 1 week. CONCLUSIONS: The findings of this study not only validated Cryptosporidium ACS (and related acyl-[acyl-carrier-protein]-ligases) as pharmacological targets but also indicate that triacsin C and analogues can be explored as potential new therapeutics against the virtually untreatable cryptosporidial infection in immunocompromised patients.


Asunto(s)
Coenzima A Ligasas/antagonistas & inhibidores , Criptosporidiosis/prevención & control , Cryptosporidium parvum/enzimología , Inhibidores Enzimáticos/farmacología , Triazenos/farmacología , Animales , Técnicas de Cultivo de Célula , Clonación de Organismos , Coenzima A Ligasas/metabolismo , Criptosporidiosis/enzimología , Humanos , Ratones
4.
Antimicrob Agents Chemother ; 58(3): 1603-14, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24366728

RESUMEN

Cryptosporidium parasites are a major cause of diarrhea and malnutrition in the developing world, a frequent cause of waterborne disease in the developed world, and a potential bioterrorism agent. Currently, available treatment is limited, and Cryptosporidium drug discovery remains largely unsuccessful. As a result, the pharmacokinetic properties required for in vivo efficacy have not been established. We have been engaged in a Cryptosporidium drug discovery program targeting IMP dehydrogenase (CpIMPDH). Here, we report the activity of eight potent and selective inhibitors of CpIMPDH in the interleukin-12 (IL-12) knockout mouse model, which mimics acute human cryptosporidiosis. Two compounds displayed significant antiparasitic activity, validating CpIMPDH as a drug target. The best compound, P131 (250 mg/kg of body weight/day), performed equivalently to paromomycin (2,000 mg/kg/day) when administered in a single dose and better than paromomycin when administered in three daily doses. One compound, A110, appeared to promote Cryptosporidium infection. The pharmacokinetic, uptake, and permeability properties of the eight compounds were measured. P131 had the lowest systemic distribution but accumulated to high concentrations within intestinal cells. A110 had the highest systemic distribution. These observations suggest that systemic distribution is not required, and may be a liability, for in vivo antiparasitic activity. Intriguingly, A110 caused specific alterations in fecal microbiota that were not observed with P131 or vehicle alone. Such changes may explain how A110 promotes parasitemia. Collectively, these observations suggest a blueprint for the development of anticryptosporidial therapy.


Asunto(s)
Coccidiostáticos/uso terapéutico , Criptosporidiosis/tratamiento farmacológico , Cryptosporidium parvum/efectos de los fármacos , IMP Deshidrogenasa/antagonistas & inhibidores , Animales , Células CACO-2/parasitología , Modelos Animales de Enfermedad , Descubrimiento de Drogas/métodos , Humanos , Interleucina-12/genética , Ratones , Ratones Endogámicos C57BL/parasitología , Ratones Noqueados/parasitología
5.
Microorganisms ; 8(6)2020 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-32532051

RESUMEN

Cryptosporidium spp. are opportunistic protozoan parasites that infect epithelial cells of the small intestine, causing diarrheal illness in humans. Differences in severity may be due to the immunological status of the host, malnutrition or prior exposure but may also be due to differences in the host gut flora. We examined changes in bacterial flora following antibiotic treatment to determine how cryptosporidial infections and gut integrity were affected by alterations in the microbiome. DNA was extracted from fecal and intestinal samples during peak infection. V4 region amplicons were generated and sequenced using 16sRNA on an Illumina MiSeq. Species evenness and richness were estimated using the Shannon diversity index. There was a significant decrease in anaerobes and overgrowth of Enterobacteriaceae in mice treated with cloxacillin. We also examined levels of short-chain fatty acids in fecal samples. There was a significant decrease in acetate, propionate, and butyrate in these same mice. Concurrent with the shift in bacterial infection was a significant increase in severity of cryptosporidial infection and increase in gut permeability. Treatment with other antibiotics significantly altered the microbiome but did not change the infection, suggesting that specific alterations in the host microbiome allow for more favorable growth of the parasite.

6.
Microbes Infect ; 20(6): 369-375, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29842985

RESUMEN

Cryptosporidium spp. are opportunistic protozoan parasites that infect epithelial cells in the intestinal tract and cause a flu-like diarrheal illness. Innate immunity is key to limiting the expansion of parasitic stages early in infection. One mechanism in which it does this is through the generation of early cytokines, such as IL-18. The processing and secretion of mature IL-18 (and IL-1ß) is mediated by caspase-1 which is activated within an inflammasome following the engagement of inflammasome-initiating sensors. We examined how the absence of caspase-1 and caspase-11, the adapter protein Asc, and other inflammasome components affects susceptibility to cryptosporidial infection by these and other key cytokines in the gut. We found that Casp-11-/-Casp-1-/- knockout mice have increased susceptibility to Cryptosporidium parvum infection as demonstrated by the 35-fold higher oocyst production (at peak infection) compared to wild-type mice. Susceptibility correlated with a lack of IL-18 in caspase-1 and caspase1/11 knockout mice, whereas IL-18 is significantly elevated in wildtype mice. IL-1ß was not generated in any significant amount following infection nor was any increased susceptibility observed in IL-1ß knockout mice. We also show that the adapter protein Asc is important to susceptibility, and that the caspase-1 canonical inflammasome signaling pathway is the dominant pathway in C. parvum resistance.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , Criptosporidiosis/genética , Criptosporidiosis/metabolismo , Cryptosporidium parvum/metabolismo , Inflamasomas/metabolismo , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Caspasa 1/deficiencia , Caspasas/deficiencia , Caspasas/metabolismo , Caspasas Iniciadoras , Cryptosporidium parvum/crecimiento & desarrollo , Predisposición Genética a la Enfermedad , Interacciones Huésped-Parásitos , Interleucina-18/metabolismo , Ratones , Ratones Noqueados , Carga de Parásitos , Transducción de Señal
7.
FEMS Microbiol Lett ; 363(14)2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27279627

RESUMEN

Auranofin is an FDA-approved gold-containing compound used for the treatment of rheumatoid arthritis. Recent reports of antimicrobial activity against protozoa and bacteria indicate that auranofin targets the reductive enzyme thioredoxin reductase (TrxR). We evaluated auranofin as well as five auranofin analogs containing N-heterocyclic carbenes (instead of the triethylphosphane present in auranofin) and five gold-carbene controls for their ability to inhibit or kill Helicobacter pylori in vitro Auranofin completely inhibited bacterial growth at 1.2 µM. Purified H. pylori TrxR was inhibited by auranofin in a cell-free assay (IC50 ∼88 nM). The most active gold(I)-N-heterocyclic carbene compounds exhibited MICs comparable to auranofin against H. pylori (2 µM), while also exhibiting lower toxicities for human embryonic kidney cells (HEK-293T cells). Median toxic concentrations (TC50) were 13-20-fold higher compared to auranofin indicating that they were less cytotoxic. The N-heterocyclic carbene analogs maybe well tolerated, but further evaluation is needed in vivo Finally, auranofin was synergistic with the antibiotic amoxicillin, suggesting that targeting both the reductive enzyme TrxR and cell wall synthesis may be effective against H. pylori infections.


Asunto(s)
Antibacterianos/farmacología , Auranofina/farmacología , Helicobacter pylori/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Metano/análogos & derivados , Compuestos Orgánicos de Oro/farmacología , Antibacterianos/química , Auranofina/química , Línea Celular Transformada , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Compuestos Heterocíclicos/química , Humanos , Concentración 50 Inhibidora , Metano/química , Metano/farmacología , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Compuestos Orgánicos de Oro/química
8.
Immunol Lett ; 158(1-2): 42-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24295591

RESUMEN

Our previous studies have described dendritic cells (DCs) to be important sources of Th1 cytokines such as IL-12 and IL-2 in vitro, following stimulation with Cryptosporidium parvum antigens. We further established the role of DCs during cryptosporidiosis using a diphtheria toxin promoter regulated transgenic CD11c-DTR/EGFP mouse model. In vivo depletion of CD11c(+) cells in CD11c-DTR-Tg mice significantly increased susceptibility to C. parvum infection. Adoptive transfer of unstimulated or antigen stimulated DCs into CD11c(+) depleted CD11c-DTR-Tg mice resulted in an early decrease in parasite load at 4 days post infection. However, this response was transient since parasite load increased in mice engrafted with either unstimulated DCs or DCs stimulated with solubilized antigen by 6 days post infection. In contrast, in mice engrafted with DCs stimulated with live sporozoites, parasite load remained low during the entire period, suggesting the development of a more effective and sustained response. A corresponding increase in IFN-γ expression in T cells from spleen and mesenteric lymph nodes was also noted. Consistent with the in vivo engraftment study, DCs that are pulsed with live sporozoites in vitro and co-cultured with CD4(+) and CD8(+) T cells produced higher IFN-γ levels. Our study establishes the importance of DCs in susceptibility to infection by C. parvum and as important mediators of immune responses.


Asunto(s)
Criptosporidiosis/inmunología , Cryptosporidium parvum/inmunología , Células Dendríticas/inmunología , Células TH1/inmunología , Traslado Adoptivo , Animales , Antígenos CD11/metabolismo , Citocinas/metabolismo , Células Dendríticas/trasplante , Toxoide Diftérico/inmunología , Susceptibilidad a Enfermedades , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Microbes Infect ; 15(8-9): 599-606, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23644177

RESUMEN

Cryptosporidium parvum is a protozoan parasite that infects the epithelial cells of the small intestine causing diarrheal illness in humans. While T cells are known to be important in resistance and recovery from infection, little has been characterized as to the phenotypic expression of surface effector and memory markers after infection. We used an acute model of infection (C57BL/6 interleukin-12p40), which develops long-standing resistance to re-infection, to characterize expression of different effector and memory cells. Using flow cytometry, we found that heterogeneous populations were generated after infection, consisting of both CD62L(high) central memory T cells (T(CM)) and CD62L(low) effector memory T cells (T(EM)) that were competent to produce the Th type 1 effector cytokine, IFN-γ. Both CD4⁺ and CD8⁺ T(CM) and T(EM) populations persisted in the absence of infection (up to 60 days post-infection). Additionally, transfer of either CD62L(low)CD4⁺ T(EM) or CD62L(high)CD4⁺ T(CM) into naive recipients resulted in a protective response. Taken together, these studies show that distinct subsets of effector and memory CD4⁺ T cells develop after infection with C. parvum, and mediate protective immunity to re-challenge.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Criptosporidiosis/prevención & control , Cryptosporidium parvum/inmunología , Memoria Inmunológica , Traslado Adoptivo , Animales , Antígenos CD/análisis , Criptosporidiosis/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA