Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Immunity ; 43(5): 840-2, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26588776

RESUMEN

Parasitic helminths are potent regulators of host immunity, including inhibition of allergic inflammation. In this issue of Immunity, Zaiss et al. (2015) reveal that microbiota compositional shifts during helminth infection contribute to the multifaceted ways that helminths modulate host immunity.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Helmintos/inmunología , Hipersensibilidad/inmunología , Inflamación/inmunología , Inflamación/parasitología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Femenino , Humanos , Masculino
2.
Infect Immun ; 89(12): e0022521, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34460289

RESUMEN

Heligmosomoides polygyrus is a helminth which naturally infects mice and is widely used as a laboratory model of chronic small intestinal helminth infection. While it is known that infection with H. polygyrus alters the composition of the host's bacterial microbiota, the functional implications of this alteration are unclear. We investigated the impact of H. polygyrus infection on short-chain fatty acid (SCFA) levels in the mouse intestine and sera. We found that helminth infection resulted in significantly upregulated levels of the branched SCFA isovaleric acid, exclusively in the proximal small intestine, which is the site of H. polygyrus colonization. We next set out to test the hypothesis that elevating local levels of isovaleric acid was a strategy used by H. polygyrus to promote its own fitness within the mammalian host. To test this, we supplemented the drinking water of mice with isovalerate during H. polygyrus infection and examined whether this affected helminth fecundity or chronicity. We did not find that isovaleric acid supplementation affected helminth chronicity; however, we found that it did promote helminth fecundity, as measured by helminth egg output in the feces of mice. Through antibiotic treatment of helminth-infected mice, we found that the bacterial microbiota was required in order to support elevated levels of isovaleric acid in the proximal small intestine during helminth infection. Overall, our data reveal that during H. polygyrus infection there is a microbiota-dependent localized increase in the production of isovaleric acid in the proximal small intestine and that this supports helminth fecundity in the murine host.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Interacciones Huésped-Parásitos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Nematospiroides dubius/fisiología , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología , Animales , Modelos Animales de Enfermedad , Metabolismo de los Lípidos , Ratones
3.
Eur J Immunol ; 48(1): 87-98, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28960280

RESUMEN

Dendritic cells (DCs) are essential in dictating the nature and effectiveness of immune responses. In the intestine DCs can be separated into discrete subsets, defined by expression of CD11b and CD103, each with different developmental requirements and distinct functional potential. Recent evidence has shown that different intestinal DC subsets are involved in the induction of T helper (Th)17 and regulatory T cell responses, but the cells that initiate Th2 immune responses are still incompletely understood. We show that in the Th2 response to an intestinal helminth in mice, only CD11b+ and not CD11b- DCs accumulate in the local lymph node, upregulate PDL2 and express markers of alternative activation. An enteric Th1 response instead activated both CD11b+ and CD11b- DCs without eliciting alternative activation in either population. Functionally, only CD11b+ DCs activated during helminth infection supported Th2 differentiation in naive CD4+ T cells. Together our data demonstrate that the ability to prime Th2 cells during intestinal helminth infection, is a selective and inducible characteristic of CD11b+ DCs.


Asunto(s)
Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Animales , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Células Dendríticas/clasificación , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Superficie Celular/inmunología , Infecciones por Strongylida/parasitología , Células TH1/inmunología
4.
Proc Natl Acad Sci U S A ; 112(2): 560-5, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548165

RESUMEN

Many significant bacterial pathogens have evolved virulence mechanisms to evade degradation and exposure to reactive oxygen (ROS) and reactive nitrogen species (RNS), allowing them to survive and replicate inside their hosts. Due to the highly reactive and short-lived nature of ROS and RNS, combined with limitations of conventional detection agents, the mechanisms underlying these evasion strategies remain poorly understood. In this study, we describe a system that uses redox-sensitive GFP to nondisruptively measure real-time fluctuations in the intrabacterial redox environment. Using this system coupled with high-throughput microscopy, we report the intrabacterial redox dynamics of Salmonella enterica Typhimurium (S. Typhimurium) residing inside macrophages. We found that the bacterial SPI-2 type III secretion system is required for ROS evasion strategies and this evasion relies on an intact Salmonella-containing vacuole (SCV) within which the bacteria reside during infection. Additionally, we found that cytosolic bacteria that escape the SCV experience increased redox stress in human and murine macrophages. These results highlight the existence of specialized evasion strategies used by intracellular pathogens that either reside inside a vacuole or "escape" into the cytosol. Taken together, the use of redox-sensitive GFP inside Salmonella significantly advances our understanding of ROS and RNS evasion strategies during infection. This technology can also be applied to measuring bacterial oxidative and nitrosative stress dynamics under different conditions in a wide variety of bacteria.


Asunto(s)
Macrófagos/metabolismo , Macrófagos/microbiología , Salmonella typhimurium/metabolismo , Animales , Línea Celular , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Ratones , Estrés Oxidativo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estallido Respiratorio , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Vacuolas/metabolismo , Vacuolas/microbiología
5.
J Infect Dis ; 215(8): 1245-1254, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28368463

RESUMEN

Intestinal helminth infections occur predominantly in regions where exposure to enteric bacterial pathogens is also common. Helminth infections inhibit host immunity against microbial pathogens, which has largely been attributed to the induction of regulatory or type 2 (Th2) immune responses. Here we demonstrate an additional 3-way interaction in which helminth infection alters the metabolic environment of the host intestine to enhance bacterial pathogenicity. We show that an ongoing helminth infection increased colonization by Salmonella independently of T regulatory or Th2 cells. Instead, helminth infection altered the metabolic profile of the intestine, which directly enhanced bacterial expression of Salmonella pathogenicity island 1 (SPI-1) genes and increased intracellular invasion. These data reveal a novel mechanism by which a helminth-modified metabolome promotes susceptibility to bacterial coinfection.


Asunto(s)
Coinfección/inmunología , Helmintiasis/inmunología , Parasitosis Intestinales/inmunología , Mucosa Intestinal/metabolismo , Metaboloma , Infecciones por Salmonella/inmunología , Células Th2/inmunología , Animales , Coinfección/microbiología , Coinfección/parasitología , Células HeLa , Humanos , Intestinos/microbiología , Intestinos/parasitología , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/genética
6.
PLoS Pathog ; 11(3): e1004676, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25816012

RESUMEN

Over 25% of the world's population are infected with helminth parasites, the majority of which colonise the gastrointestinal tract. However, no vaccine is yet available for human use, and mechanisms of protective immunity remain unclear. In the mouse model of Heligmosomoides polygyrus infection, vaccination with excretory-secretory (HES) antigens from adult parasites elicits sterilising immunity. Notably, three purified HES antigens (VAL-1, -2 and -3) are sufficient for effective vaccination. Protection is fully dependent upon specific IgG1 antibodies, but passive transfer confers only partial immunity to infection, indicating that cellular components are also required. Moreover, immune mice show greater cellular infiltration associated with trapping of larvae in the gut wall prior to their maturation. Intra-vital imaging of infected intestinal tissue revealed a four-fold increase in extravasation by LysM+GFP+ myeloid cells in vaccinated mice, and the massing of these cells around immature larvae. Mice deficient in FcRγ chain or C3 complement component remain fully immune, suggesting that in the presence of antibodies that directly neutralise parasite molecules, the myeloid compartment may attack larvae more quickly and effectively. Immunity to challenge infection was compromised in IL-4Rα- and IL-25-deficient mice, despite levels of specific antibody comparable to immune wild-type controls, while deficiencies in basophils, eosinophils or mast cells or CCR2-dependent inflammatory monocytes did not diminish immunity. Finally, we identify a suite of previously uncharacterised heat-labile vaccine antigens with homologs in human and veterinary parasites that together promote full immunity. Taken together, these data indicate that vaccine-induced immunity to intestinal helminths involves IgG1 antibodies directed against secreted proteins acting in concert with IL-25-dependent Type 2 myeloid effector populations.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Antígenos Helmínticos/inmunología , Inmunoglobulina G/inmunología , Interleucina-4/inmunología , Interleucinas/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Vacunación , Animales , Anticuerpos Antihelmínticos/genética , Humanos , Inmunoglobulina G/genética , Interleucina-4/genética , Interleucinas/genética , Larva/inmunología , Ratones , Ratones Noqueados , Infecciones por Strongylida/genética , Infecciones por Strongylida/prevención & control
7.
J Immunol ; 195(9): 4059-66, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26477048

RESUMEN

Both intestinal helminth parasites and certain bacterial microbiota species have been credited with strong immunomodulatory effects. Recent studies reported that the presence of helminth infection alters the composition of the bacterial intestinal microbiota and, conversely, that the presence and composition of the bacterial microbiota affect helminth colonization and persistence within mammalian hosts. This article reviews recent findings on these reciprocal relationships, in both human populations and mouse models, at the level of potential mechanistic pathways and the implications these bear for immunomodulatory effects on allergic and autoimmune disorders. Understanding the multidirectional complex interactions among intestinal microbes, helminth parasites, and the host immune system allows for a more holistic approach when using probiotics, prebiotics, synbiotics, antibiotics, and anthelmintics, as well as when designing treatments for autoimmune and allergic conditions.


Asunto(s)
Infecciones Bacterianas/inmunología , Helmintiasis/inmunología , Helmintiasis/microbiología , Interacciones Huésped-Patógeno , Intestinos/microbiología , Intestinos/parasitología , Microbiota , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Homeostasis , Humanos , Ratones , Receptores Toll-Like/fisiología
8.
J Immunol ; 193(6): 2984-93, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25114104

RESUMEN

Helminth parasites remain one of the most common causes of infections worldwide, yet little is still known about the immune signaling pathways that control their expulsion. C57BL/6 mice are chronically susceptible to infection with the gastrointestinal helminth parasite Heligmosomoides polygyrus. In this article, we report that C57BL/6 mice lacking the adapter protein MyD88, which mediates signaling by TLRs and IL-1 family members, showed enhanced immunity to H. polygyrus infection. Alongside increased parasite expulsion, MyD88-deficient mice showed heightened IL-4 and IL-17A production from mesenteric lymph node CD4(+) cells. In addition, MyD88(-/-) mice developed substantial numbers of intestinal granulomas around the site of infection, which were not seen in MyD88-sufficient C57BL/6 mice, nor when signaling through the adapter protein TRIF (TIR domain-containing adapter-inducing IFN-ß adapter protein) was also ablated. Mice deficient solely in TLR2, TLR4, TLR5, or TLR9 did not show enhanced parasite expulsion, suggesting that these TLRs signal redundantly to maintain H. polygyrus susceptibility in wild-type mice. To further investigate signaling pathways that are MyD88 dependent, we infected IL-1R1(-/-) mice with H. polygyrus. This genotype displayed heightened granuloma numbers compared with wild-type mice, but without increased parasite expulsion. Thus, the IL-1R-MyD88 pathway is implicated in inhibiting granuloma formation; however, protective immunity in MyD88-deficient mice appears to be granuloma independent. Like IL-1R1(-/-) and MyD88(-/-) mice, animals lacking signaling through the type 1 IFN receptor (i.e., IFNAR1(-/-)) also developed intestinal granulomas. Hence, IL-1R1, MyD88, and type 1 IFN receptor signaling may provide pathways to impede granuloma formation in vivo, but additional MyD88-mediated signals are associated with inhibition of protective immunity in susceptible C57BL/6 mice.


Asunto(s)
Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Granuloma/genética , Granuloma/inmunología , Interleucina-17/biosíntesis , Interleucina-4/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptores Tipo I de Interleucina-1/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Infecciones por Strongylida/parasitología , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 5/genética , Receptor Toll-Like 9/genética
9.
J Allergy Clin Immunol ; 135(1): 100-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25145536

RESUMEN

BACKGROUND: Resident gut microbiota are now recognized as potent modifiers of host immune responses in various scenarios. Recently, we demonstrated that perinatal exposure to vancomycin, but not streptomycin, profoundly alters gut microbiota and enhances susceptibility to a TH2 model of allergic asthma. OBJECTIVE: Here we sought to further clarify the etiology of these changes by determining whether perinatal antibiotic treatment has a similar effect on the TH1/TH17-mediated lung disease, hypersensitivity pneumonitis. METHODS: Hypersensitivity pneumonitis was induced in C57BL/6 wild-type or recombination-activating gene 1-deficient mice treated perinatally with vancomycin or streptomycin by repeated intranasal administration of Saccharopolyspora rectivirgula antigen. Disease severity was assessed by measuring lung inflammation, pathology, cytokine responses, and serum antibodies. Microbial community analyses were performed on stool samples via 16S ribosomal RNA pyrosequencing and correlations between disease severity and specific bacterial taxa were identified. RESULTS: Surprisingly, in contrast to our findings in an allergic asthma model, we found that the severity of hypersensitivity pneumonitis was unaffected by vancomycin, but increased dramatically after streptomycin treatment. This likely reflects an effect on the adaptive, rather than innate, immune response because the effects of streptomycin were not observed during the early phases of disease and were abrogated in recombination-activating gene 1-deficient mice. Interestingly, Bacteroidetes dominated the intestinal microbiota of streptomycin-treated animals, while vancomycin promoted the expansion of the Firmicutes. CONCLUSIONS: Perinatal antibiotics exert highly selective effects on resident gut flora, which, in turn, lead to very specific alterations in susceptibility to TH2- or TH1/TH17-driven lung inflammatory disease.


Asunto(s)
Alveolitis Alérgica Extrínseca/inmunología , Alveolitis Alérgica Extrínseca/microbiología , Antibacterianos/efectos adversos , Tracto Gastrointestinal/microbiología , Microbiota , Estreptomicina/efectos adversos , Alveolitis Alérgica Extrínseca/sangre , Alveolitis Alérgica Extrínseca/patología , Animales , Animales Recién Nacidos , Citocinas/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones Endogámicos C57BL , Saccharopolyspora , Índice de Severidad de la Enfermedad , Vancomicina/farmacología
10.
J Immunol ; 189(3): 1113-7, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22753928

RESUMEN

Multiple factors control susceptibility of C57BL/6 mice to infection with the helminth Heligmosomoides polygyrus, including TGF-ß signaling, which inhibits immunity in vivo. However, mice expressing a T cell-specific dominant-negative TGF-ß receptor II (TGF-ßRII DN) show dampened Th2 immunity and diminished resistance to infection. Interestingly, H. polygyrus-infected TGF-ßRII DN mice show greater frequencies of CD4(+)Foxp3(+)Helios(+) Tregs than infected wild-type mice, but levels of CD103 are greatly reduced on both these cells and on the CD4(+)Foxp3(+)Helios(-) population. Although Th9 and Th17 levels are comparable between infected TGF-ßRII DN and wild-type mice, the former develop exaggerated CD4(+) and CD8(+) T cell IFN-γ responses. Increased susceptibility conferred by TGF-ßRII DN expression was lost in IFN-γ-deficient mice, although they remained unable to completely clear infection. Hence, overexpression of IFN-γ negatively modulates immunity, and the presence of Helios(+) Tregs may maintain susceptibility on the C57BL/6 background.


Asunto(s)
Antígenos CD/biosíntesis , Diferenciación Celular/inmunología , Cadenas alfa de Integrinas/biosíntesis , Interferón gamma/biosíntesis , Nematospiroides dubius , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/genética , Animales , Diferenciación Celular/genética , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Predisposición Genética a la Enfermedad , Interferón gamma/deficiencia , Interferón gamma/genética , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Nematospiroides dubius/inmunología , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/genética , Infecciones por Strongylida/genética , Linfocitos T Reguladores/parasitología , Linfocitos T Reguladores/patología , Factor de Crecimiento Transformador beta/fisiología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
11.
Front Microbiol ; 14: 1254342, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37795301

RESUMEN

Introduction: Syphilis is a chronic, multi-stage infection caused by the extracellular bacterium Treponema pallidum ssp. pallidum. Treponema pallidum widely disseminates through the vasculature, crosses endothelial, blood-brain and placental barriers, and establishes systemic infection. Although the capacity of T. pallidum to traverse the endothelium is well-described, the response of endothelial cells to T. pallidum exposure, and the contribution of this response to treponemal traversal, is poorly understood. Methods: To address this knowledge gap, we used quantitative proteomics and cytokine profiling to characterize endothelial responses to T. pallidum. Results: Proteomic analyses detected altered host pathways controlling extracellular matrix organization, necroptosis and cell death, and innate immune signaling. Cytokine analyses of endothelial cells exposed to T. pallidum revealed increased secretion of interleukin (IL)-6, IL-8, and vascular endothelial growth factor (VEGF), and decreased secretion of monocyte chemoattractant protein-1 (MCP-1). Discussion: This study provides insight into the molecular basis of syphilis disease symptoms and the enhanced susceptibility of individuals infected with syphilis to HIV co-infection. These investigations also enhance understanding of the host response to T. pallidum exposure and the pathogenic strategies used by T. pallidum to disseminate and persist within the host. Furthermore, our findings highlight the critical need for inclusion of appropriate controls when conducting T. pallidum-host cell interactions using in vitro- and in vivo-grown T. pallidum.

12.
J Leukoc Biol ; 114(3): 223-236, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37227004

RESUMEN

Eosinophils are present in the thymus of mammals, yet their function at this site during homeostatic development is unknown. We used flow cytometry to determine the abundance and phenotype of eosinophils (here defined as SSchigh SiglecF+ CD11b+ CD45+ cells) in the thymus of mice during the neonatal period, the later postnatal period, and into adulthood. We show that both the total number of thymic eosinophils and their frequency among leukocytes increase over the first 2 wk of life and that their accumulation in the thymus is dependent on the presence of an intact bacterial microbiota. We report that thymic eosinophils express the interleukin-5 receptor (CD125), CD80, and IDO, and that subsets of thymic eosinophils express CD11c and major histocompatibility complex II (MHCII). We found that the frequency of MHCII-expressing thymic eosinophils increases over the first 2 wk of life, and that during this early-life period the highest frequency of MHCII-expressing thymic eosinophils is located in the inner medullary region. These data suggest a temporal and microbiota-dependent regulation of eosinophil abundance and functional capabilities in the thymus.


Asunto(s)
Eosinófilos , Timo , Ratones , Animales , Citometría de Flujo , Complejo Mayor de Histocompatibilidad , Mamíferos
13.
Exp Parasitol ; 132(1): 76-89, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21875581

RESUMEN

The intestinal nematode parasite Heligmosomoides polygyrus bakeri exerts widespread immunomodulatory effects on both the innate and adaptive immune system of the host. Infected mice adopt an immunoregulated phenotype, with abated allergic and autoimmune reactions. At the cellular level, infection is accompanied by expanded regulatory T cell populations, skewed dendritic cell and macrophage phenotypes, B cell hyperstimulation and multiple localised changes within the intestinal environment. In most mouse strains, these act to block protective Th2 immunity. The molecular basis of parasite interactions with the host immune system centres upon secreted products termed HES (H. polygyrus excretory-secretory antigen), which include a TGF-ß-like ligand that induces de novo regulatory T cells, factors that modify innate inflammatory responses, and molecules that block allergy in vivo. Proteomic and transcriptomic definition of parasite proteins, combined with biochemical identification of immunogenic molecules in resistant mice, will provide new candidate immunomodulators and vaccine antigens for future research.


Asunto(s)
Inmunomodulación , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Inmunidad Adaptativa/inmunología , Animales , Anticuerpos Antihelmínticos/inmunología , Especificidad de Anticuerpos , Antígenos Helmínticos/inmunología , Linfocitos B/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Humanos , Inmunidad Innata/inmunología , Ratones , Fenotipo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
14.
Front Microbiol ; 13: 888525, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35722306

RESUMEN

The etiological agent of syphilis, Treponema pallidum ssp. pallidum, is a highly invasive "stealth" pathogen that can evade the host immune response and persist within the host for decades. This obligate human pathogen is adept at establishing infection and surviving at sites within the host that have a multitude of competing microbes, sometimes including pathogens. One survival strategy employed by bacteria found at polymicrobial sites is elimination of competing microorganisms by production of antimicrobial peptides (AMPs). Antimicrobial peptides are low molecular weight proteins (miniproteins) that function directly via inhibition and killing of microbes and/or indirectly via modulation of the host immune response, which can facilitate immune evasion. In the current study, we used bioinformatics to show that approximately 7% of the T. pallidum proteome is comprised of miniproteins of 150 amino acids or less with unknown functions. To investigate the possibility that AMP production is an unrecognized defense strategy used by T. pallidum during infection, we developed a bioinformatics pipeline to analyze the complement of T. pallidum miniproteins of unknown function for the identification of potential AMPs. This analysis identified 45 T. pallidum AMP candidates; of these, Tp0451a and Tp0749 were subjected to further bioinformatic analyses to identify AMP critical core regions (AMPCCRs). Four potential AMPCCRs from the two predicted AMPs were identified and peptides corresponding to these AMPCCRs were experimentally confirmed to exhibit bacteriostatic and bactericidal activity against a panel of biologically relevant Gram-positive and Gram-negative bacteria. Immunomodulation assays performed under inflammatory conditions demonstrated that one of the AMPCCRs was also capable of differentially regulating expression of two pro-inflammatory chemokines [monocyte chemoattractant protein-1 (MCP-1) and interleukin-8 (IL-8)]. These findings demonstrate proof-of-concept for our developed AMP identification pipeline and are consistent with the novel concept that T. pallidum expresses AMPs to defend against competing microbes and modulate the host immune response.

15.
PLoS Negl Trop Dis ; 15(1): e0009052, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33471793

RESUMEN

Intestinal helminth infection can impair host resistance to co-infection with enteric bacterial pathogens. However, it is not known whether helminth drug-clearance can restore host resistance to bacterial infection. Using a mouse helminth-Salmonella co-infection system, we show that anthelmintic treatment prior to Salmonella challenge is sufficient to restore host resistance to Salmonella. The presence of the small intestine-dwelling helminth Heligmosomoides polygyrus at the point of Salmonella infection supports the initial establishment of Salmonella in the small intestinal lumen. Interestingly, if helminth drug-clearance is delayed until Salmonella has already established in the small intestinal lumen, anthelmintic treatment does not result in complete clearance of Salmonella. This suggests that while the presence of helminths supports initial Salmonella colonization, helminths are dispensable for Salmonella persistence in the host small intestine. These data contribute to the mechanistic understanding of how an ongoing or prior helminth infection can affect pathogenic bacterial colonization and persistence in the mammalian intestine.


Asunto(s)
Coinfección/microbiología , Coinfección/parasitología , Resistencia a la Enfermedad/fisiología , Helmintiasis/complicaciones , Parasitosis Intestinales/complicaciones , Nematospiroides dubius/fisiología , Salmonella/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Intestinos/microbiología , Intestinos/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Salmonella/complicaciones , Salmonella typhi
16.
ACS Biomater Sci Eng ; 6(9): 5069-5083, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-33455300

RESUMEN

We demonstrate microfluidic manufacturing of glutathione (GSH)-responsive polymer nanoparticles (PNPs) with controlled in vitro pharmacological properties for selective drug delivery. This work leverages previous fundamental work on microfluidic control of the physicochemical properties of GSH-responsive PNPs containing cleavable disulfide groups in two different locations (core and interface, DualM PNPs). In this paper, we employ a two-phase gas-liquid microfluidic reactor for the flow-directed manufacturing of paclitaxel-loaded or DiI-loaded DualM PNPs (PAX-PNPs or DiI-PNPs, where DiI is a fluorescent drug surrogate dye). We find that both PAX-PNPs and DiI-PNPs exhibit similar flow-tunable sizes, morphologies, and internal structures to those previously described for empty DualM PNPs. Fluorescent imaging of DiI-PNP formulations shows that microfluidic manufacturing greatly improves the homogeneity of drug dispersion within the PNP population compared to standard bulk microprecipitation. Encapsulation of PAX in DualM PNPs significantly increases its selectivity to cancerous cells, with various PAX-PNP formulations showing higher cytotoxicity against cancerous MCF-7 cells than against non-cancerous HaCaT cells, in contrast to free PAX, which showed similar cytotoxicity in the two cell lines. In addition, the characterization of DualM PNP formulations formed at various microfluidic flow rates reveals that critical figures of merit for drug delivery function-including encapsulation efficiencies, GSH-triggered release rates, rates of cell uptake, cytotoxicities, and selectivity to cancerous cells-exhibit microfluidic flow tunability that mirrors trends in PNP size. These results highlight the potential of two-phase microfluidic manufacturing for controlling both structure and drug delivery function of biological stimuli-responsive nanomedicines toward improved therapeutic outcomes.


Asunto(s)
Nanopartículas , Preparaciones Farmacéuticas , Polímeros de Estímulo Receptivo , Sistemas de Liberación de Medicamentos , Humanos , Microfluídica
17.
Front Immunol ; 11: 557960, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178185

RESUMEN

Conflicting data has emerged regarding a role for eosinophils in IgA production, with some reports that eosinophils support both secretory and circulating IgA levels during homeostasis. Previous studies have compared antibody levels between wildtype and eosinophil-deficient mice, but these mice were obtained from different commercial vendors and/or were not littermates. Thus, the possibility remains that extrinsic environmental factors, rather than an intrinsic lack of eosinophils, are responsible for the reports of reduced IgA in eosinophil-deficient mice. Here we used wild-type and eosinophil-deficient (ΔdblGATA) mice that were purchased from a single vendor, subsequently bred in-house and either co-housed as adults, co-reared from birth or raised as littermates. We found no differences in the levels of secretory IgA or in the numbers of small intestinal IgA-producing plasma cells between wild-type and ΔdblGATA mice, demonstrating that under controlled steady-state conditions eosinophils are not essential for the maintenance of secretory IgA in the intestinal tract. While we found that levels of IgM and IgE were significantly elevated in the serum of ΔdblGATA mice compared to co-reared or co-housed wild-type mice, no significant differences in these or other circulating antibody isotypes were identified between genotypes in littermate-controlled experiments. Our results demonstrate that eosinophils are not required to maintain secretory or circulating IgA production and the absence of eosinophils does not impact circulating IgG1, IgG2b, IgM, or IgE levels during homeostasis. These findings emphasize the importance of optimally controlling rearing and housing conditions throughout life between mice of different genotypes.


Asunto(s)
Eosinófilos/inmunología , Eosinófilos/metabolismo , Inmunoglobulina A/inmunología , Animales , Biomarcadores , Citocinas/metabolismo , Citometría de Flujo , Inmunoglobulina A/sangre , Inmunoglobulina A Secretora/inmunología , Ratones , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo
18.
Mucosal Immunol ; 11(4): 1039-1046, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29453411

RESUMEN

Intestinal helminths have well-characterized modulatory effects on mammalian immune pathways. Ongoing helminth infection has been associated with both the suppression of allergies and an altered susceptibility to microbial infections. Enteric helminths share a niche with the intestinal microbiota, and the presence of helminths alters the microbiota composition and the metabolic signature of the host. Recent studies have demonstrated that the helminth-modified intestinal microbiome has the capacity to modify host immune responses even in the absence of live helminth infection. This article discusses the mechanisms by which helminths modify the intestinal microbiome of mammals, and reviews the evidence for a helminth-modified microbiome directly influencing host immunity during infectious and inflammatory diseases. Understanding the multifaceted mechanisms that underpin helminth immunomodulation will pave the way for novel therapies to combat infectious and inflammatory diseases.


Asunto(s)
Helmintiasis/inmunología , Helmintos/inmunología , Infecciones/inmunología , Inflamación/inmunología , Intestinos/inmunología , Microbiota , Animales , Terapia Biológica/tendencias , Inmunidad Innata , Inmunomodulación , Intestinos/microbiología , Intestinos/parasitología
19.
Nat Rev Immunol ; 17(8): 518-528, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28504257

RESUMEN

The incidence of allergic disease continues to rise in industrialized countries. The rapid increase in the incidence of allergic disease throughout the past half century suggests that recently altered environmental factors are driving allergy development. Accumulating evidence suggests that environmental experiences that occur during the first months of life can influence the risk of allergic sensitization. In this Review, we present the evidence relating to specific early life exposures that affect future allergy development, and discuss how these exposures may promote either tolerance or allergic sensitization.


Asunto(s)
Hipersensibilidad/inmunología , Animales , Asma/inmunología , Asma/microbiología , Asma/terapia , Desarrollo Infantil , Modelos Animales de Enfermedad , Helmintos , Humanos , Hipersensibilidad/microbiología , Hipersensibilidad/terapia , Recién Nacido , Infecciones/inmunología
20.
Data Brief ; 7: 894-9, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27077092

RESUMEN

The redox balance in a variety of Gram-negative bacteria was explored using redox sensitive GFP (roGFP2), J. van der Heijden et al. doi:10.1016/j.freeradbiomed.2015.11.029[1]. This data article provides Supporting material to further investigate the relationship between Salmonella typhimurium survival and oxidative stress. The first set of data presented in this article, shows the percentage of surviving bacteria after exposure to hydrogen peroxide. The second set of data shows the concentration of hydrogen peroxide that was produced by S. Typhimurium in different growth phases. The last set of data shows the percentage of surviving S. Typhimurium bacteria after exposure to different antibiotics.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA