Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Immunity ; 55(7): 1250-1267.e12, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35709757

RESUMEN

The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.


Asunto(s)
Enfermedades Transmisibles , Microbiota , Animales , Eosinófilos , Homeostasis , Mucosa Intestinal , Intestino Delgado , Ratones
2.
Mucosal Immunol ; 15(6): 1283-1295, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35288644

RESUMEN

Soil-transmitted helminths cause widespread disease, infecting ~1.5 billion people living within poverty-stricken regions of tropical and subtropical countries. As adult worms inhabit the intestine alongside bacterial communities, we determined whether the bacterial microbiota impacted on host resistance against intestinal helminth infection. We infected germ-free, antibiotic-treated and specific pathogen-free mice, with the intestinal helminth Heligmosomoides polygyrus bakeri. Mice harboured increased parasite numbers in the absence of a bacterial microbiota, despite mounting a robust helminth-induced type 2 immune response. Alterations to parasite behaviour could already be observed at early time points following infection, including more proximal distribution of infective larvae along the intestinal tract and increased migration in a Baermann assay. Mice lacking a complex bacterial microbiota exhibited reduced levels of intestinal acetylcholine, a major excitatory intestinal neurotransmitter that promotes intestinal transit by activating muscarinic receptors. Both intestinal motility and host resistance against larval infection were restored by treatment with the muscarinic agonist bethanechol. These data provide evidence that a complex bacterial microbiota provides the host with resistance against intestinal helminths via its ability to regulate intestinal motility.


Asunto(s)
Helmintiasis , Parasitosis Intestinales , Nematospiroides dubius , Infecciones por Strongylida , Ratones , Animales , Motilidad Gastrointestinal
3.
Immunol Cell Biol ; 100(4): 223-234, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35156238

RESUMEN

Recent advances in the field of host immunity against parasitic nematodes have revealed the importance of macrophages in trapping tissue migratory larvae. Protective immune mechanisms against the rodent hookworm Nippostrongylus brasiliensis (Nb) are mediated, at least in part, by IL-4-activated macrophages that bind and trap larvae in the lung. However, it is still not clear how host macrophages recognize the parasite. An in vitro co-culture system of bone marrow-derived macrophages and Nb infective larvae was utilized to screen for the possible ligand-receptor pair involved in macrophage attack of larvae. Competitive binding assays revealed an important role for ß-glucan recognition in the process. We further identified a role for CD11b and the non-classical pattern recognition receptor ephrin-A2 (EphA2), but not the highly expressed ß-glucan dectin-1 receptor, in this process of recognition. This work raises the possibility that parasitic nematodes synthesize ß-glucans and it identifies CD11b and ephrin-A2 as important pattern recognition receptors involved in the host recognition of these evolutionary old pathogens. To our knowledge, this is the first time that EphA2 has been implicated in immune responses to a helminth.


Asunto(s)
Interleucina-4 , Lectinas Tipo C , Ancylostomatoidea , Animales , Interleucina-4/metabolismo , Larva , Lectinas Tipo C/metabolismo , Macrófagos/metabolismo , Receptores Inmunológicos
4.
J Exp Med ; 219(1)2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34779829

RESUMEN

Helminth parasites are adept manipulators of the immune system, using multiple strategies to evade the host type 2 response. In the intestinal niche, the epithelium is crucial for initiating type 2 immunity via tuft cells, which together with goblet cells expand dramatically in response to the type 2 cytokines IL-4 and IL-13. However, it is not known whether helminths modulate these epithelial cell populations. In vitro, using small intestinal organoids, we found that excretory/secretory products (HpES) from Heligmosomoides polygyrus blocked the effects of IL-4/13, inhibiting tuft and goblet cell gene expression and expansion, and inducing spheroid growth characteristic of fetal epithelium and homeostatic repair. Similar outcomes were seen in organoids exposed to parasite larvae. In vivo, H. polygyrus infection inhibited tuft cell responses to heterologous Nippostrongylus brasiliensis infection or succinate, and HpES also reduced succinate-stimulated tuft cell expansion. Our results demonstrate that helminth parasites reshape their intestinal environment in a novel strategy for undermining the host protective response.


Asunto(s)
Células Epiteliales/metabolismo , Células Caliciformes/metabolismo , Intestino Delgado/citología , Organoides/metabolismo , Infecciones por Strongylida/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Epiteliales/parasitología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Células Caliciformes/parasitología , Proteínas del Helminto/metabolismo , Proteínas del Helminto/farmacología , Interacciones Huésped-Parásitos , Interleucina-13/farmacología , Interleucina-4/farmacología , Intestino Delgado/parasitología , Ratones Endogámicos C57BL , Nematospiroides dubius/metabolismo , Nematospiroides dubius/fisiología , Nippostrongylus/metabolismo , Nippostrongylus/fisiología , Organoides/citología , Organoides/parasitología , Infecciones por Strongylida/parasitología , Ácido Succínico/farmacología , Transcriptoma/efectos de los fármacos
5.
Trends Parasitol ; 36(9): 761-772, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32713764

RESUMEN

Gastrointestinal helminth infection still constitutes a major public health issue, particularly in the developing world. As these parasites can undergo a large part of their lifecycle within the intestinal tract the host has developed various structural and cellular specializations at the epithelial barrier to contend with infection. Detailed characterization of these cells will provide important insights about their contributions to the protective responses mediated against helminths. Here, we discuss how key components of the intestinal epithelium may function to limit the initial establishment of helminths, and how these cells are altered during an active response to infection.


Asunto(s)
Helmintiasis/inmunología , Interacciones Huésped-Parásitos/inmunología , Mucosa Intestinal/inervación , Mucosa Intestinal/parasitología , Animales , Helmintiasis/parasitología , Helmintos/inmunología , Humanos
6.
Int J Parasitol ; 50(9): 623-633, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32659278

RESUMEN

The hunt for effective vaccines against the major helminth diseases of humans has yet to bear fruit despite much effort over several decades. No individual parasite antigen has proved to elicit full protective immunity, suggesting that combinatorial strategies may be required. Recently it has been discovered that extracellular vesicles released by parasitic helminths contain multiple potential immune modulators, which could together be targeted by a future vaccine. Increasing knowledge of helminth extracellular vesicle components, both enclosed by and exposed on the membrane, will open up a new field of targets for an effective vaccine. This review discusses the interactions between helminth extracellular vesicles and the immune system discovered thus far, and the advantages of targeting these lipid-bound packages with a vaccine. In addition, we also comment upon specific antigens that may be the best targets for an anti-helminth vaccine. In the future, extensive knowledge of the parasites' full arsenal in controlling their host may finally provide us with the ideal target for a fully effective vaccine.


Asunto(s)
Antígenos Helmínticos/inmunología , Vesículas Extracelulares/inmunología , Helmintiasis , Helmintos/inmunología , Interacciones Huésped-Parásitos/inmunología , Vacunas/inmunología , Animales , Helmintiasis/inmunología , Helmintiasis/parasitología , Helmintiasis/prevención & control , Humanos
7.
Parasite Immunol ; 42(7): e12728, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32394439

RESUMEN

Helminth infection represents a major health problem causing approximately 5 million disability-adjusted life years worldwide. Concerns that repeated anti-helminthic treatment may lead to drug resistance render it important that vaccines are developed but will require increased understanding of the immune-mediated cellular and antibody responses to helminth infection. IL-4 or antibody-activated murine macrophages are known to immobilize parasitic nematode larvae, but few studies have addressed whether this is translatable to human macrophages. In the current study, we investigated the capacity of human macrophages to recognize and attack larval stages of Ascaris suum, a natural porcine parasite that is genetically similar to the human helminth Ascaris lumbricoides. Human macrophages were able to adhere to and trap A suum larvae in the presence of either human or pig serum containing Ascaris-specific antibodies and other factors. Gene expression analysis of serum-activated macrophages revealed that CCL24, a potent eosinophil attractant, was the most upregulated gene following culture with A suum larvae in vitro, and human eosinophils displayed even greater ability to adhere to, and trap, A suum larvae. These data suggest that immune serum-activated macrophages can recruit eosinophils to the site of infection, where they act in concert to immobilize tissue-migrating Ascaris larvae.


Asunto(s)
Ascariasis/inmunología , Ascaris suum/inmunología , Quimiocina CCL24/metabolismo , Eosinófilos/inmunología , Macrófagos/inmunología , Animales , Anticuerpos Antihelmínticos/sangre , Formación de Anticuerpos , Ascaris lumbricoides/inmunología , Humanos , Sueros Inmunes/farmacología , Larva/inmunología , Recuento de Leucocitos , Ratones , Porcinos , Enfermedades de los Porcinos/inmunología , Vacunas/inmunología
8.
Parasite Immunol ; 42(7): e12717, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32249432

RESUMEN

Macrophages, the major population of tissue-resident mononuclear phagocytes, contribute significantly to the immune response during helminth infection. Alternatively activated macrophages (AAM) are induced early in the anti-helminth response following tissue insult and parasite recognition, amplifying the early type 2 immune cascade initiated by epithelial cells and ILC2s, and subsequently driving parasite expulsion. AAM also contribute to functional alterations in tissues infiltrated with helminth larvae, mediating both tissue repair and inflammation. Their activation is amplified and occurs more rapidly following reinfection, where they can play a dual role in trapping tissue migratory larvae and preventing or resolving the associated inflammation and damage. In this review, we will address both the known and emerging roles of tissue macrophages during helminth infection, in addition to considering both outstanding research questions and new therapeutic strategies.


Asunto(s)
Inmunidad Innata/inmunología , Macrófagos/inmunología , Infecciones por Strongylida/inmunología , Estrongílidos/inmunología , Animales , Anticuerpos Antihelmínticos/inmunología , Arginasa/inmunología , Quitinasas/inmunología , Inflamación/parasitología , Recuento de Leucocitos , Linfocitos/inmunología , Resistina/inmunología
9.
Cell Host Microbe ; 27(2): 277-289.e6, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-32053791

RESUMEN

Hookworms cause a major neglected tropical disease, occurring after larvae penetrate the host skin. Neutrophils are phagocytes that kill large pathogens by releasing neutrophil extracellular traps (NETs), but whether they target hookworms during skin infection is unknown. Using a murine hookworm, Nippostrongylus brasiliensis, we observed neutrophils being rapidly recruited and deploying NETs around skin-penetrating larvae. Neutrophils depletion or NET inhibition altered larvae behavior and enhanced the number of adult worms following murine infection. Nevertheless, larvae were able to mitigate the effect of NETs by secreting a deoxyribonuclease (Nb-DNase II) to degrade the DNA backbone. Critically, neutrophils were able to kill larvae in vitro, which was enhanced by neutralizing Nb-DNase II. Homologs of Nb-DNase II are present in other nematodes, including the human hookworm, Necator americanus, which also evaded NETs in vitro. These findings highlight the importance of neutrophils in hookworm infection and a potential conserved mechanism of immune evasion.


Asunto(s)
Ancylostomatoidea/inmunología , Endodesoxirribonucleasas/biosíntesis , Trampas Extracelulares/metabolismo , Evasión Inmune , Animales , Interacciones Huésped-Parásitos , Ratones , Neutrófilos/metabolismo , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología
10.
Bio Protoc ; 10(24): e3849, 2020 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-33659498

RESUMEN

Hookworms are skin penetrating parasites, however in the laboratory the hookworm model Nippostrongylus brasiliensis, the parasite is traditionally administered subcutaneously bypassing the skin (epidermis and dermis). Here, we describe two complementary approaches for infecting mice with N. brasiliensis in order to study the skin immune responses. The first approach employs a skin percutaneous injection that is poorly efficient with the laboratory strain of the parasite in mice, but represents a natural infection. The second approach employs an intradermal injection of the parasite, allowing the controlled delivery of the parasitic larvae and leads to an infection that closely mimics the natural kinetics of parasite migration and development. Both of those infection models allow the investigator to study the skin immune response mounted against the parasite, in addition to detailed investigations of the early immunomodulatory strategies employed by the parasite during skin invasion.

11.
Clin Transl Immunology ; 8(11): e01089, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31719981

RESUMEN

Type 2 immunity has recently emerged as a critical player in metabolic status, with numerous studies investigating the role of type 2 immune cells within adipose tissue. Metabolic dysfunction is often characterised as a low-grade or chronic inflammatory state within tissues, and type 2 immunity may facilitate a return to metabolic homeostasis. A complex network of type 2 resident cells including M2 macrophages, eosinophils and ILC2s has been identified within adipose tissue. Although the effector cells in this equilibrium have not been clearly identified, any alteration of the type 2 microenvironment resulted in an altered metabolic state. Historically, the type 2 immune response has been associated with helminth infection. The type 2 immune response drives host resistance and plays an important role in promoting tissue repair following the migration of helminth larvae through tissues. Although helminths are largely eradicated in developed countries, infection rates remain high in poor communities within the developing world. Interestingly, there is strong evidence that helminth infection is inversely correlated with autoimmune or inflammatory disorders. Recently, an increasing amount of epidemiological and field studies suggest that it could be the same for obesity and metabolic syndrome. In the current review, we summarise the literature linking type 2 immunity to improved adipose tissue function. We then discuss more recent evidence indicating that helminth infection can provide protection against metabolic syndrome. Lastly, we explore the possible contributions of altered nutrient uptake, adipose tissue function and/or the intestinal microbiota with the ability of helminths to alter metabolic status.

14.
Nanoscale ; 10(3): 881-906, 2018 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-29265147

RESUMEN

Extracellular Vesicles (EVs) are gaining interest as central players in liquid biopsies, with potential applications in diagnosis, prognosis and therapeutic guidance in most pathological conditions. These nanosized particles transmit signals determined by their protein, lipid, nucleic acid and sugar content, and the unique molecular pattern of EVs dictates the type of signal to be transmitted to recipient cells. However, their small sizes and the limited quantities that can usually be obtained from patient-derived samples pose a number of challenges to their isolation, study and characterization. These challenges and some possible options to overcome them are discussed in this review.


Asunto(s)
Vesículas Extracelulares/química , Carbohidratos , Humanos , Lípidos , Ácidos Nucleicos , Pronóstico , Proteínas
15.
Cell Rep ; 19(8): 1545-1557, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28538175

RESUMEN

Recent studies have demonstrated that many parasites release extracellular vesicles (EVs), yet little is known about the specific interactions of EVs with immune cells or their functions during infection. We show that EVs secreted by the gastrointestinal nematode Heligmosomoides polygyrus are internalized by macrophages and modulate their activation. EV internalization causes downregulation of type 1 and type 2 immune-response-associated molecules (IL-6 and TNF, and Ym1 and RELMα) and inhibits expression of the IL-33 receptor subunit ST2. Co-incubation with EV antibodies abrogated suppression of alternative activation and was associated with increased co-localization of the EVs with lysosomes. Furthermore, mice vaccinated with EV-alum generated protective immunity against larval challenge, highlighting an important role in vivo. In contrast, ST2-deficient mice are highly susceptible to infection, and they are unable to clear parasites following EV vaccination. Hence, macrophage activation and the IL-33 pathway are targeted by H. polygyrus EVs, while neutralization of EV function facilitates parasite expulsion.


Asunto(s)
Vesículas Extracelulares/metabolismo , Inmunidad , Activación de Macrófagos , Nematospiroides dubius/metabolismo , Parásitos/metabolismo , Vacunas/inmunología , Animales , Anticuerpos Antihelmínticos/inmunología , Formación de Anticuerpos/efectos de los fármacos , Células de la Médula Ósea/citología , Citocalasina D/farmacología , Vesículas Extracelulares/efectos de los fármacos , Inmunidad/efectos de los fármacos , Proteína 1 Similar al Receptor de Interleucina-1 , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Parásitos/efectos de los fármacos , Receptores de Interleucina/metabolismo , Vacunación
16.
J Extracell Vesicles ; 5: 30741, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27389011

RESUMEN

Extracellular vesicles (EVs) mediate communication between cells and organisms across all 3 kingdoms of life. Several reports have demonstrated that EVs can transfer molecules between phylogenetically diverse species and can be used by parasites to alter the properties of the host environment. Whilst the concept of vesicle secretion and uptake is broad reaching, the molecular composition of these complexes is expected to be diverse based on the physiology and environmental niche of different organisms. Exosomes are one class of EVs originally defined based on their endocytic origin, as these derive from multivesicular bodies that then fuse with the plasma membrane releasing them into the extracellular environment. The term exosome has also been used to describe any small EVs recovered by high-speed ultracentrifugation, irrespective of origin since this is not always well characterized. Here, we use comparative global lipidomic analysis to examine the composition of EVs, which we term exosomes, that are secreted by the gastrointestinal nematode, Heligmosomoides polygyrus, in relation to exosomes secreted by cells of its murine host. Ultra-performance liquid chromatography - tandem mass spectrometry (UPLC-MS/MS) analysis reveals a 9- to 62-fold enrichment of plasmalogens, as well as other classes of ether glycerophospholipids, along with a relative lack of cholesterol and sphingomyelin (SM) in the nematode exosomes compared with those secreted by murine cells. Biophysical analyses of the membrane dynamics of these exosomes demonstrate increased rigidity in those from the nematode, and parallel studies with synthetic vesicles support a role of plasmalogens in stabilizing the membrane structure. These results suggest that nematodes can maintain exosome membrane structure and integrity through increased plasmalogens, compensating for diminished levels of other lipids, including cholesterol and SM. This work also illuminates the prevalence of plasmalogens in some EVs, which has not been widely reported and could have implications for the biochemical or immunomodulatory properties of EVs. Further comparative analyses such as those described here will shed light on diversity in the molecular properties of EVs that enable them to function in cross-species communication.

17.
Mol Biochem Parasitol ; 208(1): 33-40, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27297184

RESUMEN

Helminths are metazoan organisms many of which have evolved parasitic life styles dependent on sophisticated manipulation of the host environment. Most notably, they down-regulate host immune responses to ensure their own survival, by exporting a range of immuno-modulatory mediators that interact with host cells and tissues. While a number of secreted immunoregulatory parasite proteins have been defined, new work also points to the release of extracellular vesicles, or exosomes, that interact with and manipulate host gene expression. These recent results are discussed in the overall context of how helminths communicate effectively with the host organism.


Asunto(s)
Comunicación Celular , Helmintos/inmunología , Helmintos/fisiología , Interacciones Huésped-Parásitos , Evasión Inmune , Animales , Proteínas del Helminto/metabolismo , Humanos , Factores Inmunológicos/metabolismo
19.
Trends Parasitol ; 31(10): 477-489, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26433251

RESUMEN

Extracellular vesicles (EVs) have emerged as a ubiquitous mechanism for transferring information between cells and organisms across all three kingdoms of life. In addition to their roles in normal physiology, vesicles also transport molecules from pathogens to hosts and can spread antigens as well as infectious agents. Although initially described in the host-pathogen context for their functions in immune surveillance, vesicles enable multiple modes of communication by, and between, parasites. Here we review the literature demonstrating that EVs are secreted by intracellular and extracellular eukaryotic parasites, as well as their hosts, and detail the functional properties of these vesicles in maturation, pathogenicity and survival. We further describe the prospects for targeting or exploiting these complexes in therapeutic and vaccine strategies.


Asunto(s)
Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Interacciones Huésped-Parásitos/fisiología , Parásitos/fisiología , Enfermedades Parasitarias/metabolismo , Animales , Comunicación Celular/fisiología , Humanos , Parásitos/patogenicidad
20.
J Vis Exp ; (98): e52412, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25867600

RESUMEN

Heligmosomoides polygyrus (formerly known as Nematospiroides dubius, and also referred to by some as H. bakeri) is a gastrointestinal helminth that employs multiple immunomodulatory mechanisms to establish chronic infection in mice and closely resembles prevalent human helminth infections. H. polygyrus has been studied extensively in the field of helminth-derived immune regulation and has been found to potently suppress experimental models of allergy and autoimmunity (both with active infection and isolated secreted products). The protocol described in this paper outlines management of the H. polygyrus life cycle for consistent production of L3 larvae, recovery of adult parasites, and collection of their excretory-secretory products (HES).


Asunto(s)
Nematospiroides dubius/crecimiento & desarrollo , Nematospiroides dubius/metabolismo , Animales , Estadios del Ciclo de Vida , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA