Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
1.
Immunol Cell Biol ; 102(5): 396-406, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38648862

RESUMEN

Increased permeability of the intestinal epithelial layer is linked to the pathogenesis and perpetuation of a wide range of intestinal and extra-intestinal diseases. Infecting humans with controlled doses of helminths, such as human hookworm (termed hookworm therapy), is proposed as a treatment for many of the same diseases. Helminths induce immunoregulatory changes in their host which could decrease epithelial permeability, which is highlighted as a potential mechanism through which helminths treat disease. Despite this, the influence of a chronic helminth infection on epithelial permeability remains unclear. This study uses the chronically infecting intestinal helminth Heligmosomoides polygyrus to reveal alterations in the expression of intestinal tight junction proteins and epithelial permeability during the infection course. In the acute infection phase (1 week postinfection), an increase in intestinal epithelial permeability is observed. Consistent with this finding, jejunal claudin-2 is upregulated and tricellulin is downregulated. By contrast, in the chronic infection phase (6 weeks postinfection), colonic claudin-1 is upregulated and epithelial permeability decreases. Importantly, this study also investigates changes in epithelial permeability in a small human cohort experimentally challenged with the human hookworm, Necator americanus. It demonstrates a trend toward small intestinal permeability increasing in the acute infection phase (8 weeks postinfection), and colonic and whole gut permeability decreasing in the chronic infection phase (24 weeks postinfection), suggesting a conserved epithelial response between humans and mice. In summary, our findings demonstrate dynamic changes in epithelial permeability during a chronic helminth infection and provide another plausible mechanism by which chronic helminth infections could be utilized to treat disease.


Asunto(s)
Mucosa Intestinal , Permeabilidad , Animales , Humanos , Mucosa Intestinal/parasitología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Enfermedad Crónica , Nematospiroides dubius/inmunología , Ratones , Necator americanus , Parasitosis Intestinales/inmunología , Uniones Estrechas/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Intestino Delgado/parasitología , Intestino Delgado/inmunología , Femenino , Ratones Endogámicos C57BL , Masculino , Helmintiasis/inmunología , Helmintiasis/parasitología , Necatoriasis/inmunología , Proteína 2 con Dominio MARVEL/metabolismo
2.
Nature ; 607(7917): 142-148, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35732734

RESUMEN

The divergence of the common dendritic cell progenitor1-3 (CDP) into the conventional type 1 and type 2 dendritic cell (cDC1 and cDC2, respectively) lineages4,5 is poorly understood. Some transcription factors act in the commitment of already specified progenitors-such as BATF3, which stabilizes Irf8 autoactivation at the +32 kb Irf8 enhancer4,6-but the mechanisms controlling the initial divergence of CDPs remain unknown. Here we report the transcriptional basis of CDP divergence and describe the first requirements for pre-cDC2 specification. Genetic epistasis analysis7 suggested that Nfil3 acts upstream of Id2, Batf3 and Zeb2 in cDC1 development but did not reveal its mechanism or targets. Analysis of newly generated NFIL3 reporter mice showed extremely transient NFIL3 expression during cDC1 specification. CUT&RUN and chromatin immunoprecipitation followed by sequencing identified endogenous NFIL3 binding in the -165 kb Zeb2 enhancer8 at three sites that also bind the CCAAT-enhancer-binding proteins C/EBPα and C/EBPß. In vivo mutational analysis using CRISPR-Cas9 targeting showed that these NFIL3-C/EBP sites are functionally redundant, with C/EBPs supporting and NFIL3 repressing Zeb2 expression at these sites. A triple mutation of all three NFIL3-C/EBP sites ablated Zeb2 expression in myeloid, but not lymphoid progenitors, causing the complete loss of pre-cDC2 specification and mature cDC2 development in vivo. These mice did not generate T helper 2 (TH2) cell responses against Heligmosomoides polygyrus infection, consistent with cDC2 supporting TH2 responses to helminths9-11. Thus, CDP divergence into cDC1 or cDC2 is controlled by competition between NFIL3 and C/EBPs at the -165 kb Zeb2 enhancer.


Asunto(s)
Diferenciación Celular , Células Dendríticas , Elementos de Facilitación Genéticos , Mutación , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/genética , Células Dendríticas/clasificación , Células Dendríticas/citología , Células Dendríticas/patología , Elementos de Facilitación Genéticos/genética , Epistasis Genética , Proteína 2 Inhibidora de la Diferenciación , Linfocitos/citología , Ratones , Células Mieloides/citología , Nematospiroides dubius/inmunología , Proteínas Represoras , Células Th2/citología , Células Th2/inmunología , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética
3.
J Biol Chem ; 298(6): 101994, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35500648

RESUMEN

The mouse intestinal helminth Heligmosomoides polygyrus modulates host immune responses by secreting a transforming growth factor (TGF)-ß mimic (TGM), to expand the population of Foxp3+ Tregs. TGM comprises five complement control protein (CCP)-like domains, designated D1-D5. Though lacking homology to TGF-ß, TGM binds directly to the TGF-ß receptors TßRI and TßRII and stimulates the differentiation of naïve T-cells into Tregs. However, the molecular determinants of binding are unclear. Here, we used surface plasmon resonance, isothermal calorimetry, NMR spectroscopy, and mutagenesis to investigate how TGM binds the TGF-ß receptors. We demonstrate that binding is modular, with D1-D2 binding to TßRI and D3 binding to TßRII. D1-D2 and D3 were further shown to compete with TGF-ß(TßRII)2 and TGF-ß for binding to TßRI and TßRII, respectively. The solution structure of TGM-D3 revealed that TGM adopts a CCP-like fold but is also modified to allow the C-terminal strand to diverge, leading to an expansion of the domain and opening potential interaction surfaces. TGM-D3 also incorporates a long structurally ordered hypervariable loop, adding further potential interaction sites. Through NMR shift perturbations and binding studies of TGM-D3 and TßRII variants, TGM-D3 was shown to occupy the same site of TßRII as bound by TGF-ß using both a novel interaction surface and the hypervariable loop. These results, together with the identification of other secreted CCP-like proteins with immunomodulatory activity in H. polygyrus, suggest that TGM is part of a larger family of evolutionarily plastic parasite effector molecules that mediate novel interactions with their host.


Asunto(s)
Proteínas del Helminto , Interacciones Huésped-Patógeno , Nematospiroides dubius , Receptores de Factores de Crecimiento Transformadores beta , Factor de Crecimiento Transformador beta , Animales , Evolución Biológica , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Interacciones Huésped-Patógeno/inmunología , Ratones , Nematospiroides dubius/clasificación , Nematospiroides dubius/genética , Nematospiroides dubius/inmunología , Nematospiroides dubius/metabolismo , Unión Proteica , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
Nature ; 599(7883): 125-130, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34671159

RESUMEN

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Asunto(s)
Homeostasis , Intestinos/inmunología , Intestinos/fisiología , Neuroglía/inmunología , Neuroglía/fisiología , Regeneración , Adventicia/inmunología , Adventicia/parasitología , Animales , Quimiocina CXCL10/inmunología , Duodeno/inmunología , Duodeno/parasitología , Duodeno/patología , Duodeno/fisiología , Femenino , Gliosis , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Intestinos/parasitología , Intestinos/patología , Masculino , Ratones , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología
5.
Indian J Tuberc ; 68(3): 340-349, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34099199

RESUMEN

BACKGROUND: Tuberculosis is a chronic infection caused by Mycobacterium tuberculosis (M.tb), which needs proper macrophage activation for control. It has been debated whether the co-infection with helminth will affect the immune response to mycobacterial infection. OBJECTIVE: To determine the effect of sequential co-infection of Heligmosomoides polygyrus (H.pg) nematodes and M.tb on T cell responses, macrophages polarization and lung histopathological changes. METHOD: This study used 49 mice divided into 7 treatment groups, with different sequence of infection of M.tb via inhalation and H.pg via oral ingestion for 8 and 16 weeks. T cells response in the lung, intestine, and peripheral blood were determined by flow cytometry. Cytokines (IL-4, IFN-γ, TGB-ß1, and IL-10) were measured in peripheral blood using ELISA. Lung macrophage polarization were determined by the expression of iNOS (M1) or Arginase 1 (M2). Mycobacterial count were done in lung tissue. Lung histopathology were measured using Dorman's semiquantitative score assessing peribronchiolitis, perivasculitis, alveolitis, and granuloma formation. RESULT: M.tb infection induced Th1 response and M1 macrophage polarization, while H.pg infection induced Th2 and M2 polarization. In sequential co-infection, the final polarization of macrophage was dictated by the sequence of co-infection. However, all groups with M.tb infection showed the same degree of mycobacterial count in lung tissues and lung tissue histopathological changes. CONCLUSION: Sequential co-infection of H.pg and M.tb induces different T cell response which leads to different macrophage polarization in lung tissue. Helminth infection induced M2 lung macrophage polarization, but did not cause different mycobacterial count nor lung histopathological changes.


Asunto(s)
Pulmón , Activación de Macrófagos/inmunología , Macrófagos Alveolares/inmunología , Mycobacterium tuberculosis/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida , Tuberculosis , Animales , Recuento de Células , Polaridad Celular/inmunología , Coinfección/inmunología , Coinfección/microbiología , Coinfección/parasitología , Citocinas/sangre , Modelos Animales de Enfermedad , Inmunidad Celular , Pulmón/inmunología , Pulmón/patología , Ratones , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Linfocitos T/inmunología , Tuberculosis/inmunología , Tuberculosis/microbiología
6.
Front Immunol ; 12: 627638, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33936040

RESUMEN

Background: Tuberculosis (TB) is still a major challenge for humankind. Because regions with the highest incidence also have a high prevalence of helminthiasis and nutritional scarcity, we wanted to understand the impact of these on TB progression. Methods: We have developed an experimental murine model for active TB in C3HeB/FeJ, coinfected with Trichuris muris and Heligmosomoides polygyrus nematodes, and exposed to an environmental mycobacterium (M. manresensis) and intermittent fasting. Cause-effect relationships among these factors were explored with Partial Least Squares Path modelling (PLSPM). Results: Previous parasitization had a major anti-inflammatory effect and reduced systemic levels of ADA, haptoglobin, local pulmonary levels of IL-1ß, IL-6, TNF-α, CXCL-1, CXCL-5 and IL-10. Oral administration of heat-killed M. manresensis resulted in a similar outcome. Both interventions diminished pulmonary pathology and bacillary load, but intermittent food deprivation reduced this protective effect increasing stress and inflammation. The PLSPM revealed nematodes might have protective effects against TB progression. Conclusions: Significantly higher cortisol levels in food-deprivation groups showed it is a stressful condition, which might explain its deleterious effect. This highlights the impact of food security on TB eradication policies and the need to prioritize food supply over deworming activities.


Asunto(s)
Coinfección , Privación de Alimentos , Helmintiasis/parasitología , Parasitosis Intestinales/parasitología , Pulmón/microbiología , Mycobacterium tuberculosis/patogenicidad , Nematospiroides dubius/patogenicidad , Infecciones por Strongylida/parasitología , Tricuriasis/parasitología , Trichuris/patogenicidad , Tuberculosis Pulmonar/microbiología , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Helmintiasis/inmunología , Helmintiasis/metabolismo , Interacciones Huésped-Parásitos , Mediadores de Inflamación/metabolismo , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Ratones Endogámicos C3H , Mycobacterium tuberculosis/inmunología , Nematospiroides dubius/inmunología , Estado Nutricional , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/metabolismo , Tricuriasis/inmunología , Tricuriasis/metabolismo , Trichuris/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/metabolismo
7.
Nat Immunol ; 22(2): 216-228, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33462454

RESUMEN

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Asunto(s)
Bacterias/patogenicidad , Infecciones Bacterianas/microbiología , Linfocitos T CD4-Positivos/microbiología , Linfocitos T CD4-Positivos/parasitología , Colon/microbiología , Colon/parasitología , Microbioma Gastrointestinal , Heligmosomatoidea/patogenicidad , Parasitosis Intestinales/parasitología , Animales , Bacterias/inmunología , Infecciones Bacterianas/genética , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Cromatina/genética , Cromatina/metabolismo , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Colon/inmunología , Colon/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Heligmosomatoidea/inmunología , Interacciones Huésped-Patógeno , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Parasitosis Intestinales/genética , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Nippostrongylus/inmunología , Nippostrongylus/patogenicidad , Fenotipo , Salmonella enterica/inmunología , Salmonella enterica/patogenicidad , Análisis de la Célula Individual , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transcriptoma
8.
Eur J Immunol ; 51(2): 433-444, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33067820

RESUMEN

Infection with parasitic worms (helminths) alters host immune responses and can inhibit pathogenic inflammation. Helminth infection promotes a strong Th2 and T regulatory response while suppressing Th1 and Th17 function. Th2 responses are largely dependent on transcriptional programs directed by Stat6-signaling. We examined the importance of intact T cell Stat6 signaling on helminth-induced suppression of murine colitis that results from T cell transfer into immune-deficient mice. Colonization with the intestinal nematode Heligmosomoides polygyrus bakeri resolves WT T cell transfer colitis. However, if the transferred T cells lack intact Stat6 then helminth exposure failed to attenuate colitis or suppress MLN T cell IFN-γ or IL17 production. Loss of Stat6 signaling resulted in decreased IL10 and increased IFN-γ co-expression by IL-17+ T cells. We also transferred T cells from mice with constitutive T cell expression of activated Stat6 (Stat6VT). These mice developed a severe eosinophilic colitis that also was not attenuated by helminth infection. These results show that T cell expression of intact but regulated Stat6 signaling is required for helminth infection-associated regulation of pathogenic intestinal inflammation.


Asunto(s)
Colitis/inmunología , Nematospiroides dubius/inmunología , Factor de Transcripción STAT6/inmunología , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Animales , Colitis/parasitología , Interferón gamma/inmunología , Interleucina-10/inmunología , Interleucina-17/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Ratones , Ratones Endogámicos C57BL , Células TH1/inmunología , Células TH1/parasitología , Células Th17/inmunología , Células Th17/parasitología , Células Th2/inmunología , Células Th2/parasitología
9.
Dev Comp Immunol ; 115: 103879, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33007336

RESUMEN

The mouse intestinal parasite Heligmosomoides polygyrus demonstrates adaptation to the inflammatory milieu as a result of colitis induced by dextran sulphate sodium (DSS). Nematodes from mice with colitis had different effects on dendritic cells than nematodes from mice without colitis. Immature JAWSII cells pre-exposed to L4 stage H. polygyrus from DSS-treated mice were adoptively transferred to mice with induced colitis. After two days, a higher disease activity index, macroscopic damage score and colon histology score were observed. MLN T cells isolated nine days after transfer demonstrated proinflammatory IFN-γ and IL-17 production. Transfer of JAWSII stimulated with male or female L4 larvae from a control invasion resulted in a slight improvement of colitis; in addition, dendritic cells exposed to H. polygyrus female L4 larvae, provoked migration of CD8+CD25+ T cells from MLN to the colon. Nematodes from an inflammatory environment changed cytokine production by dendritic cells. Inflammatory milieu changing nematode immunomodulatory activity affects dendritic cell functions, which offers new insight into the helminth-host relationship.


Asunto(s)
Colitis/terapia , Células Dendríticas/inmunología , Nematospiroides dubius/inmunología , Terapia con Helmintos/métodos , Traslado Adoptivo , Animales , Movimiento Celular/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos/inmunología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Larva , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Mesenterio , Ratones , Cultivo Primario de Células , Factores Sexuales , Linfocitos T/inmunología
10.
Sci Immunol ; 5(53)2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33188058

RESUMEN

Interleukin-33 (IL-33) is a pleiotropic cytokine that can promote type 2 inflammation but also drives immunoregulation through Foxp3+Treg expansion. How IL-33 is exported from cells to serve this dual role in immunosuppression and inflammation remains unclear. Here, we demonstrate that the biological consequences of IL-33 activity are dictated by its cellular source. Whereas IL-33 derived from epithelial cells stimulates group 2 innate lymphoid cell (ILC2)-driven type 2 immunity and parasite clearance, we report that IL-33 derived from myeloid antigen-presenting cells (APCs) suppresses host-protective inflammatory responses. Conditional deletion of IL-33 in CD11c-expressing cells resulted in lowered numbers of intestinal Foxp3+Treg cells that express the transcription factor GATA3 and the IL-33 receptor ST2, causing elevated IL-5 and IL-13 production and accelerated anti-helminth immunity. We demonstrate that cell-intrinsic IL-33 promoted mouse dendritic cells (DCs) to express the pore-forming protein perforin-2, which may function as a conduit on the plasma membrane facilitating IL-33 export. Lack of perforin-2 in DCs blocked the proliferative expansion of the ST2+Foxp3+Treg subset. We propose that perforin-2 can provide a plasma membrane conduit in DCs that promotes the export of IL-33, contributing to mucosal immunoregulation under steady-state and infectious conditions.


Asunto(s)
Células Dendríticas/inmunología , Interleucina-33/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Animales , Membrana Celular/metabolismo , Enfermedad Crónica , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Inmunidad Mucosa , Interleucina-33/análisis , Interleucina-33/genética , Masculino , Ratones , Ratones Transgénicos , Mucosa Nasal/inmunología , Mucosa Nasal/patología , Pólipos Nasales/inmunología , Pólipos Nasales/patología , Nematospiroides dubius/inmunología , Nippostrongylus/inmunología , Proteínas Citotóxicas Formadoras de Poros , Rinitis/inmunología , Rinitis/patología , Sinusitis/inmunología , Sinusitis/patología , Infecciones por Strongylida/parasitología , Linfocitos T Reguladores/metabolismo
11.
Front Immunol ; 11: 1830, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117327

RESUMEN

Helminth parasites are effective in biasing Th2 immunity and inducing regulatory pathways that minimize excessive inflammation within their hosts, thus allowing chronic infection to occur whilst also suppressing bystander atopic or autoimmune diseases. Multiple sclerosis (MS) is a severe autoimmune disease characterized by inflammatory lesions within the central nervous system; there are very limited therapeutic options for the progressive forms of the disease and none are curative. Here, we used the experimental autoimmune encephalomyelitis (EAE) model to examine if the intestinal helminth Heligmosomoides polygyrus and its excretory/secretory products (HES) are able to suppress inflammatory disease. Mice infected with H. polygyrus at the time of immunization with the peptide used to induce EAE (myelin-oligodendrocyte glycoprotein, pMOG), showed a delay in the onset and peak severity of EAE disease, however, treatment with HES only showed a marginal delay in disease onset. Mice that received H. polygyrus 4 weeks prior to EAE induction were also not significantly protected. H. polygyrus secretes a known TGF-ß mimic (Hp-TGM) and simultaneous H. polygyrus infection with pMOG immunization led to a significant expansion of Tregs; however, administering the recombinant Hp-TGM to EAE mice failed to replicate the EAE protection seen during infection, indicating that this may not be central to the disease protecting mechanism. Mice infected with H. polygyrus also showed a systemic Th2 biasing, and restimulating splenocytes with pMOG showed release of pMOG-specific IL-4 as well as suppression of inflammatory IL-17A. Notably, a Th2-skewed response was found only in mice infected with H. polygyrus at the time of EAE induction and not those with a chronic infection. Furthermore, H. polygyrus failed to protect against disease in IL-4Rα-/- mice. Together these results indicate that the EAE disease protective mechanism of H. polygyrus is likely to be predominantly Th2 deviation, and further highlights Th2-biasing as a future therapeutic strategy for MS.


Asunto(s)
Antígenos Helmínticos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Receptores de Superficie Celular/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/inmunología
12.
Int J Mol Sci ; 21(21)2020 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-33105843

RESUMEN

As the global incidences of colorectal cancer rises, there is a growing importance in understanding the interaction between external factors, such as common infections, on the initiation and progression of this disease. While certain helminth infections have been shown to alter the severity and risk of developing colitis-associated colorectal cancer, whether these parasites can directly affect colorectal cancer progression is unknown. Here, we made use of murine and human colorectal cancer cell lines to demonstrate that exposure to antigens derived from the gastrointestinal nematode Heligmosomoides polygyrus significantly reduced colorectal cancer cell proliferation in vitro. Using a range of approaches, we demonstrate that antigen-dependent reductions in cancer cell proliferation and viability are associated with increased expression of the critical cell cycle regulators p53 and p21. Interestingly, H. polygyrus-derived antigens significantly increased murine colorectal cancer cell migration, which was associated with an increased expression of the adherens junction protein ß-catenin, whereas the opposite was true for human colorectal cancer cells. Together, these findings demonstrate that antigens derived from a gastrointestinal nematode can significantly alter colorectal cancer cell behavior. Further in-depth analysis may reveal novel candidates for targeting and treating late-stage cancer.


Asunto(s)
Antígenos Helmínticos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Nematospiroides dubius/inmunología , Animales , Antígenos Helmínticos/inmunología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Células HCT116 , Humanos , Masculino , Ratones Endogámicos C57BL , Nematospiroides dubius/metabolismo , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Especificidad de la Especie , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina/metabolismo
13.
Arch Immunol Ther Exp (Warsz) ; 68(5): 30, 2020 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-32960348

RESUMEN

An effective host immune system prevents the growth of most cancer cells. However, as intestinal nematodes are able to induce both immunotolerance and immunosuppression in the host, it is possible that their presence could allow co-occurring cancer cells to proliferate and metastasize. Our findings indicate that previous, subsequent or concurrent intestinal nematode infection affects the formation of lung metastatic nodules in mice experimentally infected with Heligmosomoides polygyrus. In addition, pre-infection with nematodes renders mice resistant to metastasis development in lungs, with the inoculated EL4 cancer cells being located mainly in mesenteric lymph nodes. The present paper discusses the nematode-induced mechanisms which may influence the metastatic process.


Asunto(s)
Helmintiasis/inmunología , Parasitosis Intestinales/inmunología , Neoplasias Pulmonares/secundario , Linfoma/inmunología , Linfoma/parasitología , Nematospiroides dubius/inmunología , Animales , Modelos Animales de Enfermedad , Inmunomodulación , Neoplasias Pulmonares/parasitología , Linfoma/patología , Masculino , Ratones , Infecciones por Nematodos/inmunología , Metástasis de la Neoplasia , Factor de Crecimiento Transformador beta/metabolismo
14.
Cell Mol Gastroenterol Hepatol ; 10(4): 763-778, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32629118

RESUMEN

BACKGROUND & AIMS: Epidemiological and animal studies have indicated an inverse correlation between the rising prevalence of obesity and metabolic syndrome and exposure to helminths. Whether helminth-induced immune response contributes to microbiota remodeling in obesity remains unknown. The aim of this study is to explore the immune-regulatory role of helminth in the prevention of HFD-induced obesity through remodeling gut microbiome. METHODS: C57BL/6J WT and STAT6-/- mice were infected with Heligmosomoides polygyrus and followed by high fat diet (HFD) feeding for 6 weeks. The host immune response, body weight, and fecal microbiota composition were analyzed. We used adoptive transfer of M2 macrophages and microbiota transplantation approaches to determine the impact of these factors on HFD-obesity. We also examined stool microbiota composition and short chain fatty acids (SCFAs) concentration and determined the expression of SCFA-relevant receptors in the recipient mice. RESULTS: Helminth infection of STAT6-/- (Th2-deficient) mice and adoptive transfer of helminth-induced alternatively activated (M2) macrophages demonstrated that the helminth-associated Th2 immune response plays an important role in the protection against obesity and induces changes in microbiota composition. Microbiota transplantation showed that helminth-induced, Th2-dependent alterations of the gut microbiota are sufficient to confer protection against obesity. Collectively, these results indicate that helminth infection protects against HFD-induced obesity by Th2-dependent, M2 macrophage-mediated alterations of the intestinal microbiota. CONCLUSION: Our findings provide new mechanistic insights into the complex interplay between helminth infection, the immune system and the gut microbiota in a HFD-induced obesity model and holds promise for gut microbiome-targeted immunotherapy in obesity prevention.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal , Nematospiroides dubius/inmunología , Obesidad/prevención & control , Animales , Células Cultivadas , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/inmunología , Factores Protectores , Infecciones por Strongylida/inmunología
15.
J Exp Med ; 217(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32584413

RESUMEN

Antigen uptake and presentation by naive and germinal center (GC) B cells are different, with the former expressing even low-affinity BCRs efficiently capture and present sufficient antigen to T cells, whereas the latter do so more efficiently after acquiring high-affinity BCRs. We show here that antigen uptake and processing by naive but not GC B cells depend on Cbl and Cbl-b (Cbls), which consequently control naive B and cognate T follicular helper (Tfh) cell interaction and initiation of the GC reaction. Cbls mediate CD79A and CD79B ubiquitination, which is required for BCR-mediated antigen endocytosis and postendocytic sorting to lysosomes, respectively. Blockade of CD79A or CD79B ubiquitination or Cbls ligase activity is sufficient to impede BCR-mediated antigen processing and GC development. Thus, Cbls act at the entry checkpoint of the GC reaction by promoting naive B cell antigen presentation. This regulation may facilitate recruitment of naive B cells with a low-affinity BCR into GCs to initiate the process of affinity maturation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Centro Germinal/inmunología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Formación de Anticuerpos/inmunología , Antígenos CD/metabolismo , Linfocitos B/citología , Comunicación Celular/inmunología , Endocitosis , Inmunidad Humoral , Intestinos/inmunología , Intestinos/parasitología , Activación de Linfocitos/inmunología , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Mutagénesis/genética , Mutación/genética , Nematospiroides dubius/inmunología , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Proteínas Proto-Oncogénicas c-cbl/genética , Receptores de Antígenos de Linfocitos B/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Ubiquitinación
16.
Elife ; 92020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32420871

RESUMEN

The IL-33-ST2 pathway is an important initiator of type 2 immune responses. We previously characterised the HpARI protein secreted by the model intestinal nematode Heligmosomoides polygyrus, which binds and blocks IL-33. Here, we identify H. polygyrus Binds Alarmin Receptor and Inhibits (HpBARI) and HpBARI_Hom2, both of which consist of complement control protein (CCP) domains, similarly to the immunomodulatory HpARI and Hp-TGM proteins. HpBARI binds murine ST2, inhibiting cell surface detection of ST2, preventing IL-33-ST2 interactions, and inhibiting IL-33 responses in vitro and in an in vivo mouse model of asthma. In H. polygyrus infection, ST2 detection is abrogated in the peritoneal cavity and lung, consistent with systemic effects of HpBARI. HpBARI_Hom2 also binds human ST2 with high affinity, and effectively blocks human PBMC responses to IL-33. Thus, we show that H. polygyrus blocks the IL-33 pathway via both HpARI which blocks the cytokine, and also HpBARI which blocks the receptor.


Asunto(s)
Alternaria/inmunología , Antígenos Helmínticos/metabolismo , Asma/patología , Proteína 1 Similar al Receptor de Interleucina-1/antagonistas & inhibidores , Interleucina-33/antagonistas & inhibidores , Nematospiroides dubius/metabolismo , Animales , Línea Celular , Humanos , Factores Inmunológicos/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nematospiroides dubius/inmunología , Ovalbúmina/inmunología
17.
Parasite Immunol ; 42(9): e12700, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32027755

RESUMEN

Intestinal helminthes induce immunosuppressive responses as well as type 2 immunity. Their suppressive properties are intended to regulate inflammatory diseases such as allergies and autoimmune diseases. This study evaluated whether helminthic infections suppress obesity, a chronic inflammatory state, using an intestinal nematode, Heligmosomoides polygyrus (Hp). Infection with Hp at the same time as feeding a high-fat diet (HFD) prevented weight gain, dyslipidaemia and glucose intolerance observed in uninfected obese mice. Immunologically, Hp infection skewed M1 macrophages to M2 macrophages and induced type 2 innate lymphoid cells in adipose tissues. The expression of interleukin (IL)-33, a potent initiator of type 2 responses, was also increased in association with uncoupled protein 1 (UCP1). To further investigate the anti-obesity effects of IL-33 in mice infected with Hp, IL-33-deficient mice were fed the HFD and infected with Hp. These mutant mice rapidly gained weight compared with wild-type mice, indicating the anti-obesity effect of IL-33. In the absence of IL-33, the rapid increase in weight was not prevented, and type 2 responses and UCP1 expression were not observed even during Hp infection. These results suggested that the suppression of obesity by Hp is dependent on IL-33.


Asunto(s)
Dieta Alta en Grasa , Interleucina-33/fisiología , Parasitosis Intestinales/inmunología , Nematospiroides dubius , Obesidad/prevención & control , Infecciones por Strongylida/inmunología , Tejido Adiposo/inmunología , Animales , Inmunidad Innata , Parasitosis Intestinales/complicaciones , Linfocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Nematospiroides dubius/inmunología , Obesidad/inmunología , Terapia con Helmintos
18.
Parasite Immunol ; 42(5): e12704, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32049381

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are heterogeneous population of monocyte and granulocyte progenitors that are highly suppressive against T cells. In BALB/c mice infected with a nematode Heligmosomoides polygyrus bakeri, we studied the dynamics of MDSCs, identified as CD11b+Gr-1+, induction in different tissues along with the development of parasite infection. We observed that MDSC-like cells are induced both by larvae and adult stages of H polygyrus bakeri. Gr-1+ cells of suppressive phenotype are recruited in the bone marrow, peripheral blood and peritoneal cavity during histotropic phase of infection and are present at that time in the intestine wall, where worms reside. Later, during intestinal phase, suppressive Gr-1+ cells increased in mesenteric lymph nodes and the spleen. l-arginine metabolism was important for the protective immunity, and parasite-induced Gr-1+ cells showed elevated arginase-1 and iNOS expression. Inhibition of arginase-1 and l-arginine administration caused reduced level of infection that coincided with weaker suppressive phenotype of Gr-1+ cells. We identified that l-arginine pathway activation and induction of MDSC-like cells characterize immunosuppressive state during H polygyrus bakeri infection in mice. Our findings confirm the role of MDSCs in parasitic infections and point l-arginine pathway as a potential target for immunomodulation during nematode infections.


Asunto(s)
Arginina/inmunología , Antígeno CD11b/inmunología , Monocitos/inmunología , Nematospiroides dubius/inmunología , Receptores de Quimiocina/inmunología , Infecciones por Strongylida/inmunología , Animales , Antígeno CD11b/genética , Femenino , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos BALB C , Monocitos/parasitología , Nematospiroides dubius/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Receptores de Quimiocina/genética , Bazo/inmunología , Infecciones por Strongylida/genética , Infecciones por Strongylida/parasitología
19.
Immunohorizons ; 4(1): 23-32, 2020 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980480

RESUMEN

Tuft cells are an epithelial cell type critical for initiating type 2 immune responses to parasites and protozoa in the small intestine. To respond to these stimuli, intestinal tuft cells use taste chemosensory signaling pathways, but the role of taste receptors in type 2 immunity is poorly understood. In this study, we show that the taste receptor TAS1R3, which detects sweet and umami in the tongue, also regulates tuft cell responses in the distal small intestine. BALB/c mice, which have an inactive form of TAS1R3, as well as Tas1r3-deficient C57BL6/J mice both have severely impaired responses to tuft cell-inducing signals in the ileum, including the protozoa Tritrichomonas muris and succinate. In contrast, TAS1R3 is not required to mount an immune response to the helminth Heligmosomoides polygyrus, which infects the proximal small intestine. Examination of uninfected Tas1r3-/- mice revealed a modest reduction in the number of tuft cells in the proximal small intestine but a severe decrease in the distal small intestine at homeostasis. Together, these results suggest that TAS1R3 influences intestinal immunity by shaping the epithelial cell landscape at steady-state.


Asunto(s)
Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Receptores Acoplados a Proteínas G/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Epiteliales/metabolismo , Microbioma Gastrointestinal , Homeostasis , Íleon/inmunología , Íleon/parasitología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nematospiroides dubius/inmunología , Receptores Acoplados a Proteínas G/deficiencia , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Gusto/fisiología , Tritrichomonas/inmunología
20.
J Immunol ; 204(4): 923-932, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31900338

RESUMEN

The transcription factor BHLHE40 is an emerging regulator of the immune system. Recent studies suggest that BHLHE40 regulates type 2 immunity, but this has not been demonstrated in vivo. We found that BHLHE40 is required in T cells for a protective TH2 cell response in mice infected with the helminth Heligmosomoides polygyrus bakeri H. polygyrus elicited changes in gene and cytokine expression by lamina propria CD4+ T cells, many of which were BHLHE40 dependent, including production of the common ß (CSF2RB) chain family cytokines GM-CSF and IL-5. In contrast to deficiency in GM-CSF or IL-5 alone, loss of both GM-CSF and IL-5 signaling impaired protection against H. polygyrus Overall, we show that BHLHE40 regulates the TH2 cell transcriptional program during helminth infection to support normal expression of Csf2, Il5, and other genes required for protection and reveal unexpected redundancy of common ß chain-dependent cytokines previously thought to possess substantially divergent functions.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas de Homeodominio/metabolismo , Interleucina-5/metabolismo , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Subunidad beta Común de los Receptores de Citocinas/genética , Subunidad beta Común de los Receptores de Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Proteínas de Homeodominio/genética , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Interleucina-5/antagonistas & inhibidores , Interleucina-5/genética , Interleucina-5/inmunología , Ratones , Ratones Noqueados , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Infecciones por Strongylida/parasitología , Células Th2/efectos de los fármacos , Transcripción Genética/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA