Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(24): 11988-11996, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31138692

RESUMEN

Undernutrition in children is a pressing global health problem, manifested in part by impaired linear growth (stunting). Current nutritional interventions have been largely ineffective in overcoming stunting, emphasizing the need to obtain better understanding of its underlying causes. Treating Bangladeshi children with severe acute malnutrition with therapeutic foods reduced plasma levels of a biomarker of osteoclastic activity without affecting biomarkers of osteoblastic activity or improving their severe stunting. To characterize interactions among the gut microbiota, human milk oligosaccharides (HMOs), and osteoclast and osteoblast biology, young germ-free mice were colonized with cultured bacterial strains from a 6-mo-old stunted infant and fed a diet mimicking that consumed by the donor population. Adding purified bovine sialylated milk oligosaccharides (S-BMO) with structures similar to those in human milk to this diet increased femoral trabecular bone volume and cortical thickness, reduced osteoclasts and their bone marrow progenitors, and altered regulators of osteoclastogenesis and mediators of Th2 responses. Comparisons of germ-free and colonized mice revealed S-BMO-dependent and microbiota-dependent increases in cecal levels of succinate, increased numbers of small intestinal tuft cells, and evidence for activation of a succinate-induced tuft cell signaling pathway linked to Th2 immune responses. A prominent fucosylated HMO, 2'-fucosyllactose, failed to elicit these changes in bone biology, highlighting the structural specificity of the S-BMO effects. These results underscore the need to further characterize the balance between, and determinants of, osteoclastic and osteoblastic activity in stunted infants/children, and suggest that certain milk oligosaccharides may have therapeutic utility in this setting.


Asunto(s)
Huesos/efectos de los fármacos , Vida Libre de Gérmenes/efectos de los fármacos , Desnutrición/tratamiento farmacológico , Leche Humana/metabolismo , Oligosacáridos/administración & dosificación , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Animales , Bacterias/efectos de los fármacos , Bovinos , Dieta , Modelos Animales de Enfermedad , Heces/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Lactante , Intestino Delgado/microbiología , Masculino , Desnutrición/microbiología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos
2.
Infect Immun ; 84(8): 2317-2323, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27271747

RESUMEN

Clostridium difficile is a major, life-threatening hospital-acquired pathogen that causes mild to severe colitis in infected individuals. The tissue destruction and inflammation which characterize C. difficile infection (CDI) are primarily due to the Rho-glucosylating toxins A and B. These toxins cause epithelial cell death and induce robust inflammatory signaling by activating the transcription factor NF-κB, leading to chemokine and cytokine secretion. The toxins also activate the inflammasome complex, which leads to secretion of the pyrogenic cytokine IL-1ß. In this study, we utilized glucosylation-deficient toxin A to show that activation of the inflammasome by this toxin is dependent on Rho glucosylation, confirming similar findings reported for toxin B. We also demonstrated that tissue destruction and in vivo inflammatory cytokine production are critically dependent on the enzymatic activity of toxin A, suggesting that inhibiting toxin glucosyltransferase activity may be effective in combating this refractory disease.


Asunto(s)
Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Clostridioides difficile/inmunología , Clostridioides difficile/metabolismo , Infecciones por Clostridium/metabolismo , Infecciones por Clostridium/microbiología , Enterotoxinas/inmunología , Enterotoxinas/metabolismo , Inmunidad Innata , Animales , Toxinas Bacterianas/genética , Biomarcadores , Infecciones por Clostridium/patología , Citocinas/metabolismo , Enterotoxinas/genética , Glicosilación , Mediadores de Inflamación/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo
3.
Infect Immun ; 84(10): 2824-32, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27456830

RESUMEN

Intestinal segmented filamentous bacteria (SFB) protect from ameba infection, and protection is transferable with bone marrow dendritic cells (BMDCs). SFB cause an increase in serum amyloid A (SAA), suggesting that SAA might mediate SFB's effects on BMDCs. Here we further explored the role of bone marrow in SFB-mediated protection. Transient gut colonization with SFB or SAA administration alone transiently increased the H3K27 histone demethylase Jmjd3, persistently increased bone marrow Csf2ra expression and granulocyte monocyte precursors (GMPs), and protected from ameba infection. Pharmacologic inhibition of Jmjd3 H3K27 demethylase activity during SAA treatment or blockade of granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling in SFB-colonized mice prevented GMP expansion, decreased gut neutrophils, and blocked protection from ameba infection. These results indicate that alteration of the microbiota and systemic exposure to SAA can influence myelopoiesis and susceptibility to amebiasis via epigenetic mechanisms. Gut microbiota-marrow communication is a previously unrecognized mechanism of innate protection from infection.


Asunto(s)
Células de la Médula Ósea/citología , Entamoeba histolytica/fisiología , Entamebiasis/fisiopatología , Tracto Gastrointestinal/microbiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Proteína Amiloide A Sérica/fisiología , Animales , Bacterias , Médula Ósea/metabolismo , Células de la Médula Ósea/fisiología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Células Progenitoras de Granulocitos y Macrófagos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
4.
Anaerobe ; 30: 205-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25223264

RESUMEN

Clostridium difficile is a Gram-positive, spore forming bacillus and the most common cause of antibiotic-associated diarrhea in the United States. Clinical outcomes of C. difficile infection (CDI) range from asymptomatic colonization to pseudomembranous colitis, sepsis and death. Disease is primarily mediated by the action of the Rho-glucosylating toxins A and B, which induce potent pro-inflammatory signaling within the host. The role of this inflammatory response during infection is just beginning to be appreciated, with recent data suggesting inflammatory markers correlate closely with disease severity. In addition to the toxins, multiple innate immune signaling pathways have been implicated in establishing an inflammatory response during infection. In intoxication-based models of disease, inflammation typically enhances pathogenesis, while protection from infection seems to require some level of inflammatory response. Thus, the host immune response plays a key role in shaping the course of infection and a balanced inflammatory response which eradicates infection without damaging host tissues is likely required for successful resolution of disease.


Asunto(s)
Clostridioides difficile/inmunología , Clostridioides difficile/fisiología , Interacciones Huésped-Patógeno , Inmunidad Innata , Toxinas Bacterianas/toxicidad , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/patología , Humanos , Transducción de Señal
5.
Microbiome ; 12(1): 71, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38589975

RESUMEN

BACKGROUND: Childhood undernutrition is a major global health challenge with devastating lifelong consequences. Linear growth stunting due to undernutrition has been linked to poor health outcomes, and mothers who experience growth stunting in childhood are more likely to give birth to stunted children later in life. Based on these findings, we hypothesized that intergenerational colonization of mice with microbiota from human donors with undernutrition may recapitulate certain immune and growth changes observed in this disorder. RESULTS: To test this hypothesis, we developed a gnotobiotic murine model of undernutrition using microbiota from human infants with healthy or stunted growth trajectories. Intergenerational colonization with microbiota derived from children with growth stunting lead to less linear growth and the development of immune features of undernutrition and enteropathy, including intestinal villus blunting, lower liver IGF-1 and accumulation of intraepithelial lymphocytes and plasma cells in the small intestine. In contrast, colonization after weaning lead to fewer host phenotypic changes between these distinct microbial communities. CONCLUSIONS: These results are broadly consistent with previous findings demonstrating that exposure of the immune system to microbial products during the weaning phase is a critical determinant of later life immune function. Overall, our results suggest intergenerational colonization with human microbiota samples is a useful approach with which to investigate microbiota-dependent changes in growth and immunity in early life. Murine models that capture the intergenerational and multifactorial nature of undernutrition are critical to understanding the underlying biology of this disorder. Video Abstract.


Asunto(s)
Microbioma Gastrointestinal , Desnutrición , Microbiota , Animales , Humanos , Lactante , Ratones , Trastornos del Crecimiento , Intestino Delgado
6.
Cell Host Microbe ; 31(5): 685-687, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37167948

RESUMEN

In a recent report in Science, Schwarzer and colleagues demonstrate the growth benefits of treatment with Lactiplantibacillus plantarum strain WJL in a preclinical mouse model of chronic undernutrition. L. plantarum influences the somatotropic axis to promote growth through intestinal epithelial NOD2 sensing.


Asunto(s)
Mucosa Intestinal , Desnutrición , Animales , Ratones , Microbioma Gastrointestinal/genética , Crecimiento/genética , Crecimiento/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Desnutrición/genética , Desnutrición/microbiología , Desnutrición/fisiopatología , Desnutrición/terapia
7.
bioRxiv ; 2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37461523

RESUMEN

Childhood undernutrition is a major global health challenge with devastating lifelong consequences. Linear growth stunting due to undernutrition has been linked to poor outcomes, and mothers who experience stunting are more likely to give birth to stunted children. Murine models that capture the intergenerational and multifactorial nature of undernutrition are critical to understanding the underlying biology of this disorder. Here we report a gnotobiotic mouse model of undernutrition using microbiota from human infants with healthy or stunted growth trajectories. Intergenerational transmission of microbiota from parents to offspring leads to the development of growth and immune features of undernutrition and enteropathy, including reduced linear growth, intestinal villus blunting and accumulation of intraepithelial lymphocytes. In contrast, colonization after weaning reduces sensitivity to detect changes driven by distinct microbial communities. Overall, these results suggest intergenerational colonization is a useful approach with which to investigate microbiota-dependent growth and immunity in early life.

8.
Nat Rev Gastroenterol Hepatol ; 20(4): 223-237, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36526906

RESUMEN

Environmental enteric dysfunction (EED) is a subclinical syndrome of intestinal inflammation, malabsorption and barrier disruption that is highly prevalent in low- and middle-income countries in which poverty, food insecurity and frequent exposure to enteric pathogens impair growth, immunity and neurodevelopment in children. In this Review, we discuss advances in our understanding of EED, intestinal adaptation and the gut microbiome over the 'first 1,000 days' of life, spanning pregnancy and early childhood. Data on maternal EED are emerging, and they mirror earlier findings of increased risks for preterm birth and fetal growth restriction in mothers with either active inflammatory bowel disease or coeliac disease. The intense metabolic demands of pregnancy and lactation drive gut adaptation, including dramatic changes in the composition, function and mother-to-child transmission of the gut microbiota. We urgently need to elucidate the mechanisms by which EED undermines these critical processes so that we can improve global strategies to prevent and reverse intergenerational cycles of undernutrition.


Asunto(s)
Síndromes de Malabsorción , Microbiota , Nacimiento Prematuro , Lactante , Recién Nacido , Femenino , Preescolar , Humanos , Embarazo , Transmisión Vertical de Enfermedad Infecciosa , Intestino Delgado
9.
Sci Transl Med ; 14(640): eabk1107, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35417188

RESUMEN

Disrupted development of the gut microbiota is a contributing cause of childhood malnutrition. Bifidobacterium longum subspecies infantis is a prominent early colonizer of the infant gut that consumes human milk oligosaccharides (HMOs). We found that the absolute abundance of Bifidobacterium infantis is lower in 3- to 24-month-old Bangladeshi infants with severe acute malnutrition (SAM) compared to their healthy age-matched counterparts. A single-blind, placebo-controlled trial (SYNERGIE) was conducted in 2- to 6-month-old Bangladeshi infants with SAM. A commercial U.S. donor-derived B. infantis strain (EVC001) was administered daily with or without the HMO lacto-N-neotetraose for 28 days. This intervention increased fecal B. infantis abundance in infants with SAM, although to levels still 10- to 100-fold lower than in untreated healthy controls. EVC001 treatment promoted weight gain that was associated with reduced intestinal inflammation markers in infants with SAM. We cultured fecal B. infantis strains from Bangladeshi infants and colonized gnotobiotic mice with these cultured strains. The gnotobiotic mice were fed a diet representative of that consumed by 6-month-old Bangladeshi infants, with or without HMO supplementation. One B. infantis strain, Bg_2D9, expressing two gene clusters involved in uptake and utilization of N-glycans and plant-derived polysaccharides, exhibited superior fitness over EVC001. The fitness advantage of Bg_2D9 was confirmed in a gnotobiotic mouse model of mother-to-infant gut microbiota transmission where dams received a pretreatment fecal community from a SAM infant in the SYNERGIE trial. Whether Bg_2D9 is superior to EVC001 for treating malnourished infants who consume a diet with limited breastmilk requires further clinical testing.


Asunto(s)
Bifidobacterium longum subspecies infantis , Desnutrición Aguda Severa , Animales , Bifidobacterium , Heces/microbiología , Humanos , Lactante , Ratones , Leche Humana , Método Simple Ciego , Aumento de Peso
10.
Cell Host Microbe ; 25(5): 756-765.e5, 2019 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-31003940

RESUMEN

Clostridium difficile infection (CDI) is the number one hospital-acquired infection in the United States. CDI is more common and severe in inflammatory bowel disease patients. Here, we studied the mechanism by which prior colitis exacerbates CDI. Mice were given dextran sulfate sodium (DSS) colitis, recovered for 2 weeks, and then were infected with C. difficile. Mortality and CDI severity were increased in DSS-treated mice compared to controls. Severe CDI is dependent on CD4+ T cells, which persist after colitis-associated inflammation subsides. Adoptive transfer of Th17 cells to naive mice is sufficient to increase CDI-associated mortality through elevated IL-17 production. Finally, in humans, the Th17 cytokines IL-6 and IL-23 associate with severe CDI, and patients with high serum IL-6 are 7.6 times more likely to die post infection. These findings establish a central role for Th17 cells in CDI pathogenesis following colitis and identify them as a potential target for preventing severe disease.


Asunto(s)
Clostridioides difficile/crecimiento & desarrollo , Infecciones por Clostridium/inmunología , Colitis/complicaciones , Colitis/patología , Susceptibilidad a Enfermedades , Células Th17/inmunología , Adolescente , Traslado Adoptivo , Adulto , Anciano , Animales , Niño , Infecciones por Clostridium/mortalidad , Infecciones por Clostridium/patología , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/patología , Subunidad p19 de la Interleucina-23/sangre , Interleucina-6/sangre , Masculino , Ratones , Persona de Mediana Edad , Medición de Riesgo , Análisis de Supervivencia , Adulto Joven
11.
Nat Commun ; 10(1): 2712, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-31221971

RESUMEN

Clostridium difficile (C. difficile) incidence has tripled over the past 15 years and is attributed to the emergence of hypervirulent strains. While it is clear that C. difficile toxins cause damaging colonic inflammation, the immune mechanisms protecting from tissue damage require further investigation. Through a transcriptome analysis, we identify IL-33 as an immune target upregulated in response to hypervirulent C. difficile. We demonstrate that IL-33 prevents C. difficile-associated mortality and epithelial disruption independently of bacterial burden or toxin expression. IL-33 drives colonic group 2 innate lymphoid cell (ILC2) activation during infection and IL-33 activated ILC2s are sufficient to prevent disease. Furthermore, intestinal IL-33 expression is regulated by the microbiota as fecal microbiota transplantation (FMT) rescues antibiotic-associated depletion of IL-33. Lastly, dysregulated IL-33 signaling via the decoy receptor, sST2, predicts C. difficile-associated mortality in human patients. Thus, IL-33 signaling to ILC2s is an important mechanism of defense from C. difficile colitis.


Asunto(s)
Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/inmunología , Inmunidad Innata , Interleucina-33/metabolismo , Linfocitos/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antibacterianos/efectos adversos , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidad , Colon/citología , Colon/inmunología , Colon/microbiología , Colon/patología , Modelos Animales de Enfermedad , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/mortalidad , Enterocolitis Seudomembranosa/terapia , Trasplante de Microbiota Fecal , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Perfilación de la Expresión Génica , Humanos , Interleucina-33/inmunología , Linfocitos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Virulencia/inmunología , Adulto Joven
12.
Science ; 365(6449)2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31296738

RESUMEN

To examine the contributions of impaired gut microbial community development to childhood undernutrition, we combined metabolomic and proteomic analyses of plasma samples with metagenomic analyses of fecal samples to characterize the biological state of Bangladeshi children with severe acute malnutrition (SAM) as they transitioned, after standard treatment, to moderate acute malnutrition (MAM) with persistent microbiota immaturity. Host and microbial effects of microbiota-directed complementary food (MDCF) prototypes targeting weaning-phase bacterial taxa underrepresented in SAM and MAM microbiota were characterized in gnotobiotic mice and gnotobiotic piglets colonized with age- and growth-discriminatory bacteria. A randomized, double-blind controlled feeding study identified a lead MDCF that changes the abundances of targeted bacteria and increases plasma biomarkers and mediators of growth, bone formation, neurodevelopment, and immune function in children with MAM.


Asunto(s)
Trastornos de la Nutrición del Niño/dietoterapia , Trastornos de la Nutrición del Niño/microbiología , Microbioma Gastrointestinal , Vida Libre de Gérmenes , Interacciones Microbiota-Huesped , Fenómenos Fisiológicos Nutricionales del Lactante , Animales , Bangladesh , Proteínas Sanguíneas/análisis , Trastornos de la Nutrición del Niño/metabolismo , Preescolar , Humanos , Lactante
13.
mBio ; 8(1)2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28246365

RESUMEN

The parasite Entamoeba histolytica is a cause of diarrhea in infants in low-income countries. Previously, it was shown that tumor necrosis factor alpha (TNF-α) production was associated with increased risk of E. histolytica diarrhea in children. Interleukin-25 (IL-25) is a cytokine that is produced by intestinal epithelial cells that has a role in maintenance of gut barrier function and inhibition of TNF-α production. IL-25 expression was decreased in humans and in the mouse model of amebic colitis. Repletion of IL-25 blocked E. histolytica infection and barrier disruption in mice, increased gut eosinophils, and suppressed colonic TNF-α. Depletion of eosinophils with anti-Siglec-F antibody prevented IL-25-mediated protection. In contrast, depletion of TNF-α resulted in resistance to amebic infection. We concluded that IL-25 provides protection from amebiasis, which is dependent upon intestinal eosinophils and suppression of TNF-α.IMPORTANCE The intestinal epithelial barrier is important for protection from intestinal amebiasis. We discovered that the intestinal epithelial cytokine IL-25 was suppressed during amebic colitis in humans and that protection could be restored in the mouse model by IL-25 administration. IL-25 acted via eosinophils and suppressed TNF-α. This work illustrates a previously unrecognized pathway of innate mucosal immune response.


Asunto(s)
Disentería Amebiana/inmunología , Entamoeba histolytica/inmunología , Eosinófilos/inmunología , Interleucina-17/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Interleucinas/metabolismo , Ratones
14.
Cell Rep ; 16(2): 432-443, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27346351

RESUMEN

Clostridium difficile infection (CDI) is the most common cause of hospital-acquired infection in the United States. Host susceptibility and the severity of infection are influenced by disruption of the microbiota and the immune response. However, how the microbiota regulate immune responses to mediate CDI outcome remains unclear. Here, we have investigated the role of the microbiota-linked cytokine IL-25 during infection. Intestinal IL-25 was suppressed during CDI in humans and mice. Restoration of IL-25 reduced CDI-associated mortality and tissue pathology even though equivalent levels of C. difficile bacteria and toxin remained in the gut. IL-25 protection was mediated by gut eosinophils, as demonstrated by an increase in intestinal eosinophils and a loss of IL-25 protection upon eosinophil depletion. These findings support a mechanism whereby the induction of IL-25-mediated eosinophilia can reduce host mortality during active CDI. This work may provide targets for future development of microbial or immune-based therapies.


Asunto(s)
Clostridioides difficile/inmunología , Enterocolitis Seudomembranosa/metabolismo , Eosinófilos/inmunología , Interleucinas/fisiología , Animales , Enterocolitis Seudomembranosa/inmunología , Enterocolitis Seudomembranosa/microbiología , Mucinas Gástricas/biosíntesis , Microbioma Gastrointestinal , Humanos , Interleucina-4/metabolismo , Interleucinas/farmacología , Recuento de Leucocitos , Masculino , Ratones Endogámicos C57BL , Membrana Mucosa/inmunología , Membrana Mucosa/patología , Factores Protectores
15.
Nat Microbiol ; 1(8): 16108, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27573114

RESUMEN

Clostridium difficile is the most common hospital acquired pathogen in the USA, and infection is, in many cases, fatal. Toxins A and B are its major virulence factors, but expression of a third toxin, known as C. difficile transferase (CDT), is increasingly common. An adenosine diphosphate (ADP)-ribosyltransferase that causes actin cytoskeletal disruption, CDT is typically produced by the major, hypervirulent strains and has been associated with more severe disease. Here, we show that CDT enhances the virulence of two PCR-ribotype 027 strains in mice. The toxin induces pathogenic host inflammation via a Toll-like receptor 2 (TLR2)-dependent pathway, resulting in the suppression of a protective host eosinophilic response. Finally, we show that restoration of TLR2-deficient eosinophils is sufficient for protection from a strain producing CDT. These findings offer an explanation for the enhanced virulence of CDT-expressing C. difficile and demonstrate a mechanism by which this binary toxin subverts the host immune response.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Proteínas Bacterianas/metabolismo , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/patología , Colon/inmunología , Eosinófilos/inmunología , Factores de Virulencia/metabolismo , Animales , Clostridioides difficile/clasificación , Clostridioides difficile/genética , Infecciones por Clostridium/microbiología , Modelos Animales de Enfermedad , Ratones , Ribotipificación
16.
PLoS Negl Trop Dis ; 10(7): e0004820, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27467505

RESUMEN

Cryptosporidium is a major cause of severe diarrhea, especially in malnourished children. Using a murine model of C. parvum oocyst challenge that recapitulates clinical features of severe cryptosporidiosis during malnutrition, we interrogated the effect of protein malnutrition (PM) on primary and secondary responses to C. parvum challenge, and tested the differential ability of mucosal priming strategies to overcome the PM-induced susceptibility. We determined that while PM fundamentally alters systemic and mucosal primary immune responses to Cryptosporidium, priming with C. parvum (106 oocysts) provides robust protective immunity against re-challenge despite ongoing PM. C. parvum priming restores mucosal Th1-type effectors (CD3+CD8+CD103+ T-cells) and cytokines (IFNγ, and IL12p40) that otherwise decrease with ongoing PM. Vaccination strategies with Cryptosporidium antigens expressed in the S. Typhi vector 908htr, however, do not enhance Th1-type responses to C. parvum challenge during PM, even though vaccination strongly boosts immunity in challenged fully nourished hosts. Remote non-specific exposures to the attenuated S. Typhi vector alone or the TLR9 agonist CpG ODN-1668 can partially attenuate C. parvum severity during PM, but neither as effectively as viable C. parvum priming. We conclude that although PM interferes with basal and vaccine-boosted immune responses to C. parvum, sustained reductions in disease severity are possible through mucosal activators of host defenses, and specifically C. parvum priming can elicit impressively robust Th1-type protective immunity despite ongoing protein malnutrition. These findings add insight into potential correlates of Cryptosporidium immunity and future vaccine strategies in malnourished children.


Asunto(s)
Criptosporidiosis/prevención & control , Cryptosporidium/inmunología , Proteínas en la Dieta/administración & dosificación , Desnutrición/patología , Vacunas Antiprotozoos/inmunología , Administración Intranasal , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Vacunas Antiprotozoos/administración & dosificación
17.
mBio ; 6(1)2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25626905

RESUMEN

UNLABELLED: Clostridium difficile is the most common hospital-acquired pathogen, causing antibiotic-associated diarrhea in over 250,000 patients annually in the United States. Disease is primarily mediated by toxins A and B, which induce potent proinflammatory signaling in host cells and can activate an ASC-containing inflammasome. Recent findings suggest that the intensity of the host response to infection correlates with disease severity. Our lab has identified the proinflammatory cytokine interleukin-23 (IL-23) as a pathogenic mediator during C. difficile infection (CDI). The mechanisms by which C. difficile induces IL-23, however, are not well understood, and the role of toxins A and B in this process is unclear. Here, we show that toxins A and B alone are not sufficient for IL-23 production but synergistically increase the amount of IL-23 produced in response to MyD88-dependent danger signals, including pathogen-associated molecular patterns (PAMPs) and host-derived damage associated molecular patterns (DAMPs). Danger signals also enhanced the secretion of IL-1ß in response to toxins A and B, and subsequent IL-1 receptor signaling accounted for the majority of the increase in IL-23 that occurred in the presence of the toxins. Inhibition of inflammasome activation in the presence of extracellular K(+) likewise decreased IL-23 production. Finally, we found that IL-1ß was increased in the serum of patients with CDI, suggesting that this systemic response could influence downstream production of pathogenic IL-23. Identification of the synergy of danger signals with toxins A and B via inflammasome signaling represents a novel finding in the mechanistic understanding of C. difficile-induced inflammation. IMPORTANCE: Clostridium difficile is among the leading causes of death due to health care-associated infection, and factors determining disease severity are not well understood. C. difficile secretes toxins A and B, which cause inflammation and tissue damage, and recent findings suggest that some of this tissue damage may be due to an inappropriate host immune response. We have found that toxins A and B, in combination with both bacterium- and host-derived danger signals, can induce expression of the proinflammatory cytokines IL-1ß and IL-23. Our results demonstrate that IL-1ß signaling enhances IL-23 production and could lead to increased pathogenic inflammation during CDI.


Asunto(s)
Clostridioides difficile/inmunología , Infecciones por Clostridium/inmunología , Inflamasomas/inmunología , Interleucina-23/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Clostridioides difficile/genética , Clostridioides difficile/fisiología , Infecciones por Clostridium/genética , Infecciones por Clostridium/microbiología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Enterotoxinas/inmunología , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-23/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA