Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 735
Filtrar
1.
PLoS One ; 19(7): e0292408, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38950025

RESUMEN

Co-infections are a common reality but understanding how the immune system responds in this context is complex and can be unpredictable. Heligmosomoides bakeri (parasitic roundworm, previously Heligmosomoides polygyrus) and Toxoplasma gondii (protozoan parasite) are well studied organisms that stimulate a characteristic Th2 and Th1 response, respectively. Several studies have demonstrated reduced inflammatory cytokine responses in animals co-infected with such organisms. However, while general cytokine signatures have been examined, the impact of the different cytokine producing lymphocytes on parasite control/clearance is not fully understood. We investigated five different lymphocyte populations (NK, NKT, γδ T, CD4+ T and CD8+ T cells), five organs (small intestine, Peyer's patches, mesenteric lymph nodes, spleen and liver), and 4 cytokines (IFN©, IL-4, IL-10 and IL-13) at two different time points (days 5 and 10 post T. gondii infection). We found that co-infected animals had significantly higher mortality than either single infection. This was accompanied by transient and local changes in parasite loads and cytokine profiles. Despite the early changes in lymphocyte and cytokine profiles, severe intestinal pathology in co-infected mice likely contributed to early mortality due to significant damage by both parasites in the small intestine. Our work demonstrates the importance of taking a broad view during infection research, studying multiple cell types, organs/tissues and time points to link and/or uncouple immunological from pathological findings. Our results provide insights into how co-infection with parasites stimulating different arms of the immune system can lead to drastic changes in infection dynamics.


Asunto(s)
Coinfección , Citocinas , Nematospiroides dubius , Toxoplasma , Animales , Coinfección/inmunología , Coinfección/parasitología , Toxoplasma/inmunología , Ratones , Citocinas/metabolismo , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/mortalidad , Toxoplasmosis/inmunología , Toxoplasmosis/mortalidad , Toxoplasmosis/complicaciones , Femenino , Toxoplasmosis Animal/inmunología , Toxoplasmosis Animal/mortalidad , Toxoplasmosis Animal/parasitología , Bazo/inmunología , Bazo/patología , Bazo/parasitología , Carga de Parásitos , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Tejido Linfoide/parasitología
2.
Nat Commun ; 15(1): 5226, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38890291

RESUMEN

IL-33 plays a significant role in inflammation, allergy, and host defence against parasitic helminths. The model gastrointestinal nematode Heligmosomoides polygyrus bakeri secretes the Alarmin Release Inhibitor HpARI2, an effector protein that suppresses protective immune responses and asthma in its host by inhibiting IL-33 signalling. Here we reveal the structure of HpARI2 bound to mouse IL-33. HpARI2 contains three CCP-like domains, and we show that it contacts IL-33 primarily through the second and third of these. A large loop which emerges from CCP3 directly contacts IL-33 and structural comparison shows that this overlaps with the binding site on IL-33 for its receptor, ST2, preventing formation of a signalling complex. Truncations of HpARI2 which lack the large loop from CCP3 are not able to block IL-33-mediated signalling in a cell-based assay and in an in vivo female mouse model of asthma. This shows that direct competition between HpARI2 and ST2 is responsible for suppression of IL-33-dependent responses.


Asunto(s)
Asma , Proteínas del Helminto , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Nematospiroides dubius , Animales , Interleucina-33/metabolismo , Interleucina-33/química , Nematospiroides dubius/inmunología , Proteínas del Helminto/metabolismo , Proteínas del Helminto/química , Proteínas del Helminto/inmunología , Ratones , Femenino , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Asma/inmunología , Asma/metabolismo , Humanos , Transducción de Señal , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/metabolismo , Unión Proteica , Modelos Animales de Enfermedad , Sitios de Unión , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
3.
Gut Microbes ; 16(1): 2370917, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38944838

RESUMEN

Polyphenols are phytochemicals commonly found in plant-based diets which have demonstrated immunomodulatory and anti-inflammatory properties. However, the interplay between polyphenols and pathogens at mucosal barrier surfaces has not yet been elucidated in detail. Here, we show that proanthocyanidin (PAC) polyphenols interact with gut parasites to influence immune function and gut microbial-derived metabolites in mice. PAC intake inhibited mastocytosis during infection with the small intestinal roundworm Heligmosomoides polygyrus, and altered the host tissue transcriptome at the site of infection with the large intestinal whipworm Trichuris muris, with a notable enhancement of type-1 inflammatory and interferon-driven gene pathways. In the absence of infection, PAC intake promoted the expansion of Turicibacter within the gut microbiota, increased fecal short chain fatty acids, and enriched phenolic metabolites such as phenyl-γ-valerolactones in the cecum. However, these putatively beneficial effects were reduced in PAC-fed mice infected with T. muris, suggesting concomitant parasite infection can attenuate gut microbial-mediated PAC catabolism. Collectively, our results suggest an inter-relationship between a phytonutrient and infection, whereby PAC may augment parasite-induced inflammation (most prominently with the cecum dwelling T. muris), and infection may abrogate the beneficial effects of health-promoting phytochemicals.


Asunto(s)
Microbioma Gastrointestinal , Nematospiroides dubius , Polifenoles , Proantocianidinas , Tricuriasis , Trichuris , Animales , Ratones , Polifenoles/farmacología , Polifenoles/metabolismo , Trichuris/metabolismo , Tricuriasis/parasitología , Tricuriasis/inmunología , Nematospiroides dubius/inmunología , Proantocianidinas/metabolismo , Proantocianidinas/farmacología , Ratones Endogámicos C57BL , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/metabolismo , Femenino , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Heces/parasitología , Heces/microbiología
4.
Allergy ; 79(8): 2157-2172, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38924546

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) infection in infants is a major cause of viral bronchiolitis and hospitalisation. We have previously shown in a murine model that ongoing infection with the gut helminth Heligmosomoides polygyrus protects against RSV infection through type I interferon (IFN-I) dependent reduction of viral load. Yet, the cellular basis for this protection has remained elusive. Given that recruitment of mononuclear phagocytes to the lung is critical for early RSV infection control, we assessed their role in this coinfection model. METHODS: Mice were infected by oral gavage with H. polygyrus. Myeloid immune cell populations were assessed by flow cytometry in lung, blood and bone marrow throughout infection and after secondary infection with RSV. Monocyte numbers were depleted by anti-CCR2 antibody or increased by intravenous transfer of enriched monocytes. RESULTS: H. polygyrus infection induces bone marrow monopoiesis, increasing circulatory monocytes and lung mononuclear phagocytes in a IFN-I signalling dependent manner. This expansion causes enhanced lung mononuclear phagocyte counts early in RSV infection that may contribute to the reduction of RSV load. Depletion or supplementation of circulatory monocytes prior to RSV infection confirms that these are both necessary and sufficient for helminth induced antiviral protection. CONCLUSIONS: H. polygyrus infection induces systemic monocytosis contributing to elevated mononuclear phagocyte numbers in the lung. These cells are central to an anti-viral effect that reduces the peak viral load in RSV infection. Treatments to promote or modulate these cells may provide novel paths to control RSV infection in high risk individuals.


Asunto(s)
Modelos Animales de Enfermedad , Monocitos , Infecciones por Virus Sincitial Respiratorio , Carga Viral , Animales , Infecciones por Virus Sincitial Respiratorio/inmunología , Ratones , Monocitos/inmunología , Nematospiroides dubius/inmunología , Pulmón/inmunología , Pulmón/virología , Infecciones por Strongylida/inmunología , Virus Sincitiales Respiratorios/inmunología , Interferón Tipo I/metabolismo
5.
Sci Rep ; 14(1): 10773, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730262

RESUMEN

The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.


Asunto(s)
Hipocampo , Plasticidad Neuronal , Animales , Femenino , Hipocampo/metabolismo , Hipocampo/parasitología , Embarazo , Ratones , Infecciones por Nematodos/inmunología , Infecciones por Nematodos/parasitología , Potenciación a Largo Plazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Masculino , Neuroinmunomodulación
6.
PLoS Negl Trop Dis ; 18(5): e0012188, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38805557

RESUMEN

BACKGROUND: Angiostrongylus cantonensis is a parasite that mainly infects the heart and pulmonary arteries of rats and causes human eosinophilic meningitis or meningoencephalitis in certain geographical areas. Current diagnostic methods include detection of the parasite in cerebrospinal fluid (CSF) and eosinophilic immune examination after lumbar puncture, which may be risky and produce false-positive results. 18F- Fluorodeoxyglucose (FDG), a Positron emission tomography (PET) tracer, has been used to assess different pathological or inflammatory changes in the brains of patients. In this study, we hypothesized that A. cantonensis infection-induced inflammatory and immunomodulatory factors of eosinophils result in localized pathological changes in the brains of non-permissive hosts, which could be analyzed using in vivo 18F-FDG PET imaging. METHODOLOGY/FINDINGS: Non-permissive host ICR mice and permissive host SD rats were infected with A. cantonensis, and the effects of the resulting inflammation on 18F-FDG uptake were characterized using PET imaging. We also quantitatively measured the distributed uptake values of different brain regions to build an evaluated imaging model of localized neuropathological damage caused by eosinophilic inflammation. Our results showed that the uptake of 18F-FDG increased in the cerebellum, brainstem, and limbic system of mice at three weeks post-infection, whereas the uptake in the rat brain was not significant. Immunohistochemical staining and western blotting revealed that Iba-1, a microglia-specific marker, significantly increased in the hippocampus and its surrounding area in mice after three weeks of infection, and then became pronounced after four weeks of infection; while YM-1, an eosinophilic chemotactic factor, in the hippocampus and midbrain, increased significantly from two weeks post-infection, sharply escalated after three weeks of infection, and peaked after four weeks of infection. Cytometric bead array (CBA) analysis revealed that the expression of TNF in the serum of mice increased concomitantly with the prolongation of infection duration. Furthermore, IFN-γ and IL-4 in rat serum were significantly higher than in mouse serum at two weeks post-infection, indicating significantly different immune responses in the brains of rats and mice. We suggest that 18F-FDG uptake in the host brain may be attributed to the accumulation of large numbers of immune cells, especially the metabolic burst of activated eosinophils, which are attracted to and induced by activated microglia in the brain. CONCLUSIONS: An in vivo 18F-FDG/PET imaging model can be used to evaluate live neuroinflammatory pathological changes in the brains of A. cantonensis-infected mice and rats.


Asunto(s)
Angiostrongylus cantonensis , Encéfalo , Fluorodesoxiglucosa F18 , Tomografía de Emisión de Positrones , Ratas Sprague-Dawley , Infecciones por Strongylida , Animales , Angiostrongylus cantonensis/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/diagnóstico por imagen , Infecciones por Strongylida/patología , Encéfalo/parasitología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Encéfalo/inmunología , Ratones , Ratas , Eosinófilos/inmunología , Inflamación/inmunología , Masculino , Modelos Animales de Enfermedad , Lectinas/metabolismo , Femenino , beta-N-Acetilhexosaminidasas
7.
Front Immunol ; 14: 1130933, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37063913

RESUMEN

The initiation of type 2 immune responses at mucosal barriers is regulated by rapidly secreted cytokines called alarmins. The alarmins IL-33, IL-25 and TSLP are mainly secreted by stromal and epithelial cells in tissues and were linked to chronic inflammatory diseases, such as allergic lung inflammation, or to resistance against worm infections. Receptors for alarmins are expressed by a variety of immune cells, including group 2 innate lymphoid cells (ILC2s), an early source of the type 2 cytokines, such as IL-5 and IL-13, which have been linked to atopic diseases and anti-worm immunity as well. However, the precise contribution of the IL-33 receptor signals for ILC2 activation still needs to be completed due to limitations in targeting genes in ILC2. Using the newly established Nmur1 iCre-eGFP mouse model, we obtained specific conditional genetic ablation of the IL-33 receptor subunit ST2 in ILC2s. ST2-deficient ILC2s were unresponsive to IL-33 but not to stimulation with the alarmin IL-25. As a result of defective ST2 signals, ILC2s produced limited amounts of IL-5 and IL-13 and failed to support eosinophil homeostasis. Further, ST2-deficient ILC2s were unable to expand and promote the recruitment of eosinophils during allergic lung inflammation provoked by papain administration. During infection with Nippostrongylus brasiliensis, ILC2-intrinsic ST2 signals were required to mount an effective type 2 immune response against the parasite leading to higher susceptibility against worm infection in conditional knockout mice. Therefore, this study argues for a non-redundant role of cell-intrinsic ST2 signals triggering proper activation of ILC2 for initiation of type 2 immunity.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1 , Eosinofilia Pulmonar , Infecciones por Strongylida , Animales , Ratones , Alarminas , Citocinas/inmunología , Inmunidad Innata , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-13 , Interleucina-33 , Interleucina-5 , Linfocitos , Eosinofilia Pulmonar/inmunología , Nippostrongylus , Infecciones por Strongylida/inmunología
8.
Elife ; 122023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36753434

RESUMEN

The myeloid C-type lectin receptor (CLR) MINCLE senses the mycobacterial cell wall component trehalose-6,6'-dimycolate (TDM). Recently, we found that IL-4 downregulates MINCLE expression in macrophages. IL-4 is a hallmark cytokine in helminth infections, which appear to increase the risk for mycobacterial infection and active tuberculosis. Here, we investigated functional consequences of IL-4 and helminth infection on MINCLE-driven macrophage activation and Th1/Th17 adjuvanticity. IL-4 inhibited MINCLE and cytokine induction after macrophage infection with Mycobacterium bovis bacille Calmette-Guerin (BCG). Infection of mice with BCG upregulated MINCLE on myeloid cells, which was inhibited by IL-4 plasmid injection and by infection with the nematode Nippostrongylus brasiliensis in monocytes. To determine the impact of helminth infection on MINCLE-dependent immune responses, we vaccinated mice with a recombinant protein together with the MINCLE ligand trehalose-6,6-dibehenate (TDB) as adjuvant. Concurrent infection with N. brasiliensis or with Schistosoma mansoni promoted T cell-derived IL-4 production and suppressed Th1/Th17 differentiation in the spleen. In contrast, helminth infection did not reduce Th1/Th17 induction by TDB in draining peripheral lymph nodes, where IL-4 levels were unaltered. Upon use of the TLR4-dependent adjuvant G3D6A, N. brasiliensis infection impaired selectively the induction of splenic antigen-specific Th1 but not of Th17 cells. Inhibition of MINCLE-dependent Th1/Th17 responses in mice infected with N. brasiliensis was dependent on IL-4/IL-13. Thus, helminth infection attenuated the Th17 response to MINCLE-dependent immunization in an organ- and adjuvant-specific manner via the Th2 cytokines IL-4/IL-13. Taken together, our results demonstrate downregulation of MINCLE expression on monocytes and macrophages by IL-4 as a possible mechanism of thwarted Th17 vaccination responses by underlying helminth infection.


Asunto(s)
Interleucina-4 , Lectinas Tipo C , Proteínas de la Membrana , Infecciones por Strongylida , Animales , Ratones , Adyuvantes Inmunológicos , Vacuna BCG , Citocinas/inmunología , Interleucina-13 , Interleucina-4/inmunología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos/inmunología , Mycobacterium bovis , Células TH1 , Células Th17/inmunología , Proteínas de la Membrana/metabolismo , Nippostrongylus , Infecciones por Strongylida/inmunología
9.
Eur J Immunol ; 52(12): 1925-1933, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36116042

RESUMEN

Infection of mice with Nippostrongylus brasiliensis (Nb) serves as a model for human hookworm infection affecting about 600 million people world-wide. Expulsion of Nb from the intestine requires IL-13-mediated mucus secretion from goblet cells and activation of smooth muscles cells. Type 2 innate lymphoid cells (ILC2s) are a major cellular source of IL-13 but it remains unclear whether IL-13 secretion from ILC2s is required for Nb expulsion. Here, we compared the immune response to Nb infection in mixed bone marrow chimeras with wild-type or IL-4/IL-13-deficient ILC2s. ILC2-derived IL-4/IL-13 was required for recruitment of eosinophils to the lung but had no influence of systemic eosinophil levels. In the small intestine, goblet cell hyperplasia and tuft cell accumulation was largely dependent on IL-4/IL-13 secretion from ILC2s. This further translated to higher eggs counts and impaired worm expulsion in mice with IL-4/IL-13-deficient ILC2s. Overall, we demonstrate that ILC2s constitute a non-redundant source of IL-4/IL-13 required for protective immunity against primary Nb infection.


Asunto(s)
Inmunidad Innata , Linfocitos , Infecciones por Strongylida , Animales , Ratones , Interleucina-13 , Interleucina-4 , Nippostrongylus , Infecciones por Strongylida/inmunología
10.
Cell Rep ; 38(2): 110215, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021079

RESUMEN

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Asunto(s)
Pulmón/inmunología , Macrófagos Alveolares/inmunología , Infecciones por Strongylida/inmunología , Animales , Arginasa/metabolismo , Diferenciación Celular , Citocinas , Femenino , Pulmón/parasitología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Nippostrongylus , Infecciones por Strongylida/parasitología
11.
J Nutr Biochem ; 100: 108887, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34655757

RESUMEN

Phytonutrients such as cinnamaldehyde (CA) have been studied for their effects on metabolic diseases, but their influence on mucosal inflammation and immunity to enteric infection are not well documented. Here, we show that consumption of CA in mice significantly down-regulates transcriptional pathways connected to inflammation in the small intestine, and alters T-cell populations in mesenteric lymph nodes. During infection with the enteric helminth Heligomosomoides polygyrus, CA treatment attenuated infection-induced changes in biological pathways connected to cell cycle and mitotic activity, and tended to reduce worm burdens. Mechanistically, CA did not appear to exert activity through a prebiotic effect, as CA treatment did not significantly change the composition of the gut microbiota. Instead, in vitro experiments showed that CA directly induced xenobiotic metabolizing pathways in intestinal epithelial cells and suppressed endotoxin-induced inflammatory responses in macrophages. Collectively, our results show that CA down-regulates inflammatory pathways in the intestinal mucosa and can limit the pathological response to enteric infection. These properties appear to be largely independent of the gut microbiota, and instead connected to the ability of CA to induce antioxidant pathways in intestinal cells. Our results encourage further investigation into the use of CA and related phytonutrients as functional food components to promote intestinal health in humans and animals.


Asunto(s)
Acroleína/análogos & derivados , Suplementos Dietéticos , Inflamación/inmunología , Intestino Delgado/metabolismo , Fitoquímicos/administración & dosificación , Infecciones por Strongylida/inmunología , Acroleína/administración & dosificación , Acroleína/farmacología , Animales , Células Cultivadas , Femenino , Microbioma Gastrointestinal , Inmunidad Mucosa , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Ganglios Linfáticos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nematospiroides dubius , Fitoquímicos/farmacología , Linfocitos T/inmunología , Transcripción Genética , Transcriptoma , Xenobióticos/metabolismo
12.
Mucosal Immunol ; 15(1): 74-83, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34420044

RESUMEN

Macrophages are a heterogeneous population of innate immune cells that are often divided into two major subsets: classically activated, typically pro-inflammatory (M1) macrophages that mediate host defense, and alternatively activated, tolerance-inducing (M2) macrophages that exert homeostatic and tissue-regenerative functions. Disturbed macrophage function/differentiation results either in inadequate, excessive immune activation or in a failure to induce efficient protective immune responses against pathogens. Loss-of-function variants in protein tyrosine phosphatase non-receptor type 2 (PTPN2) are associated with chronic inflammatory disorders, but the effect of macrophage-intrinsic PTPN2 loss is still poorly understood. Here we report that PTPN2-deficient macrophages fail to acquire an alternatively activated/M2 phenotype. This was the consequence of reduced IL-6 receptor expression and a failure to induce IL-4 receptor in response to IL-6, resulting in an inability to respond to the key M2-inducing cytokine IL-4. Ultimately, failure to adequately respond to IL-6 and IL-4 resulted in increased levels of M1 macrophage marker expression in vitro and exacerbated lung inflammation upon infection with Nippostrongylus brasiliensis in vivo. These results demonstrate that PTPN2 loss interferes with the ability of macrophages to adequately respond to inflammatory stimuli and might explain the increased susceptibility of PTPN2 loss-of-function carriers to developing inflammatory diseases.


Asunto(s)
Inflamación/inmunología , Pulmón/inmunología , Macrófagos/inmunología , Nippostrongylus/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Infecciones por Strongylida/inmunología , Animales , Diferenciación Celular , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-4/metabolismo , Pulmón/parasitología , Ratones , Ratones Noqueados , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Células THP-1 , Células TH1/inmunología , Células Th2/inmunología
13.
Science ; 374(6568): eabe6723, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34735226

RESUMEN

A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.


Asunto(s)
Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Microbioma Gastrointestinal , Bacterias Grampositivas/fisiología , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Nematospiroides dubius , Infecciones por Strongylida/inmunología , Animales , Carga Bacteriana , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Proteínas Ricas en Prolina del Estrato Córneo/genética , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Células Caliciformes/metabolismo , Humanos , Inmunidad Innata , Mucosa Intestinal/microbiología , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Ratones , Viabilidad Microbiana , Células de Paneth/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/microbiología
14.
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
15.
Front Immunol ; 12: 711907, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484215

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are early effectors of mucosal type 2 immunity, producing cytokines such as interleukin (IL)-13 to mediate responses to helminth infection and allergen-induced inflammation. ILC2s are also present in lymph nodes (LNs) and can express molecules required for antigen presentation, but to date there are limited data on their dynamic behaviour. We used a CD2/IL-13 dual fluorescent reporter mouse for in vivo imaging of ILC2s and Th2 T cells in real time following a type 2 priming helminth infection or egg injection. After helminth challenge, we found that ILC2s were the main source of IL-13 in lymphoid organs (Peyer's patches and peripheral LNs), and were located in T cell areas. Intravital imaging demonstrated an increase in IL-13+ ILC2 size and movement following helminth infection, but reduced duration of interactions with T cells compared with those in homeostasis. In contrast, in the intestinal mucosa, we observed an increase in ILC2-T cell interactions post-infection, including some of prolonged duration, as well as increased IL-13+ ILC2 movement. These data suggest that ILC2 activation enhances cell motility, with the potential to increase the area of distribution of cytokines to optimise the early generation of type 2 responses. The prolonged ILC2 interactions with T cells within the intestinal mucosa are consistent with the conclusion that contact-based T cell activation may occur within inflamed tissues rather than lymphoid organs. Our findings have important implications for our understanding of the in vivo biology of ILC2s and the way in which these cells facilitate adaptive immune responses.


Asunto(s)
Parasitosis Intestinales/inmunología , Subgrupos Linfocitarios/inmunología , Nippostrongylus , Esquistosomiasis mansoni/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Animales , Genes Reporteros , Interleucina-13/análisis , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Intestino Delgado/parasitología , Microscopía Intravital , Recuento de Linfocitos , Subgrupos Linfocitarios/química , Ratones , Especificidad de Órganos , Organismos Libres de Patógenos Específicos , Células Th2/química
16.
Nat Metab ; 3(9): 1150-1162, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34531575

RESUMEN

Macrophages exhibit a spectrum of activation states ranging from classical to alternative activation1. Alternatively, activated macrophages are involved in diverse pathophysiological processes such as confining tissue parasites2, improving insulin sensitivity3 or promoting an immune-tolerant microenvironment that facilitates tumour growth and metastasis4. Recently, the metabolic regulation of macrophage function has come into focus as both the classical and alternative activation programmes require specific regulated metabolic reprogramming5. While most of the studies regarding immunometabolism have focussed on the catabolic pathways activated to provide energy, little is known about the anabolic pathways mediating macrophage alternative activation. In this study, we show that the anabolic transcription factor sterol regulatory element binding protein 1 (SREBP1) is activated in response to the canonical T helper 2 cell cytokine interleukin-4 to trigger the de novo lipogenesis (DNL) programme, as a necessary step for macrophage alternative activation. Mechanistically, DNL consumes NADPH, partitioning it away from cellular antioxidant defences and raising reactive oxygen species levels. Reactive oxygen species serves as a second messenger, signalling sufficient DNL, and promoting macrophage alternative activation. The pathophysiological relevance of this mechanism is validated by showing that SREBP1/DNL is essential for macrophage alternative activation in vivo in a helminth infection model.


Asunto(s)
Antioxidantes/metabolismo , Ácidos Grasos/biosíntesis , Macrófagos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Dexametasona/farmacología , Humanos , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Nippostrongylus/aislamiento & purificación , Nippostrongylus/patogenicidad , Células RAW 264.7 , Análisis de Secuencia de ARN/métodos , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Regulación hacia Arriba
17.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34290141

RESUMEN

"Taste-like" tuft cells in the intestine trigger type 2 immunity in response to worm infection. The secretion of interleukin-13 (IL-13) from type 2 innate lymphoid cells (ILC2) represents a key step in the tuft cell-ILC2 cell-intestinal epithelial cell circuit that drives the clearance of worms from the gut via type 2 immune responses. Hallmark features of type 2 responses include tissue remodeling, such as tuft and goblet cell expansion, and villus atrophy, yet it remains unclear if additional molecular changes in the gut epithelium facilitate the clearance of worms from the gut. Using gut organoids, we demonstrated that IL-4 and IL-13, two type 2 cytokines with similar functions, not only induced the classical type 2 responses (e.g., tuft cell expansion) but also drastically up-regulated the expression of gasdermin C genes (Gsdmcs). Using an in vivo worm-induced type 2 immunity model, we confirmed the up-regulation of Gsdmcs in Nippostrongylus brasiliensis-infected wild-type C57BL/6 mice. Consistent with gasdermin family members being principal effectors of pyroptosis, overexpression of Gsdmc2 in human embryonic kidney 293 (HEK293) cells triggered pyroptosis and lytic cell death. Moreover, in intestinal organoids treated with IL-4 or IL-13, or in wild-type mice infected with N. brasiliensis, lytic cell death increased, which may account for villus atrophy observed in worm-infected mice. Thus, we propose that the up-regulated Gsdmc family may be major effectors for type 2 responses in the gut and that Gsdmc-mediated pyroptosis may provide a conduit for the release of antiparasitic factors from enterocytes to facilitate the clearance of worms.


Asunto(s)
Muerte Celular , Proteínas de Unión al ADN/metabolismo , Enterocitos/patología , Inmunidad Innata/inmunología , Intestino Delgado/patología , Infecciones por Strongylida/complicaciones , Células Th2/inmunología , Animales , Proliferación Celular , Proteínas de Unión al ADN/genética , Enterocitos/inmunología , Enterocitos/metabolismo , Enterocitos/parasitología , Femenino , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Intestino Delgado/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Nippostrongylus/fisiología , Transducción de Señal , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología
18.
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
19.
PLoS Pathog ; 17(6): e1009602, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34106992

RESUMEN

The CD4+ T cell response is critical to host protection against helminth infection. How this response varies across different hosts and tissues remains an important gap in our understanding. Using IL-4-reporter mice to identify responding CD4+ T cells to Nippostrongylus brasiliensis infection, T cell receptor sequencing paired with novel clustering algorithms revealed a broadly reactive and clonally diverse CD4+ T cell response. While the most prevalent clones and clonotypes exhibited some tissue selectivity, most were observed to reside in both the lung and lung-draining lymph nodes. Antigen-reactivity of the broader repertoires was predicted to be shared across both tissues and individual mice. Transcriptome, trajectory, and chromatin accessibility analysis of lung and lymph-node repertoires revealed three unique but related populations of responding IL-4+ CD4+ T cells consistent with T follicular helper, T helper 2, and a transitional population sharing similarity with both populations. The shared antigen reactivity of lymph node and lung repertoires combined with the adoption of tissue-specific gene programs allows for the pairing of cellular and humoral responses critical to the orchestration of anti-helminth immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Strongylida/inmunología , Animales , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Ratones , Nippostrongylus , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Análisis de la Célula Individual
20.
Sci Immunol ; 6(60)2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117110

RESUMEN

Effective antiviral immunity requires generation of T and B lymphocytes expressing the transcription factor T-bet, a regulator of type 1 inflammatory responses. Using T-bet expression as an endogenous marker for cells participating in a type 1 response, we report coordinated interactions of T-bet-expressing T and B lymphocytes on the basis of their dynamic colocalization at the T cell zone and B follicle boundary (T-B boundary) and germinal centers (GCs) during lung influenza infection. We demonstrate that the assembly of this circuit takes place in distinct anatomical niches within the draining lymph node, guided by CXCR3 that enables positioning of TH1 cells at the T-B boundary. The encounter of B and TH1 cells at the T-B boundary enables IFN-γ produced by the latter to induce IgG2c class switching. Within GCs, T-bet+ TFH cells represent a specialized stable sublineage required for GC growth but dispensable for IgG2c class switching. Our studies show that during respiratory viral infection, T-bet-expressing T and B lymphocytes form a circuit assembled in a spatiotemporally controlled manner that acts as a functional unit enabling a robust and coherent humoral response tailored for optimal antiviral immunity.


Asunto(s)
Linfocitos B/inmunología , Inmunidad Humoral , Gripe Humana/inmunología , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Animales , Linfocitos B/metabolismo , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Femenino , Centro Germinal/citología , Centro Germinal/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina , Virus de la Influenza A/inmunología , Gripe Humana/patología , Gripe Humana/virología , Interferón gamma/genética , Interferón gamma/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Transgénicos , Nippostrongylus/inmunología , Ratas , Receptores CXCR3/metabolismo , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/metabolismo , Células TH1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA