Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
J Immunol ; 210(3): 297-309, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36524995

RESUMEN

CD8 virtual memory T (TVM) cells are Ag-naive CD8 T cells that have undergone partial differentiation in response to common γ-chain cytokines, particularly IL-15 and IL-4. TVM cells from young individuals are highly proliferative in response to TCR and cytokine stimulation but, with age, they lose TCR-mediated proliferative capacity and exhibit hallmarks of senescence. Helminth infection can drive an increase in TVM cells, which is associated with improved pathogen clearance during subsequent infectious challenge in young mice. Given the cytokine-dependent profile of TVM cells and their age-associated dysfunction, we traced proliferative and functional changes in TVM cells, compared with true naive CD8 T cells, after helminth infection of young and aged C57BL/6 mice. We show that IL-15 is essential for the helminth-induced increase in TVM cells, which is driven only by proliferation of existing TVM cells, with negligible contribution from true naive cell differentiation. Additionally, TVM cells showed the greatest proliferation in response to helminth infection and IL-15 compared with other CD8 T cells. Furthermore, TVM cells from aged mice did not undergo expansion after helminth infection due to both TVM cell-intrinsic and -extrinsic changes associated with aging.


Asunto(s)
Helmintiasis , Interleucina-15 , Animales , Ratones , Envejecimiento/inmunología , Linfocitos T CD8-positivos/parasitología , Citocinas , Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintos/patogenicidad , Memoria Inmunológica , Interleucina-15/metabolismo , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T
2.
Nat Commun ; 13(1): 5192, 2022 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-36057627

RESUMEN

Dynamic regulation of intestinal epithelial cell (IEC) differentiation is crucial for both homeostasis and the response to helminth infection. SIRT6 belongs to the NAD+-dependent deacetylases and has established diverse roles in aging, metabolism and disease. Here, we report that IEC Sirt6 deletion leads to impaired tuft cell development and type 2 immunity in response to helminth infection, thereby resulting in compromised worm expulsion. Conversely, after helminth infection, IEC SIRT6 transgenic mice exhibit enhanced epithelial remodeling process and more efficient worm clearance. Mechanistically, Sirt6 ablation causes elevated Socs3 expression, and subsequently attenuated tyrosine 641 phosphorylation of STAT6 in IECs. Notably, intestinal epithelial overexpression of constitutively activated STAT6 (STAT6vt) in mice is sufficient to induce the expansion of tuft and goblet cell linage. Furthermore, epithelial STAT6vt overexpression remarkedly reverses the defects in intestinal epithelial remodeling caused by Sirt6 ablation. Our results reveal a novel function of SIRT6 in regulating intestinal epithelial remodeling and mucosal type 2 immunity in response to helminth infection.


Asunto(s)
Helmintiasis/inmunología , Mucosa Intestinal , Factor de Transcripción STAT6/metabolismo , Sirtuinas/metabolismo , Animales , Células Epiteliales/metabolismo , Células Caliciformes/metabolismo , Helmintiasis/metabolismo , Inmunidad Mucosa , Mucosa Intestinal/metabolismo , Intestinos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT6/genética , Sirtuinas/genética
3.
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
4.
Sci Immunol ; 6(57)2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674322

RESUMEN

Group 2 innate lymphoid cells (ILC2s) reside in multiple tissues, including lymphoid organs and barrier surfaces, and secrete type 2 cytokines including interleukin-5 (IL-5), IL-9, and IL-13. These cells participate in multiple physiological processes including allergic inflammation, tissue repair, metabolic homeostasis, and host defense against helminth infections. Recent studies indicate that neurotransmitters and neuropeptides can play an important role in regulating ILC2 responses; however, the mechanisms that underlie these processes in vivo remain incompletely defined. Here, we identify that activated ILC2s up-regulate choline acetyltransferase (ChAT)-the enzyme responsible for the biosynthesis of acetylcholine (ACh)-after infection with the helminth parasite Nippostrongylus brasiliensis or treatment with alarmins or cytokines including IL-25, IL-33, and thymic stromal lymphopoietin (TSLP). ILC2s also express acetylcholine receptors (AChRs), and ACh administration promotes ILC2 cytokine production and elicits expulsion of helminth infection. In accordance with this, ChAT deficiency in ILC2s leads to defective ILC2 responses and impaired immunity against helminth infection. Together, these results reveal a previously unrecognized role of the ChAT-ACh pathway in promoting type 2 innate immunity to helminth infection.


Asunto(s)
Acetilcolina/metabolismo , Colina O-Acetiltransferasa/metabolismo , Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintos/inmunología , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Animales , Biomarcadores , Colina O-Acetiltransferasa/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Eosinófilos/metabolismo , Expresión Génica , Helmintiasis/parasitología , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunofenotipificación , Ratones
5.
Front Immunol ; 11: 594520, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193446

RESUMEN

Helminths, including nematodes, cestodes and trematodes, are complex parasitic organisms that infect at least one billion people globally living in extreme poverty. Helminthic infections are associated with severe morbidity particularly in young children who often harbor the highest burden of disease. While each helminth species completes a distinct life cycle within the host, several helminths incite significant lung disease. This impact on the lungs occurs either directly from larval migration and host immune activation or indirectly from a systemic inflammatory immune response. The impact of helminths on the pulmonary immune response involves a sophisticated orchestration and activation of the host innate and adaptive immune cells. The consequences of activating pulmonary host immune responses are variable with several helminthic infections leading to severe, pulmonary compromise while others providing immune tolerance and protection against the development of pulmonary diseases. Further delineation of the convoluted interface between helminth infection and the pulmonary host immune responses is critical to the development of novel therapeutics that are critically needed to prevent the significant global morbidity caused by these parasites.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/parasitología , Helmintos/inmunología , Interacciones Huésped-Parásitos/inmunología , Enfermedades Pulmonares Parasitarias/inmunología , Enfermedades Pulmonares Parasitarias/parasitología , Inmunidad Adaptativa , Animales , Biomarcadores , Susceptibilidad a Enfermedades , Helmintiasis/metabolismo , Helmintos/crecimiento & desarrollo , Humanos , Inmunidad , Inmunidad Innata , Inmunomodulación , Estadios del Ciclo de Vida , Enfermedades Pulmonares Parasitarias/metabolismo , Especificidad de Órganos/inmunología
6.
Molecules ; 25(21)2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33139647

RESUMEN

Neglected parasitic diseases remain a major public health issue worldwide, especially in tropical and subtropical areas. Human parasite diversity is very large, ranging from protozoa to worms. In most cases, more effective and new drugs are urgently needed. Previous studies indicated that the gold(I) drug auranofin (Ridaura®) is effective against several parasites. Among new gold(I) complexes, the phosphole-containing gold(I) complex {1-phenyl-2,5-di(2-pyridyl)phosphole}AuCl (abbreviated as GoPI) is an irreversible inhibitor of both purified human glutathione and thioredoxin reductases. GoPI-sugar is a novel 1-thio-ß-d-glucopyranose 2,3,4,6-tetraacetato-S-derivative that is a chimera of the structures of GoPI and auranofin, designed to improve stability and bioavailability of GoPI. These metal-ligand complexes are of particular interest because of their combined abilities to irreversibly target the essential dithiol/selenol catalytic pair of selenium-dependent thioredoxin reductase activity, and to kill cells from breast and brain tumors. In this work, screening of various parasites-protozoans, trematodes, and nematodes-was undertaken to determine the in vitro killing activity of GoPI-sugar compared to auranofin. GoPI-sugar was found to efficiently kill intramacrophagic Leishmania donovani amastigotes and adult filarial and trematode worms.


Asunto(s)
Antihelmínticos , Antineoplásicos , Antiprotozoarios , Auranofina , Complejos de Coordinación , Oro , Helmintiasis/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Infecciones por Protozoos/tratamiento farmacológico , Animales , Antihelmínticos/química , Antihelmínticos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Antiprotozoarios/química , Antiprotozoarios/farmacología , Auranofina/química , Auranofina/farmacología , Bovinos , Línea Celular Tumoral , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Evaluación de Medicamentos , Oro/química , Oro/farmacología , Helmintiasis/metabolismo , Helmintiasis/patología , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Infecciones por Protozoos/metabolismo , Infecciones por Protozoos/patología
7.
J Cell Mol Med ; 24(24): 14325-14338, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33124146

RESUMEN

CD4+ T cells differentiate into distinct functional effector and inhibitory subsets are facilitated by distinct cytokine cues present at the time of antigen recognition. Maintaining a balance between T helper 17 (Th17) and regulatory T (Treg) cells are critical for the control of the immunopathogenesis of liver diseases. Here, by using the mouse model of helminth Schistosoma japonicum (S japonicum) infection, we show that the hepatic mRNA levels of P21-activated kinase 1 (PAK1), a key regulator of the actin cytoskeleton, adhesion and cell motility, are significantly increased and associated with the development of liver pathology during S japonicum infection. In addition, PAK1-deficient mice are prone to suppression of Th17 cell responses but increased Treg cells. Furthermore, PAK1 enhances macrophage activation through promoting IRF1 nuclear translocation in an NF-κB-dependent pathway, resulting in promoting Th17 cell differentiation through inducing IL-6 production. These findings highlight the importance of PAK1 in macrophages fate determination and suggest that PAK1/IRF1 axis-dependent immunomodulation can ameliorate certain T cell-based immune pathologies.


Asunto(s)
Helmintiasis/metabolismo , Helmintiasis/parasitología , Macrófagos/inmunología , Macrófagos/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Quinasas p21 Activadas/metabolismo , Animales , Antígenos Helmínticos/inmunología , Biomarcadores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inmunofenotipificación , Ratones , Esquistosomiasis Japónica/inmunología , Esquistosomiasis Japónica/metabolismo , Esquistosomiasis Japónica/parasitología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Eur J Pharmacol ; 881: 173209, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32454117

RESUMEN

Asthma and allergic diseases are a group of chronic inflammatory disorders that arise as a result of excessive responses of the immune system against intrinsically harmless environmental substances. It is well known that substantial joint characteristics exist between the immune and nervous systems. The semaphorins (Semas) were initially characterized as axon-guidance molecules that play a crucial role during the development of the nervous system. However, increasing evidence indicates that a subset of Semas, termed "immune Semas", acting through their cognate receptors, namely, plexins (Plxns), and neuropilins (Nrps), also contributes to both physiological and pathological responses of the immune system. Notably, immune Semas exert critical roles in regulating a broad spectrum of biological processes, including immune cell-cell interactions, activation, differentiation, cell migration and mobility, angiogenesis, tumor progression, as well as inflammatory responses. Accumulating evidence indicates that the modification in the signaling of immune Semas could lead to various immune-mediated inflammatory diseases, ranging from cancer to autoimmunity and allergies. This review summarizes the recent evidence regarding the role of immune Semas in the pathogenesis of asthma and allergic diseases and discusses their therapeutic potential for treating these diseases.


Asunto(s)
Asma/metabolismo , Hipersensibilidad/metabolismo , Semaforinas/metabolismo , Animales , Antihelmínticos/uso terapéutico , Antialérgicos/uso terapéutico , Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Asma/inmunología , Helmintiasis/tratamiento farmacológico , Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintiasis/parasitología , Humanos , Hipersensibilidad/inmunología , Hipersensibilidad/fisiopatología , Mediadores de Inflamación/metabolismo , Neuropilinas/metabolismo , Receptores Inmunológicos/metabolismo , Semaforinas/inmunología , Semaforinas/uso terapéutico , Transducción de Señal
9.
Dokl Biol Sci ; 487(1): 101-104, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31571075

RESUMEN

In this ecological and physiological study of the common eider (Somateria mollissima) nesting on the coast of Eastern Murman, the species composition of the bird helminth fauna, as well as the infection quantitative parameters, were determined. The common eider small intestine proved to be infected with trematodes of the genus Microphallus; three species of cestodes, namely, Lateriporus teres (Cestoda: Dilepididae), Fimbriarioides intermedia (Cestoda: Hymenolepididae), and Microsomacanthus diorchis (Cestoda: Hymenolepididae); and one species of acanthocephalan, Polymorphus phippsi (Palaeacanthocephala: Polymorphidae). At the sites of F. intermedia and M. diorchis locations within the intestine, the protease activity was reduced while in the foci infected with acanthocephalan P. phippsi, it was, on the contrary, increased. Glycosidase activity in the intestinal mucosa was reduced as compared to the control in birds infected by the cestodes M. diorchis. Hematological indices of the infected individuals were higher than the control parameters.


Asunto(s)
Anseriformes/parasitología , Enfermedades de las Aves/parasitología , Helmintiasis/parasitología , Intestino Delgado/parasitología , Acantocéfalos/patogenicidad , Animales , Proteínas Aviares/metabolismo , Enfermedades de las Aves/metabolismo , Cestodos/patogenicidad , Glicósido Hidrolasas/metabolismo , Helmintiasis/metabolismo , Mucosa Intestinal/metabolismo , Péptido Hidrolasas/metabolismo , Trematodos/patogenicidad
10.
Nat Commun ; 10(1): 4408, 2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31562318

RESUMEN

Intestinal epithelial cells (IEC) have important functions in nutrient absorption, barrier integrity, regeneration, pathogen-sensing, and mucus secretion. Goblet cells are a specialized cell type of IEC that secrete Trefoil factor 3 (TFF3) to regulate mucus viscosity and wound healing, but whether TFF3-responsiveness requires a receptor is unclear. Here, we show that leucine rich repeat receptor and nogo-interacting protein 2 (LINGO2) is essential for TFF3-mediated functions. LINGO2 immunoprecipitates with TFF3, co-localizes with TFF3 on the cell membrane of IEC, and allows TFF3 to block apoptosis. We further show that TFF3-LINGO2 interactions disrupt EGFR-LINGO2 complexes resulting in enhanced EGFR signaling. Excessive basal EGFR activation in Lingo2 deficient mice increases disease severity during colitis and augments immunity against helminth infection. Conversely, TFF3 deficiency reduces helminth immunity. Thus, TFF3-LINGO2 interactions de-repress inhibitory LINGO2-EGFR complexes, allowing TFF3 to drive wound healing and immunity.


Asunto(s)
Colitis/inmunología , Receptores ErbB/inmunología , Helmintiasis/inmunología , Mucosa Intestinal/inmunología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/inmunología , Factor Trefoil-3/inmunología , Animales , Línea Celular Tumoral , Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran , Receptores ErbB/genética , Receptores ErbB/metabolismo , Células Caliciformes/inmunología , Células Caliciformes/metabolismo , Células Caliciformes/parasitología , Células HEK293 , Helmintiasis/metabolismo , Helmintiasis/parasitología , Helmintos/inmunología , Helmintos/fisiología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/parasitología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Organofosfonatos , Factor Trefoil-3/genética , Factor Trefoil-3/metabolismo , Células U937
11.
Parasitology ; 146(11): 1371-1378, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31258097

RESUMEN

The multifaceted interactions occurring between gastrointestinal (GI) parasitic helminths and the host gut microbiota are emerging as a key area of study within the broader research domain of host-pathogen relationships. Over the past few years, a wealth of investigations has demonstrated that GI helminths interact with the host gut flora, and that such interactions result in modifications of the host immune and metabolic statuses. Nevertheless, whilst selected changes in gut microbial composition are consistently observed in response to GI helminth infections across several host-parasite systems, research in this area to date is largely characterised by inconsistent findings. These discrepancies are particularly evident when data from studies of GI helminth-microbiota interactions conducted in humans from parasite-endemic regions are compared. In this review, we provide an overview of the main sources of variance that affect investigations on helminth-gut microbiota interactions in humans, and propose a series of methodological approaches that, whilst accounting for the inevitable constraints of fieldwork, are aimed at minimising confounding factors and draw biologically meaningful interpretations from highly variable datasets.


Asunto(s)
Microbioma Gastrointestinal , Helmintiasis , Parasitosis Intestinales , Microbioma Gastrointestinal/inmunología , Helmintiasis/inmunología , Helmintiasis/metabolismo , Humanos , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/metabolismo
12.
Proc Biol Sci ; 286(1902): 20190456, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31064304

RESUMEN

Over a billion people on earth are infected with helminth parasites and show remarkable variation in parasite burden and chronicity. These parasite distributions are captured well by classic statistics, such as the negative binomial distribution. But the within-host processes underlying this variation are not well understood. In this study, we explain variation in macroparasite infection outcomes on the basis of resource flows within hosts. Resource flows realize the interactions between parasites and host immunity and metabolism. When host metabolism is modulated by parasites, we find a positive feedback of parasites on their own resources. While this positive feedback results in parasites improving their resource availability at high burdens, giving rise to chronic infections, it also results in a threshold biomass required for parasites to establish in the host, giving rise to acute infections when biomass fails to clear the threshold. Our finding of chronic and acute outcomes in bistability contrasts with classic theory, yet is congruent with the variation in helminth burdens observed in human and wildlife populations.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintos/fisiología , Interacciones Huésped-Parásitos , Animales , Animales Salvajes , Humanos , Modelos Biológicos
13.
Trends Immunol ; 40(6): 538-552, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31103422

RESUMEN

Infection with helminth parasites poses a significant challenge to the mammalian immune system. The type 2 immune response to helminth infection is critical in limiting worm-induced tissue damage and expelling parasites. Conversely, aberrant type 2 inflammation can cause debilitating allergic disease. Recent studies have revealed that key type 2 inflammation-associated immune and epithelial cell types respond to Notch signaling, broadly regulating gene expression programs in cell development and function. Here, we discuss new advances demonstrating that Notch is active in the development, recruitment, localization, and cytokine production of immune and epithelial effector cells during type 2 inflammation. Understanding how Notch signaling controls type 2 inflammatory processes could inform the development of Notch pathway modulators to treat helminth infections and allergies.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintiasis/parasitología , Helmintos/inmunología , Interacciones Huésped-Parásitos/inmunología , Receptores Notch/metabolismo , Transducción de Señal , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Inmunidad Innata/inmunología , Leucocitos/inmunología , Leucocitos/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
14.
Front Immunol ; 10: 623, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31019505

RESUMEN

Group 2 innate lymphoid cells (ILC2s) were first discovered in experimental studies of intestinal helminth infection-and much of our current knowledge of ILC2 activation and function is based on the use of these models. It is perhaps not surprising therefore that these cells have also been found to play a key role in mediating protection against these large multicellular parasites. ILC2s have been intensively studied over the last decade, and are known to respond quickly and robustly to the presence of helminths-both by increasing in number and producing type 2 cytokines. These mediators function to activate and repair epithelial barriers, to recruit other innate cells such as eosinophils, and to help activate T helper 2 cells. More recent investigations have focused on the mechanisms by which the host senses helminth parasites to activate ILC2s. Such studies have identified novel stromal cell types as being involved in this process-including intestinal tuft cells and enteric neurons, which respond to the presence of helminths and activate ILC2s by producing IL-25 and Neuromedin, respectively. In the current review, we will outline the latest insights into ILC2 activation and discuss the requirement for-or redundancy of-ILC2s in providing protective immunity against intestinal helminth parasites.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/parasitología , Helmintos/inmunología , Interacciones Huésped-Parásitos/inmunología , Inmunidad Innata , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/parasitología , Linfocitos/inmunología , Inmunidad Adaptativa , Alarminas/metabolismo , Animales , Biomarcadores , Helmintiasis/metabolismo , Humanos , Inmunomodulación , Mediadores de Inflamación/metabolismo , Parasitosis Intestinales/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Activación de Linfocitos/inmunología , Linfocitos/metabolismo
15.
Front Immunol ; 9: 2353, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30405603

RESUMEN

Type III IFNs are important players in immunity to viral and bacterial infections. However, their association with helminth infections has not been examined. To explore the association of Type III IFNs with Strongyloides stercoralis (Ss) infection, we examined the systemic levels of IFN lambda-1, IFN lambda-2 and IFN lambda-3, IL-10, and CXCL10/IP-10 in Ss infected (INF, n = 44), helminth-uninfected (UN, n = 44) and in post-treatment INF individuals. We also examined the levels of IFN lambda-1, IFN lambda-2 and IFN lambda-3, IL-10, and CXCL10/IP-10 in whole blood culture supernatants stimulated with Ss somatic antigens, or PPD or LPS. Finally, we performed correlations of systemic Type III IFN levels with absolute numbers of dendritic cell subsets. Ss infection is characterized by elevated systemic levels of IFN lambda-1, IFN lambda-2 and IFN lambda-3, IL-10, and CXCL10/IP-10 in comparison to UN individuals and a significant reduction following anthelmintic treatment. Ss infection is also characterized by elevated levels of unstimulated or Ss antigen stimulated levels of IFN lambda-1, IFN lambda-2 and IFN lambda-3, CXCL10/IP-10 and a significant reduction following treatment. In addition, Ss infection is characterized by increased numbers of plasmacytoid and myeloid dendritic cells in comparison to UN individuals, with a significant reduction following anthelmintic treatment of INF individuals. Finally, Ss infection exhibits a significant positive correlation between the systemic levels of IFN lambda-2 and IFN lambda-3 and the numbers of plasmacytoid dendritic cells. Thus, Ss infection is characterized by elevations in systemic and antigen-induced levels of Type III IFNs, which is positively associated with the numbers of plasmacytoid dendritic cells and reversed upon anthelmintic treatment.


Asunto(s)
Antígenos Helmínticos/inmunología , Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintos/inmunología , Interferones/metabolismo , Adolescente , Adulto , Anciano , Animales , Antihelmínticos/farmacología , Antihelmínticos/uso terapéutico , Biomarcadores , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Helmintiasis/tratamiento farmacológico , Helmintiasis/parasitología , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
16.
Front Immunol ; 9: 2349, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30369927

RESUMEN

Helminth parasites are masters at manipulating host immune responses, using an array of sophisticated mechanisms. One of the major mechanisms enabling helminths to establish chronic infections is the targeting of pattern recognition receptors (PRRs) including toll-like receptors, C-type lectin receptors, and the inflammasome. Given the critical role of these receptors and their intracellular pathways in regulating innate inflammatory responses, and also directing adaptive immunity toward Th1 and Th2 responses, recognition of the pathways triggered and/or modulated by helminths and their products will provide detailed insights about how helminths are able to establish an immunoregulatory environment. However, helminths also target PRRs-independent mechanisms (and most likely other yet unknown mechanisms and pathways) underpinning the battery of different molecules helminths produce. Herein, the current knowledge on intracellular pathways in antigen presenting cells activated by helminth-derived biomolecules is reviewed. Furthermore, we discuss the importance of helminth-derived vesicles as a less-appreciated components released during infection, their role in activating these host intracellular pathways, and their implication in the development of new therapeutic approaches for inflammatory diseases and the possibility of designing a new generation of vaccines.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintos/inmunología , Interacciones Huésped-Parásitos/inmunología , Inmunomodulación , Animales , Biomarcadores , Vesículas Extracelulares/metabolismo , Helmintiasis/parasitología , Helmintiasis/prevención & control , Helmintos/metabolismo , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Espacio Intracelular/metabolismo , Vacunas Antiprotozoos/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Vesículas Secretoras/metabolismo , Transducción de Señal
17.
Expert Rev Gastroenterol Hepatol ; 12(10): 997-1006, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30113218

RESUMEN

INTRODUCTION: Macrophages represent a highly heterogeneous and plastic cell type found in most tissues of the body; the intestine is home to enormous numbers of these cells. Considerable interest surrounds the 'M2 macrophage,' as it is able to control and regulate inflammation, while promoting tissue repair. Areas covered: As potent inducers of M2 macrophages, intestinal helminths and helminth-derived products are ideal candidates for small molecule drug design to drive M2 macrophage polarization. Several gastrointestinal helminths have been found to cause M2 macrophage-inducing infections. This review covers current knowledge of helminth products and their impact on macrophage polarization, which may in the future lead to new therapeutic strategies. A literature search was performed using the following search terms in PubMed: M2 macrophage, alternative activation, helminth products, helminth ES, helminth therapy, nanoparticle, intestinal macrophages. Other studies were selected by using references from articles identified through our original literature search. Expert commentary: While the immunomodulatory potential of helminth products is well established, we have yet to fully characterize many components of the intestinal helminth product library. Current work aims to identify the protein motifs responsible for modulation of macrophages and other components of the immune system.


Asunto(s)
Antígenos Helmínticos/uso terapéutico , Enfermedades Gastrointestinales/terapia , Helmintiasis/inmunología , Macrófagos/inmunología , Terapia con Helmintos , Animales , Enfermedad Celíaca/terapia , Helmintiasis/metabolismo , Humanos , Inflamación/inmunología , Inflamación/terapia , Enfermedades Inflamatorias del Intestino/terapia
18.
Front Immunol ; 9: 1763, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30108588

RESUMEN

Helminth infections, by nematodes, trematodes, or cestodes, can lead to the modulation of host immune responses. This allows long-duration parasite infections and also impacts responses to co-infections. Surface, secreted, excreted, and shed proteins are thought to play a major role in modulation. A commonly reported feature of such immune modulation is the role of T regulatory (Treg) cells and IL-10. Efforts to identify helminth proteins, which cause immunomodulation, have identified candidates but not provided clarity as to a uniform mechanism driving modulation. In this study, we applied a bioinformatics systems approach, allowing us to analyze predicted T-cell epitopes of 17 helminth species and the responses to their surface proteins. In addition to major histocompatibility complex (MHC) binding, we analyzed amino acid motifs that would be recognized by T-cell receptors [T-cell-exposed motifs (TCEMs)]. All the helminth species examined have, within their surface proteins, peptides, which combine very common TCEMs with predicted high affinity binding to many human MHC alleles. This combination of features would result in large cognate T cell and a high probability of eliciting Treg responses. The TCEMs, which determine recognition by responding T-cell clones, are shared to a high degree between helminth species and with Plasmodium falciparum and Mycobacterium tuberculosis, both common co-infecting organisms. The implication of our observations is not only that Treg cells play a significant role in helminth-induced immune modulation but also that the epitope specificities of Treg responses are shared across species and genera of helminth. Hence, the immune response to a given helminth cannot be considered in isolation but rather forms part of an epitope ecosystem, or microenvironment, in which potentially immunosuppressive peptides in the helminth network via their common T-cell receptor recognition signals with T-cell epitopes in self proteins, microbiome, other helminths, and taxonomically unrelated pathogens. Such a systems approach provides a high-level view of the antigen-immune system signaling dynamics that may bias a host's immune response to helminth infections toward immune modulation. It may indicate how helminths have evolved to select for peptides that favor long-term parasite host coexistence.


Asunto(s)
Epítopos de Linfocito T/inmunología , Helmintiasis/inmunología , Helmintos/inmunología , Inmunomodulación/inmunología , Linfocitos T Reguladores/inmunología , Animales , Epítopos de Linfocito T/metabolismo , Helmintiasis/metabolismo , Helmintiasis/parasitología , Helmintos/clasificación , Helmintos/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Malaria Falciparum/inmunología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/fisiología , Plasmodium falciparum/inmunología , Plasmodium falciparum/fisiología , Linfocitos T Reguladores/metabolismo , Tuberculosis/inmunología , Tuberculosis/metabolismo , Tuberculosis/microbiología
19.
Front Immunol ; 9: 56, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29459856

RESUMEN

Parasitic helminths are extremely resilient in their ability to maintain chronic infection burdens despite (or maybe because of) their hosts' immune response. Explaining how parasites maintain these lifelong infections, identifying the protective immune mechanisms that regulate helminth infection burdens, and designing prophylactics and therapeutics that combat helminth infection, while preserving host health requires a far better understanding of how the immune system functions in natural habitats than we have at present. It is, therefore, necessary to complement mechanistic laboratory-based studies with studies on wild populations and their natural parasite communities. Unfortunately, the relative paucity of immunological tools for non-model species has held these types of studies back. Thankfully, recent progress in high-throughput 'omics platforms provide powerful and increasingly practical means for immunologists to move beyond traditional lab-based model organisms. Yet, assigning both metabolic and immune function to genes, transcripts, and proteins in novel species and assessing how they interact with other physiological and environmental factors requires identifying quantitative relationships between their expression and infection. Here, we used supervised machine learning to identify gene networks robustly associated with burdens of the gastrointestinal nematode Heligmosomoides polygyrus in its natural host, the wild wood mice Apodemus sylvaticus. Across 34 mice spanning two wild populations and across two different seasons, we found 17,639 transcripts that clustered in 131 weighted gene networks. These clusters robustly predicted H. polygyrus burden and included well-known effector and regulatory immune genes, but also revealed a number of genes associated with the maintenance of tissue homeostasis and hematopoiesis that have so far received little attention. We then tested the effect of experimentally reducing helminth burdens through drug treatment on those putatively protective immune factors. Despite the near elimination of H. polygyrus worms, the treatment had surprisingly little effect on gene expression. Taken together, these results suggest that hosts balance tissue homeostasis and protective immunity, resulting in relatively stable immune and, consequently, parasitological profiles. In the future, applying our approach to larger numbers of samples from additional populations will help further increase our ability to detect the immune pathways that determine chronic gastrointestinal helminth burdens in the wild.


Asunto(s)
Helmintiasis/inmunología , Helmintiasis/parasitología , Helmintos/inmunología , Interacciones Huésped-Parásitos/inmunología , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/parasitología , Transducción de Señal , Animales , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Helmintiasis/genética , Helmintiasis/metabolismo , Parasitosis Intestinales/genética , Parasitosis Intestinales/metabolismo , Masculino , Ratones , Nematospiroides dubius/inmunología , Carga de Parásitos , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Transcriptoma
20.
Am J Physiol Gastrointest Liver Physiol ; 312(6): G537-G549, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28302598

RESUMEN

Helminth parasites are highly prevalent in many low- and middle-income countries, in which inflammatory bowel disease and other immunopathologies are less frequent than in the developed world. Many of the most common helminths establish themselves in the gastrointestinal tract and can exert counter-inflammatory influences on the host immune system. For these reasons, interest has arisen as to how parasites may ameliorate intestinal inflammation and whether these organisms, or products they release, could offer future therapies for immune disorders. In this review, we discuss interactions between helminth parasites and the mucosal immune system, as well as the progress being made toward identifying mechanisms and molecular mediators through which it may be possible to attenuate pathology in the intestinal tract.


Asunto(s)
Tracto Gastrointestinal/parasitología , Helmintiasis/parasitología , Helmintos/fisiología , Inmunidad Mucosa , Parasitosis Intestinales/parasitología , Animales , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintiasis/patología , Helmintos/inmunología , Helmintos/metabolismo , Interacciones Huésped-Parásitos , Humanos , Hipótesis de la Higiene , Inmunoterapia/métodos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/parasitología , Enfermedades Inflamatorias del Intestino/terapia , Parasitosis Intestinales/inmunología , Parasitosis Intestinales/metabolismo , Parasitosis Intestinales/patología , Simbiosis , Terapia con Helmintos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...