Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gastroenterology ; 162(1): 150-165, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34536451

RESUMEN

BACKGROUND & AIMS: G protein-coupled receptor (GPR) 120 has been implicated in regulating metabolic syndromes with anti-inflammatory function. However, the role of GPR120 in intestinal inflammation is unknown. Here, we investigated whether and how GPR120 regulates CD4+ T cell function to inhibit colitis development. METHODS: Dextran sodium sulfate (DSS)-induced colitis model, Citrobacter rodentium infection model, and CD4+ T cell adoptive transfer model were used to analyze the role of GPR120 in regulating colitis development. The effect of GPR120 on CD4+ T cell functions was analyzed by RNA sequencing, flow cytometry, and Seahorse metabolic assays. Mice were administered GPR120 agonist for investigating the potential of GPR120 agonist in preventing and treating colitis. RESULTS: Deficiency of GPR120 in CD4+ T cells resulted in more severe colitis in mice upon dextran sodium sulfate insult and enteric infection. Transfer of GPR120-deficient CD4+CD45Rbhi T cells induced more severe colitis in Rag-/- mice with lower intestinal interleukin (IL) 10+CD4+ T cells. Treatment with the GPR120 agonist CpdA promoted CD4+ T cell production of IL10 by up-regulating Blimp1 and enhancing glycolysis, which was regulated by mTOR. GPR120 agonist-treated wild-type, but not IL10-deficient and Blimp1-deficient, T helper 1 cells induced less severe colitis. Furthermore, oral administration of GPR120 agonist protected mice from intestinal inflammation in both prevention and treatment schemes. Gpr120 expression was positively correlated with Il10 expression in the human colonic mucosa, including patients with inflammatory bowel diseases. CONCLUSIONS: Our findings show the role of GPR120 in regulating intestinal CD4+ T cell production of IL10 to inhibit colitis development, which identifies GPR120 as a potential therapeutic target for treating inflammatory bowel diseases.


Asunto(s)
Acetatos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Colitis/prevención & control , Colon/efectos de los fármacos , Interleucina-10/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Tiramina/análogos & derivados , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Estudios de Casos y Controles , Colitis/inmunología , Colitis/metabolismo , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Colon/inmunología , Colon/metabolismo , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Modelos Animales de Enfermedad , Glucólisis/efectos de los fármacos , Interleucina-10/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Tiramina/farmacología
2.
J Immunol ; 205(7): 1944-1952, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32859726

RESUMEN

The role of IL-21, produced mainly by Th17 cells and T follicular helper cells, has been intensively investigated in B cell differentiation and Ab class switch. However, how IL-21 regulates memory IgA+ B cell development and memory IgA responses in the intestines is still not completely understood. In this study, we found the total IgA+ B cells as well as CD38+CD138-IgA+ memory B cells were significantly increased in intestinal lamina propria (LP) of TCRßxδ-/- mice after transfer of microbiota Ag-specific Th17 cells but not Th1 cells. Although IL-21R-/- mice or IL-17R-/- mice showed decreased Ag-specific memory IgA production in the intestines upon infection with Citrobacter rodentium, the percentage of IgA+CD38+CD138- memory B cells in Peyer's patches and LP was decreased only in IL-21R-/- mice, but not in IL-17R-/- mice, after reinfection with C. rodentium compared with wild-type mice. Blockade IL-21 in vivo suppressed intestinal C. rodentium-specific IgA production as well as IgA+CD38+CD138- memory B cells in Peyer's patches and LP. Furthermore, IL-21 significantly induced B cell IgA production in vitro, with the increased expression of genes related with class-switching and memory B cell development, including Aicda, Ski, Bmi1, and Klf2. Consistently, Aicda and Ski expression was decreased in B cells of IL-21R-/- mice after C. rodentium reinfection. In conclusion, our study demonstrated that IL-21 promotes intestinal memory IgA B cell development, possibly through upregulating differentiation-related and class switching-related genes, indicating a potential role of IL-21 in memory IgA+ B cell responses in the intestines.


Asunto(s)
Linfocitos B/inmunología , Citrobacter rodentium/fisiología , Infecciones por Enterobacteriaceae/inmunología , Interleucinas/metabolismo , Mucosa Intestinal/inmunología , Receptores de Interleucina-21/metabolismo , Células Th17/inmunología , Animales , Células Cultivadas , Inmunoglobulina A/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina-21/genética
3.
FASEB J ; 34(11): 15417-15430, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32969062

RESUMEN

Stimulator of interferon genes (STING) has been shown to play a critical role in orchestrating immune responses to various pathogens through sensing cyclic dinucleotides. However, how STING regulates intestinal homeostasis is still not completely understood. In this study, we found that STING-/- mice were more susceptible to enteric infection with Citrobacter rodentium compared to wild-type (WT) mice evidenced by more severe intestinal inflammation and impaired bacterial clearance. STING-/- mice demonstrated lower expression of REG3γ but not ß-defensins and Cramp in IECs. Consistently, STING-/- IECs showed reduced capacity to inhibit bacterial growth. STING agonists, both 10-carboxymethyl-9-acridanone (CMA) and 5,6-dimethylxanthenone-4-acetic acid (DMXAA), promoted REG3γ expression IECs. Furthermore, STING agonists promoted WT but not REG3γ-deficient IEC bacterial killing. Mechanistically, STING agonists activated STAT3 and promoted glycolysis in IECs. Inhibition of STAT3 pathway and glycolysis suppressed STING-induced REG3γ production in IECs, and abrogated STING-mediated IEC killing of C. rodentium. Additionally, treatment with the STING ligand, 2,3-cGAMP, inhibited C. rodentium-induced colitis in vivo. Overall, STING promotes IEC REG3γ expression to inhibit enteric infection and intestinal inflammation, thus, maintaining the intestinal homeostasis.


Asunto(s)
Colitis/tratamiento farmacológico , Infecciones por Enterobacteriaceae/complicaciones , Células Epiteliales/efectos de los fármacos , Inflamación/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Proteínas de la Membrana/fisiología , Proteínas Citotóxicas Formadoras de Poros/farmacología , Animales , Citrobacter rodentium/efectos de los fármacos , Citrobacter rodentium/crecimiento & desarrollo , Colitis/etiología , Colitis/patología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Homeostasis , Inmunidad Innata , Inflamación/etiología , Inflamación/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo
4.
J Immunol ; 202(1): 79-92, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30478092

RESUMEN

The role of retinoid-related orphan receptor γ t (RORγt) in Th17 cell differentiation has been well established; however, how it regulates other T cell lineages is still not clearly understood. In this study, we report that in mice, while promoting Th17 cell differentiation, RORγt inhibited IL-10 production by T cells, thereby preserving the pathogenicity of Th17 cells. Treatment with RORγt-specific inhibitor suppressed Th17 cell signature cytokines, but promoted IL-10 production. RORγt inhibitor-treated Th17 cells induce less severe colitis compared with control Th17 cells. Mechanistically, the RORγt inhibitor induced T cell expression of Blimp-1 (encoded by Prdm1). Prdm1-/- T cells produced significantly fewer IL-10 when treated with RORγt inhibitor compared with wild-type T cells. Furthermore, RORγt inhibitor-treated Prdm1-/- Th17 cells induce more severe colitis compared with RORγt inhibitor-treated wild-type Th17 cells. Collectively, our studies reveal a novel mechanism by which RORγt drives and maintains pathogenic Th17 cell development by inhibiting IL-10 production.


Asunto(s)
Colitis/inmunología , Interleucina-10/metabolismo , Intestinos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Células Th17/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Represión Epigenética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética
5.
J Immunol ; 201(2): 548-559, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29898958

RESUMEN

The lumen-dwelling protozoan Giardia is an important parasitic cause of diarrheal disease worldwide. Infection can persist over extended periods with minimal intestinal inflammation, suggesting that Giardia may attenuate host responses to ensure its survival, although clearance eventually occurs in most cases. IL-10 is an anti-inflammatory regulator critical for intestinal homeostasis and controlling host responses to bacterial exposure, yet its potential role in coordinating antiprotozoal host defense in the intestine is not known. In this study, we found that murine infection with the natural enteric pathogen Giardia muris induced a transient IL-10 response after 2-4 wk at the primary site of infection in the upper small intestine, but parasite colonization and eradication were not affected by the absence of the cytokine in gene-targeted mice. However, IL-10 was critical for controlling infection-associated immunological sequelae in the colon because severe and persistent diarrhea and colitis were observed in IL-10-deficient mice within 1-2 wk postinfection but not in uninfected littermate controls. Inflammation was characterized by epithelial hyperplasia, neutrophil and macrophage expansion, and Th1 induction and could be prevented by blockade of IL-12/IL-23 p40 but not depletion of CD11c+ dendritic cells. Furthermore, the intestinal microbiota underwent characteristic shifts in composition and was required for disease because antibiotics and loss of TLR signaling in MyD88-deficient mice protected against colitis. Together, our data suggest that transient infection by a luminal and seemingly noninflammatory pathogen can trigger sustained colitis in genetically susceptible hosts, which has broader implications for understanding postinfectious syndromes and other chronic intestinal inflammatory conditions.


Asunto(s)
Colitis/inmunología , Giardia/fisiología , Giardiasis/inmunología , Interleucina-10/metabolismo , Mucosa Intestinal/inmunología , Intestino Delgado/fisiología , Células TH1/inmunología , Animales , Células Cultivadas , Enfermedad Crónica , Predisposición Genética a la Enfermedad , Humanos , Interleucina-10/genética , Interleucina-12/metabolismo , Mucosa Intestinal/parasitología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores Toll-Like/metabolismo
6.
J Immunol ; 201(8): 2492-2501, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30171165

RESUMEN

Neutrophils are the first responders to sites of inflammation when the intestinal epithelial barrier is breached and the gut microbiota invade. Despite current efforts in understanding the role of neutrophils in intestinal homeostasis, the complex interactions between neutrophils and intestinal epithelial cells (IECs) is still not well characterized. In this study, we demonstrated that neutrophils enhanced production of amphiregulin (AREG), a member of the EGFR ligand family, by IECs, which promoted IEC barrier function and tissue repair. Depletion of neutrophils resulted in more severe colitis in mice because of decreased AREG production by IECs upon dextran sodium sulfate (DSS) insult. Administration of AREG restored epithelial barrier function and ameliorated colitis. Furthermore, neutrophil-derived TGF-ß promoted AREG production by IECs. Mechanistically, TGF-ß activated MEK1/2 signaling, and inhibition of MEK1/2 abrogated TGF-ß-induced AREG production by IECs. Collectively, these findings reveal that neutrophils play an important role in the maintenance of IEC barrier function and homeostasis.


Asunto(s)
Anfirregulina/metabolismo , Colitis/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/fisiología , Neutrófilos/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Células Cultivadas , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Femenino , Homeostasis , Humanos , MAP Quinasa Quinasa 1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal
7.
Parasitol Res ; 118(7): 2277-2285, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31119381

RESUMEN

Malaria-associated bacteremia accounts for up to one-third of deaths from severe malaria, and non-typhoidal Salmonella (NTS) has been reported as a major complication of severe malarial infection. Patients who develop NTS bacteremia during Plasmodium infection show higher mortality rates than individuals with malaria alone. Systemic bacteremia can be caused by a wound or translocation from epithelial or endothelial sites. NTS is an intestinal pathogen, however the contribution of bacterial translocation from the intestinal tract during Plasmodium infection is not well studied. Here, we investigated the integrity of the intestinal barrier function of P. chabaudi-infected mice using large molecules and Salmonella infection. Intestinal histology and the adaptive immune response to malaria were also studied using light microscopy and flow cytometry. P. chabaudi infection compromised intestinal barrier function, which led to increased intestinal cellular infiltration. In addition, we observed increased serum lipopolysaccharide binding protein and leakage of soluble molecules from the intestine into the blood in infected mice. Plasmodium infection also increased intestinal translocation and dissemination of NTS to the liver. The adaptive immune response to P. chabaudi infection was also significantly impacted by NTS translocation. Reduced B and T cell activation were observed in co-infected animals, suggesting interference in the malaria-specific immune responses by bacteremia. These studies demonstrate that P. chabaudi infection induces failure of the barrier function of the intestinal wall and enhanced intestinal bacterial translocation, affecting anti-malarial immunity.


Asunto(s)
Inmunidad Adaptativa , Malaria/inmunología , Plasmodium chabaudi/inmunología , Infecciones por Salmonella/inmunología , Salmonella/inmunología , Animales , Bacteriemia , Coinfección , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Intestinos/microbiología , Intestinos/patología , Activación de Linfocitos , Malaria/complicaciones , Malaria/parasitología , Malaria/patología , Ratones , Ratones Endogámicos C57BL , Parasitemia , Infecciones por Salmonella/complicaciones , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología
8.
Artículo en Inglés | MEDLINE | ID: mdl-29109161

RESUMEN

Earlier, we reported that three Food and Drug Administration-approved drugs, trifluoperazine (TFP; an antipsychotic), amoxapine (AXPN; an antidepressant), and doxapram (DXP; a breathing stimulant), identified from an in vitro murine macrophage cytotoxicity screen, provided mice with 40 to 60% protection against pneumonic plague when administered at the time of infection for 1 to 3 days. In the present study, the therapeutic potential of these drugs against pneumonic plague in mice was further evaluated when they were administered at up to 48 h postinfection. While the efficacy of TFP was somewhat diminished as treatment was delayed to 24 h, the protection of mice with AXPN and DXP increased as treatment was progressively delayed to 24 h. At 48 h postinfection, these drugs provided the animals with significant protection (up to 100%) against challenge with the agent of pneumonic or bubonic plague when they were administered in combination with levofloxacin. Likewise, when they were used in combination with vancomycin, all three drugs provided mice with 80 to 100% protection from fatal oral Clostridium difficile infection when they were administered at 24 h postinfection. Furthermore, AXPN provided 40 to 60% protection against respiratory infection with Klebsiella pneumoniae when it was administered at the time of infection or at 24 h postinfection. Using the same in vitro cytotoxicity assay, we identified an additional 76/780 nonantibiotic drugs effective against K. pneumoniae For Acinetobacter baumannii, 121 nonantibiotic drugs were identified to inhibit bacterium-induced cytotoxicity in murine macrophages. Of these 121 drugs, 13 inhibited the macrophage cytotoxicity induced by two additional multiple-antibiotic-resistant strains. Six of these drugs decreased the intracellular survival of all three A. baumannii strains in macrophages. These results provided further evidence of the broad applicability and utilization of drug repurposing screening to identify new therapeutics to combat multidrug-resistant pathogens of public health concern.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Peste/tratamiento farmacológico , Acinetobacter baumannii/efectos de los fármacos , Amoxapina/farmacología , Animales , Línea Celular , Modelos Animales de Enfermedad , Doxapram/farmacología , Reposicionamiento de Medicamentos/métodos , Femenino , Klebsiella pneumoniae/efectos de los fármacos , Levofloxacino/farmacología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Peste/microbiología , Células RAW 264.7 , Trifluoperazina/farmacología
9.
J Immunol ; 196(10): 4390-9, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27067005

RESUMEN

It has been shown recently that neutrophils are able to produce IL-22 and IL-17, which differentially regulate the pathogenesis of inflammatory bowel disease. However, it is still largely unknown how the neutrophil production of IL-22 and IL-17 is regulated, and their role in the pathogenesis of inflammatory bowel disease. In this study, we found that IL-23 promoted neutrophil production of IL-17 and IL-22. IL-23 stimulated the neutrophil expression of IL-23R as well as rorc and ahr. Retinoid acid receptor-related orphan receptor γ t and aryl-hydrocarbon receptor differentially regulated IL-23 induction of neutrophil IL-17 and IL-22. In addition, IL-23 induced the activation of mTOR in neutrophils. Blockade of the mTOR pathway inhibited IL-23-induced expression of rorc and ahr, as well as IL-17 and IL-22 production. By using a microbiota Ag-specific T cell-mediated colitis model, we demonstrated that depletion of neutrophils, as well as blockade of IL-22, resulted in a significant increase in the severity of colitis, thereby indicating a protective role of neutrophils and IL-22 in chronic colitis. Collectively, our data revealed that neutrophils negatively regulate microbiota Ag-specific T cell induction of colitis, and IL-23 induces neutrophil production of IL-22 and IL-17 through induction of rorc and ahr, which is mediated by the mTOR pathway.


Asunto(s)
Interleucina-17/biosíntesis , Interleucina-23/metabolismo , Interleucinas/biosíntesis , Neutrófilos/metabolismo , Receptores de Interleucina/metabolismo , Serina-Treonina Quinasas TOR/genética , Animales , Ciego/patología , Diferenciación Celular , Colitis/inmunología , Colitis/patología , Colon/patología , Interleucina-23/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Receptores de Interleucina/genética , Transducción de Señal , Células Th17/inmunología , Interleucina-22
11.
Anaerobe ; 54: 83-91, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30099125

RESUMEN

BACKGROUND: Morbidity and mortality associated with Clostridioides (formerly Clostridium) difficile infection (CDI) rises progressively with advanced age (≥65 years) due in part to perturbations of the gut microbiota and immune dysfunction. Epidemiological data of community-acquired CDI suggests increased susceptibility may begin earlier during middle-age (45-64 years) but the causation remains unknown. METHODS: Middle-aged (12-14 months) and young (2-4 months) adult mice were infected with C. difficile, and disease severity, gut microbiome and innate immune response were compared. Cytokine reconstitution studies were performed in infected middle-aged mice. RESULTS: Infection of middle-aged mice with C. difficile led to greater disease compared to young controls, which was associated with increases in C. difficile burden and toxin titers, and elevated bacterial translocation. With the exception of an expansion of C. difficile in middle-aged mice, microbiome analysis revealed no age-related differences. In contrast, middle-aged mice displayed a significant defect in neutrophil recruitment to the colon, with diminished levels of innate immune cytokines IL-6, IL-23 and IL-22. Importantly, recombinant IL-22 administration during CDI reduced morbidity and prevented death in middle-aged mice. CONCLUSION: Increased susceptibility to C. difficile occurs in middle-aged mice modeling the community-acquired CDI demographics and is driven by an impaired innate immune response.


Asunto(s)
Envejecimiento/inmunología , Clostridioides difficile/fisiología , Infecciones por Clostridium/inmunología , Interleucinas/inmunología , Neutrófilos/inmunología , Factores de Edad , Animales , Clostridioides difficile/inmunología , Infecciones por Clostridium/genética , Infecciones por Clostridium/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Inmunidad Innata , Interleucinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Interleucina-22
12.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28760934

RESUMEN

Integration of antibiotic and probiotic therapy has the potential to lessen the public health burden of antimicrobial-associated diseases. Clostridium difficile infection (CDI) represents an important example where the rational design of next-generation probiotics is being actively pursued to prevent disease recurrence. Because intrinsic resistance to clinically relevant antibiotics used to treat CDI (vancomycin, metronidazole, and fidaxomicin) is a desired trait in such probiotic species, we screened several bacteria and identified Lactobacillus reuteri to be a promising candidate for adjunct therapy. Human-derived L. reuteri bacteria convert glycerol to the broad-spectrum antimicrobial compound reuterin. When supplemented with glycerol, strains carrying the pocR gene locus were potent reuterin producers, with L. reuteri 17938 inhibiting C. difficile growth at a level on par with the level of growth inhibition by vancomycin. Targeted pocR mutations and complementation studies identified reuterin to be the precursor-induced antimicrobial agent. Pathophysiological relevance was demonstrated when the codelivery of L. reuteri with glycerol was effective against C. difficile colonization in complex human fecal microbial communities, whereas treatment with either glycerol or L. reuteri alone was ineffective. A global unbiased microbiome and metabolomics analysis independently confirmed that glycerol precursor delivery with L. reuteri elicited changes in the composition and function of the human microbial community that preferentially targets C. difficile outgrowth and toxicity, a finding consistent with glycerol fermentation and reuterin production. Antimicrobial resistance has thus been successfully exploited in the natural design of human microbiome evasion of C. difficile, and this method may provide a prototypic precursor-directed probiotic approach. Antibiotic resistance and substrate bioavailability may therefore represent critical new determinants of probiotic efficacy in clinical trials.


Asunto(s)
Antibacterianos/biosíntesis , Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/prevención & control , Gliceraldehído/análogos & derivados , Glicerol/administración & dosificación , Limosilactobacillus reuteri/metabolismo , Probióticos , Propano/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Clostridioides difficile/efectos de los fármacos , Infecciones por Clostridium/inmunología , Infecciones por Clostridium/terapia , Descubrimiento de Drogas/métodos , Farmacorresistencia Bacteriana , Heces/microbiología , Fermentación , Microbioma Gastrointestinal , Gliceraldehído/metabolismo , Gliceraldehído/farmacología , Gliceraldehído/uso terapéutico , Glicerol/inmunología , Glicerol/metabolismo , Humanos , Metabolómica , Propano/farmacología , Propano/uso terapéutico , Vancomicina/farmacología
13.
Antimicrob Agents Chemother ; 60(6): 3717-29, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27067323

RESUMEN

Antibiotic resistance in medically relevant bacterial pathogens, coupled with a paucity of novel antimicrobial discoveries, represents a pressing global crisis. Traditional drug discovery is an inefficient and costly process; however, systematic screening of Food and Drug Administration (FDA)-approved therapeutics for other indications in humans offers a rapid alternative approach. In this study, we screened a library of 780 FDA-approved drugs to identify molecules that rendered RAW 264.7 murine macrophages resistant to cytotoxicity induced by the highly virulent Yersinia pestis CO92 strain. Of these compounds, we identified 94 not classified as antibiotics as being effective at preventing Y. pestis-induced cytotoxicity. A total of 17 prioritized drugs, based on efficacy in in vitro screens, were chosen for further evaluation in a murine model of pneumonic plague to delineate if in vitro efficacy could be translated in vivo Three drugs, doxapram (DXP), amoxapine (AXPN), and trifluoperazine (TFP), increased animal survivability despite not exhibiting any direct bacteriostatic or bactericidal effect on Y. pestis and having no modulating effect on crucial Y. pestis virulence factors. These findings suggested that DXP, AXPN, and TFP may modulate host cell pathways necessary for disease pathogenesis. Finally, to further assess the broad applicability of drugs identified from in vitro screens, the therapeutic potential of TFP, the most efficacious drug in vivo, was evaluated in murine models of Salmonella enterica serovar Typhimurium and Clostridium difficile infections. In both models, TFP treatment resulted in increased survivability of infected animals. Taken together, these results demonstrate the broad applicability and potential use of nonantibiotic FDA-approved drugs to combat respiratory and gastrointestinal bacterial pathogens.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Reposicionamiento de Medicamentos , Enterocolitis Seudomembranosa/tratamiento farmacológico , Peste/tratamiento farmacológico , Infecciones por Salmonella/tratamiento farmacológico , Trifluoperazina/farmacología , Amoxapina/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/crecimiento & desarrollo , Clostridioides difficile/patogenicidad , Modelos Animales de Enfermedad , Doxapram/farmacología , Esquema de Medicación , Enterocolitis Seudomembranosa/metabolismo , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/mortalidad , Femenino , Ensayos Analíticos de Alto Rendimiento , Macrófagos/efectos de los fármacos , Ratones , Peste/metabolismo , Peste/microbiología , Peste/mortalidad , Medicamentos bajo Prescripción/farmacología , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/mortalidad , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/patogenicidad , Bibliotecas de Moléculas Pequeñas/farmacología , Análisis de Supervivencia , Yersinia pestis/efectos de los fármacos , Yersinia pestis/crecimiento & desarrollo , Yersinia pestis/patogenicidad
14.
Eur J Immunol ; 45(4): 1010-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25605286

RESUMEN

Differentiated CD4(+) T cells preserve plasticity under various conditions. However, the stability of Th1 cells is unclear, as is whether Th1 cells can convert into Th17 cells and thereby contribute to the generation of IFN-γ(+) IL-17(+) CD4(+) T cells, the number of which correlates with severity of colitis. We investigated whether IFN-γ(+) Th1 cells can convert into Th17 cells under intestinal inflammation and the mechanisms involved. IFN-γ(Thy1.1+) Th1 cells were generated by culturing naïve CD4(+) T cells from IFN-γ(Thy1.1) CBir1 TCR-Tg reporter mice, whose TCR is specific for an immunodominant microbiota antigen, CBir1 flagellin, under Th1 polarizing conditions. IFN-γ(Thy1.1+) Th1 cells induced colitis in Rag(-/-) mice after adoptive transfer and converted into IL-17(+) Th17, but not Foxp3(+) Treg cells in the inflamed intestines. TGF-ß and IL-6, but not IL-1ß and IL-23, regulated Th1 conversion into Th17 cells. TGF-ß induction of transcriptional factor Runx1 is crucial for the conversion, since silencing Runx1 by siRNA inhibited Th1 conversion into Th17 cells. Furthermore, TGF-ß enhanced histone H3K9 acetylation but inhibited H3K9 trimethylation of Runx1- and ROR-γt-binding sites on il-17 or rorc gene in Th1 cells. We conclude that Th1 cells convert into Th17 cells under inflammatory conditions in intestines, which is possibly mediated by TGF-ß induction of Runx1.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/biosíntesis , Mucosa Intestinal/inmunología , Células TH1/citología , Células Th17/citología , Factor de Crecimiento Transformador beta/metabolismo , Acetilación , Animales , Sitios de Unión , Diferenciación Celular/inmunología , Células Cultivadas , Colitis/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Flagelina/inmunología , Histonas/metabolismo , Proteínas de Homeodominio/genética , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Interleucina-1beta/metabolismo , Interleucina-2/metabolismo , Interleucina-23/metabolismo , Interleucina-6/metabolismo , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Células TH1/inmunología , Células Th17/inmunología
15.
J Immunol ; 191(2): 837-47, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23772033

RESUMEN

The p53 protein has not only important tumor suppressor activity but also additional immunological and other functions, whose nature and extent are just beginning to be recognized. In this article, we show that p53 has a novel inflammation-promoting action in the intestinal tract, because loss of p53 or the upstream activating kinase, ATM, protects against acute intestinal inflammation in murine models. Mechanistically, deficiency in p53 leads to increased survival of epithelial cells and lamina propria macrophages, higher IL-6 expression owing to enhanced glucose-dependent NF-κB activation, and increased mucosal STAT3 activation. Blockade or loss of IL-6 signaling reverses the protective effects of p53 deficiency. Conversely, IL-6 treatment protects against acute colitis in a manner dependent on STAT3 signaling and induction of cytoprotective factors in epithelial cells. Together, these results indicate that p53 promotes inflammation in the intestinal tract through suppression of epithelium-protective factors, thus significantly expanding the spectrum of physiological and immunological p53 activities unrelated to cancer formation.


Asunto(s)
Colitis/inmunología , Colitis/prevención & control , Inflamación/inmunología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Células de la Médula Ósea/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Colitis/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/metabolismo , Activación Enzimática , Células Epiteliales/metabolismo , Inflamación/prevención & control , Interleucina-6/biosíntesis , Interleucina-6/farmacología , Interleucinas/biosíntesis , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Interleucina-22
16.
Exp Parasitol ; 156: 68-78, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26071205

RESUMEN

Giardia lamblia is a leading protozoan cause of diarrheal disease worldwide. It colonizes the lumen and epithelial surface of the small intestine, but does not invade the mucosa. Acute infection causes only minimal mucosal inflammation. Effective immune defenses exist, yet their identity and mechanisms remain incompletely understood. Interleukin (IL)-17A has emerged as an important cytokine involved in inflammation and antimicrobial defense against bacterial pathogens at mucosal surfaces. In this study, we demonstrate that IL-17A has a crucial function in host defense against Giardia infection. Using murine infection models with G. muris and G. lamblia, we observed marked and selective induction of intestinal IL-17A with peak expression after 2 weeks. Th17 cells in the lamina propria and innate immune cells in the epithelial compartment of the small intestine were responsible for the IL-17A response. Experiments in gene-targeted mice revealed that the cytokine, and its cognate receptor IL-17RA, were required for eradication of the parasite. The actions of the cytokine were mediated by hematopoietic cells, and were required for the transport of IgA into the intestinal lumen, since IL-17A deficiency led to marked reduction of fecal IgA levels, as well as for increased intestinal expression of several other potential effectors, including ß-defensin 1 and resistin-like molecule ß. In contrast, intestinal hypermotility, another major antigiardial defense mechanism, was not impacted by IL-17A loss. Taken together, these findings demonstrate that IL-17A and IL-17 receptor signaling are essential for intestinal defense against the important lumen-dwelling intestinal parasite Giardia.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Giardia/inmunología , Giardiasis/inmunología , Inmunoglobulina A/biosíntesis , Interleucina-17/metabolismo , Animales , Anticuerpos Antiprotozoarios/inmunología , Linfocitos T CD4-Positivos/inmunología , Quimera , Giardia lamblia/inmunología , Células Madre Hematopoyéticas/inmunología , Inmunoglobulina A/inmunología , Interleucina-17/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Intestino Delgado/inmunología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos , Células Th17/inmunología
17.
Infect Immun ; 82(5): 1949-58, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24566625

RESUMEN

Interleukin-10 (IL-10) curtails immune responses to microbial infection and autoantigens and contributes to intestinal immune homeostasis, yet administration of IL-10 has not been effective at attenuating chronic intestinal inflammatory conditions, suggesting that its immune functions may be context dependent. To gain a broader understanding of the importance of IL-10 in controlling mucosal immune responses to infectious challenges, we employed the murine attaching and effacing pathogen Citrobacter rodentium, which colonizes primarily the surfaces of the cecum and colon and causes transient mucosal inflammation driven by Th17 and Th1 T helper cells. Infection induced macrophage and dendritic cell production of IL-10, which diminished antibacterial host defenses, because IL-10-deficient mice cleared infection faster than wild-type controls. In parallel, the mice had less acute infection-associated colitis and resolved it more rapidly than controls. Importantly, transient C. rodentium infection protected IL-10-deficient mice against the later development of spontaneous colitis that normally occurs with aging in these mice. Genome-wide expression studies revealed that IL-10 deficiency was associated with downregulation of proinflammatory pathways but increased expression of the anti-inflammatory cytokine IL-27 in response to infection. IL-27 was found to suppress in vitro Th17 and, to a lesser degree, Th1 differentiation independent of IL-10. Furthermore, neutralization of IL-27 resulted in more severe colitis in infected IL-10-deficient mice. Together, these findings indicate that IL-10 is dispensable for resolving C. rodentium-associated colitis and further suggest that IL-27 may be a critical factor for controlling intestinal inflammation and Th17 and Th1 development by IL-10-independent mechanisms.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae/microbiología , Inflamación/microbiología , Interleucina-10/metabolismo , Envejecimiento , Animales , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/patología , Femenino , Regulación de la Expresión Génica/inmunología , Interleucina-10/genética , Interleucinas/genética , Interleucinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Eur J Immunol ; 43(7): 1716-26, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23620016

RESUMEN

Although the development of T-cell subsets is mainly regulated by a master transcriptional regulator and phosphorylation of the STAT protein in response to distinct cytokine stimulation, accumulating data indicate that other signaling pathways are also involved in regulating or fine-tuning T-cell lineage commitment. In this report, we investigated the role of ERK, mitogen-activated protein kinase (MAPK), in Th17 and Treg cell development. We demonstrate that blockade of ERK activation inhibited Th17-cell development while upregulating Treg cells under Th17 polarization conditions. Inhibition of ERK decreased IL-6 induction of RAR-related orphan receptor γt but enhanced TGF-ß induction of Foxp3, and ERK inhibitor-treated T cells under Th17 conditions possessed suppressive function in vitro because they produced more IL-10 and TGF-ß and inhibited naïve T-cell proliferation and IFN-γ production at levels comparable with that of Treg cells. Furthermore, ERK inhibitor-treated T cells under Th17 polarization conditions had a decreased potency to induce colitis in vivo. Collectively, our data demonstrated that the ERK pathway differentially regulates Th17- and Treg-cell differentiation, and thus interfering with the ERK pathway could represent a therapeutic treatment for inflammatory bowel diseases and other Th17-related autoimmune diseases.


Asunto(s)
Diferenciación Celular , Colitis/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Immunoblotting , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T Reguladores/citología , Células Th17/citología , Transfección
19.
J Infect Dis ; 208(8): 1342-8, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23878324

RESUMEN

Cryptosporidium parasites infect intestinal cells, causing cryptosporidiosis. Despite its high morbidity and association with stunting in the developing world, current therapies for cryptosporidiosis have limited efficacy. Calcium-dependent protein kinases (CDPKs) are essential enzymes in the biology of protozoan parasites. CDPK1 was cloned from the genome of Cryptosporidium parvum, and potent and specific inhibitors have been developed based on structural studies. In this study, we evaluated the anti-Cryptosporidium activity of a novel CDPK1 inhibitor, 1294, and demonstrated that 1294 significantly reduces parasite infection in vitro, with a half maximal effective concentration of 100 nM. Pharmacokinetic studies revealed that 1294 is well absorbed, with a half-life supporting daily administration. Oral therapy with 1294 eliminated Cryptosporidium parasites from 6 of 7 infected severe combined immunodeficiency-beige mice, and the parasites did not recur in these immunosuppressed mice. Mice treated with 1294 had less epithelial damage, corresponding to less apoptosis. Thus, 1294 is an important lead for the development of drugs for treatment of cryptosporidiosis.


Asunto(s)
Antiprotozoarios/farmacología , Criptosporidiosis/tratamiento farmacológico , Cryptosporidium parvum/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antiprotozoarios/química , Antiprotozoarios/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular , Criptosporidiosis/enzimología , Criptosporidiosis/parasitología , Cryptosporidium parvum/enzimología , Cryptosporidium parvum/genética , Cryptosporidium parvum/aislamiento & purificación , Genes Protozoarios , Humanos , Intestinos/parasitología , Intestinos/patología , Ratones , Ratones SCID , Carga de Parásitos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
20.
Curr Opin Infect Dis ; 26(5): 447-53, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23982235

RESUMEN

PURPOSE OF REVIEW: Clostridium difficile infection (CDI) is the leading cause of antibiotic-associated diarrhea and pseudomembranous colitis in the healthcare setting. An emerging consensus suggests that CDI is caused by pathogenic toxin production, gut microbial dysbiosis and altered host inflammatory responses. The aim of this review is to summarize and highlight recent advances focused on CDI pathogenic mechanisms. RECENT FINDINGS: Potential paradigm shifts relating to the mechanisms of toxin action and inhibition have recently been reported, with new insights into spore germination and surface protein function also gaining traction. Multiomic analysis of microbiome dysbiosis has identified important CDI-associated microbial community shifts that may form the basis of future targeted bacteriotherapy, and functional metabolite biomarkers that require further characterization. Classical innate and adaptive immunity against CDI is rapidly being delineated, with novel innate S-nitrosylation signals also being identified. SUMMARY: Studies in patients and animal disease models are shedding new light on the critical roles of the microbiota, metabolome and host responses in primary and recurrent CDI. An improved understanding of the CDI disease pathogenesis will provide the basis for developing new therapies for treating disease and preventing recurrence.


Asunto(s)
Clostridioides difficile/patogenicidad , Infecciones por Clostridium/microbiología , Animales , Infecciones por Clostridium/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Interacciones Huésped-Patógeno , Humanos , Microbiota
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA