Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Annu Rev Immunol ; 39: 639-665, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33646858

RESUMEN

Coevolutionary adaptation between humans and helminths has developed a finely tuned balance between host immunity and chronic parasitism due to immunoregulation. Given that these reciprocal forces drive selection, experimental models of helminth infection are ideally suited for discovering how host protective immune responses adapt to the unique tissue niches inhabited by these large metazoan parasites. This review highlights the key discoveries in the immunology of helminth infection made over the last decade, from innate lymphoid cells to the emerging importance of neuroimmune connections. A particular emphasis is placed on the emerging areas within helminth immunology where the most growth is possible, including the advent of genetic manipulation of parasites to study immunology and the use of engineered T cells for therapeutic options. Lastly,we cover the status of human challenge trials with helminths as treatment for autoimmune disease, which taken together, stand to keep the study of parasitic worms at the forefront of immunology for years to come.


Asunto(s)
Helmintiasis , Helmintos , Parásitos , Animales , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Linfocitos , Linfocitos T
2.
Nat Immunol ; 23(5): 743-756, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35437326

RESUMEN

Phenotypic and transcriptional profiling of regulatory T (Treg) cells at homeostasis reveals that T cell receptor activation promotes Treg cells with an effector phenotype (eTreg) characterized by the production of interleukin-10 and expression of the inhibitory receptor PD-1. At homeostasis, blockade of the PD-1 pathway results in enhanced eTreg cell activity, whereas during infection with Toxoplasma gondii, early interferon-γ upregulates myeloid cell expression of PD-L1 associated with reduced Treg cell populations. In infected mice, blockade of PD-L1, complete deletion of PD-1 or lineage-specific deletion of PD-1 in Treg cells prevents loss of eTreg cells. These interventions resulted in a reduced ratio of pathogen-specific effector T cells: eTreg cells and increased levels of interleukin-10 that mitigated the development of immunopathology, but which could compromise parasite control. Thus, eTreg cell expression of PD-1 acts as a sensor to rapidly tune the pool of eTreg cells at homeostasis and during inflammatory processes.


Asunto(s)
Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Linfocitos T Reguladores , Toxoplasmosis Animal , Animales , Antígeno B7-H1/inmunología , Homeostasis , Interleucina-10/inmunología , Ratones , Receptor de Muerte Celular Programada 1/inmunología , Linfocitos T Reguladores/inmunología , Toxoplasma/inmunología , Toxoplasmosis Animal/inmunología
3.
PLoS Pathog ; 17(7): e1009709, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34237106

RESUMEN

Helminths are distinct from microbial pathogens in both size and complexity, and are the likely evolutionary driving force for type 2 immunity. CD4+ helper T cells can both coordinate worm clearance and prevent immunopathology, but issues of T cell antigen specificity in the context of helminth-induced Th2 and T regulatory cell (Treg) responses have not been addressed. Herein, we generated a novel transgenic line of the gastrointestinal nematode Strongyloides ratti expressing the immunodominant CD4+ T cell epitope 2W1S as a fusion protein with green fluorescent protein (GFP) and FLAG peptide in order to track and study helminth-specific CD4+ T cells. C57BL/6 mice infected with this stable transgenic line (termed Hulk) underwent a dose-dependent expansion of activated CD44hiCD11ahi 2W1S-specific CD4+ T cells, preferentially in the lung parenchyma. Transcriptional profiling of 2W1S-specific CD4+ T cells isolated from mice infected with either Hulk or the enteric bacterial pathogen Salmonella expressing 2W1S revealed that pathogen context exerted a dominant influence over CD4+ T cell phenotype. Interestingly, Hulk-elicited 2W1S-specific CD4+ T cells exhibited both Th2 and Treg phenotypes and expressed high levels of the EGFR ligand amphiregulin, which differed greatly from the phenotype of 2W1S-specific CD4+ T cells elicited by 2W1S-expressing Salmonella. While immunization with 2W1S peptide did not enhance clearance of Hulk infection, immunization did increase total amphiregulin production as well as the number of amphiregulin-expressing CD3+ cells in the lung following Hulk infection. Altogether, this new model system elucidates effector as well as immunosuppressive and wound reparative roles of helminth-specific CD4+ T cells. This report establishes a new resource for studying the nature and function of helminth-specific T cells.


Asunto(s)
Epítopos de Linfocito T/genética , Estrongiloidiasis/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Animales , Animales Modificados Genéticamente , Antígenos Helmínticos , Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/inmunología , Ratones , Ratones Endogámicos C57BL , Strongyloides ratti/inmunología
4.
Am J Pathol ; 191(2): 266-273, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33245913

RESUMEN

IL-33 is an IL-1 family cytokine that signals through its cognate receptor, ST2, to regulate inflammation. Whether IL-33 serves a pathogenic or protective role during inflammatory bowel disease is controversial. Herein, two different strains of cell-specific conditionally deficient mice were used to compare the role of myeloid- versus intestinal epithelial cell-derived IL-33 during dextran sodium sulfate-induced colitis. Data show that loss of CD11c-restricted IL-33 exacerbated tissue pathology, coinciding with increased tissue Il6 levels and loss of intestinal forkhead box p3+ regulatory T cells. Surprisingly, the lack of intestinal epithelial cell-derived IL-33 had no impact on disease severity or tissue recovery. Thus, we show that myeloid-derived IL-33 functionally restrains colitic disease, whereas intestinal epithelial cell-derived IL-33 is dispensable.


Asunto(s)
Colitis/inmunología , Colitis/patología , Interleucina-33/metabolismo , Células Mieloides/inmunología , Animales , Colitis/inducido químicamente , Sulfato de Dextran/toxicidad , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Células Mieloides/metabolismo
5.
Scand J Gastroenterol ; 56(7): 791-805, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33941035

RESUMEN

Aim: Recovery of damaged mucosal surfaces following inflammatory insult requires diverse regenerative mechanisms that remain poorly defined. Previously, we demonstrated that the reparative actions of Trefoil Factor 3 (TFF3) depend upon the enigmatic receptor, leucine rich repeat and immunoglobulin-like domain containing nogo receptor 2 (LINGO2). This study examined the related orphan receptor LINGO3 in the context of intestinal tissue damage to determine whether LINGO family members are generally important for mucosal wound healing and maintenance of the intestinal stem cell (ISC) compartment needed for turnover of mucosal epithelium.Methods and Results: We find that LINGO3 is broadly expressed on human enterocytes and sparsely on discrete cells within the crypt niche, that contains ISCs. Loss of function studies indicate that LINGO3 is involved in recovery of normal intestinal architecture following dextran sodium sulfate (DSS)-induced colitis, and that LINGO3 is needed for therapeutic action of the long acting TFF2 fusion protein (TFF2-Fc), including a number of signaling pathways critical for cell proliferation and wound repair. LINGO3-TFF2 protein-protein interactions were relatively weak however and LINGO3 was only partially responsible for TFF2 induced MAPK signaling suggesting additional un-identified components of a receptor complex. However, deficiency in either TFF2 or LINGO3 abrogated budding/growth of intestinal organoids and reduced expression of the intestinal ISC gene leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5), indicating homologous roles for these proteins in tissue regeneration, possibly via regulation of ISCs in the crypt niche.Conclusion: We propose that LINGO3 serves a previously unappreciated role in promoting mucosal wound healing.


Asunto(s)
Colitis , Mucosa Intestinal , Humanos , Organoides , Factor Trefoil-2 , Cicatrización de Heridas
6.
Eur J Immunol ; 49(12): 2252-2254, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31429932

RESUMEN

Microbial flow cytometry is a powerful emerging technology with a broad range of applications including the study of complex microbial communities. Immunologists are increasingly using this technology to study antibody responses against pathogenic and commensal microbes. We employed microbial flow cytometry to quantify the proportion of fecal microbes bound by six different Ig isotypes: IgA, IgM, IgG1, IgG2b, IgG2c, and IgG3. In healthy mammals, secretory IgA (sIgA) binds to a subset of commensal microbes in the gut whereas IgG is not typically found in the intestinal tract of healthy mammals. Unexpectedly, fecal microbes isolated from SPF C57BL/6 mice housed in the Hill facility and imported from the vendors The Jackson Laboratory and Taconic Biosciences showed a strong signal in the Brilliant Violet 711 (BV711) channel. Unstained fecal samples from these mice demonstrated that the BV711 signal was due to bacterial autofluorescence. We found that murine diets containing alfalfa induce ex vivo microbial autofluorescence in the far red spectrum, likely due to chlorophyll. Analysis of unstained intestinal microbes is an important step in microbial flow cytometry to identify diet-induced autofluorescence. We recommend fluorophores with emission spectra below 650 nm (e.g. BV421, PE).


Asunto(s)
Alimentación Animal , Anticuerpos Antibacterianos/inmunología , Heces/microbiología , Citometría de Flujo , Microbioma Gastrointestinal , Imagen Óptica , Animales , Ratones
7.
J Pathol ; 248(3): 339-351, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30883742

RESUMEN

Exon skipping is a promising genetic therapeutic strategy for restoring dystrophin expression in the treatment of Duchenne muscular dystrophy (DMD). The potential for newly synthesized dystrophin to trigger an immune response in DMD patients, however, is not well established. We have evaluated the effect of chronic phosphorodiamidate morpholino oligomer (PMO) treatment on skeletal muscle pathology and asked whether sustained dystrophin expression elicits a dystrophin-specific autoimmune response. Here, two independent cohorts of dystrophic mdx mice were treated chronically with either 800 mg/kg/month PMO for 6 months (n = 8) or 100 mg/kg/week PMO for 12 weeks (n = 11). We found that significant muscle inflammation persisted after exon skipping in skeletal muscle. Evaluation of humoral responses showed serum-circulating antibodies directed against de novo dystrophin in a subset of mice, as assessed both by Western blotting and immunofluorescent staining; however, no dystrophin-specific antibodies were observed in the control saline-treated mdx cohorts (n = 8) or in aged (12-month-old) mdx mice with expanded 'revertant' dystrophin-expressing fibers. Reactive antibodies recognized both full-length and truncated exon-skipped dystrophin isoforms in mouse skeletal muscle. We found more antigen-specific T-cell cytokine responses (e.g. IFN-g, IL-2) in dystrophin antibody-positive mice than in dystrophin antibody-negative mice. We also found expression of major histocompatibility complex class I on some of the dystrophin-expressing fibers along with CD8+ and perforin-positive T cells in the vicinity, suggesting an activation of cell-mediated damage had occurred in the muscle. Evaluation of complement membrane attack complex (MAC) deposition on the muscle fibers further revealed lower MAC deposition on muscle fibers of dystrophin antibody-negative mice than on those of dystrophin antibody-positive mice. Our results indicate that de novo dystrophin expression after exon skipping can trigger both cell-mediated and humoral immune responses in mdx mice. Our data highlights the need to further investigate the autoimmune response and its long-term consequences after exon-skipping therapy. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Distrofina/farmacología , Exones/efectos de los fármacos , Morfolinos/farmacología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Distrofina/genética , Exones/genética , Terapia Genética/métodos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética
8.
Int J Mol Sci ; 20(9)2019 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-31072011

RESUMEN

Group 2 innate lymphoid cells (ILC2) have emerged as a major component of type 2 inflammation in mice and humans. ILC2 secrete large amounts of interleukins 5 and 13, which are largely responsible for host protective immunity against helminth parasites because these cytokines induce profound changes in host physiology that include: goblet cell metaplasia, mucus accumulation, smooth muscle hypercontractility, eosinophil and mast cell recruitment, and alternative macrophage activation (M2). This review covers the initial recognition of ILC2 as a distinct cell lineage, the key studies that established their biological importance, particularly in helminth infection, and the new directions that are likely to be the focus of emerging work that further explores this unique cell population in the context of health and disease.


Asunto(s)
Helmintiasis/inmunología , Helmintos/inmunología , Inmunidad Innata/inmunología , Linfocitos/inmunología , Animales , Linaje de la Célula/inmunología , Helmintiasis/parasitología , Helmintos/patogenicidad , Humanos , Inmunidad Innata/genética , Interleucina-13/genética , Interleucina-5/genética , Ratones
9.
Biochemistry ; 56(1): 96-106, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27959494

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) is a known regulator of the insulin and leptin signaling pathways and is an active target for the design of inhibitors for the treatment of type II diabetes and obesity. Recently, cichoric acid (CHA) and chlorogenic acid (CGA) were predicted by docking methods to be allosteric inhibitors that bind distal to the active site. However, using a combination of steady-state inhibition kinetics, solution nuclear magnetic resonance experiments, and molecular dynamics simulations, we show that CHA is a competitive inhibitor that binds in the active site of PTP1B. CGA, while a noncompetitive inhibitor, binds in the second aryl phosphate binding site, rather than the predicted benzfuran binding pocket. The molecular dynamics simulations of the apo enzyme and cysteine-phosphoryl intermediate states with and without bound CGA suggest CGA binding inhibits PTP1B by altering hydrogen bonding patterns at the active site. This study provides a mechanistic understanding of the allosteric inhibition of PTP1B.


Asunto(s)
Ácidos Cafeicos/farmacología , Ácido Clorogénico/farmacología , Inhibidores Enzimáticos/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Succinatos/farmacología , Algoritmos , Regulación Alostérica , Sitios de Unión , Unión Competitiva , Ácidos Cafeicos/química , Ácidos Cafeicos/metabolismo , Dominio Catalítico , Ácido Clorogénico/química , Ácido Clorogénico/metabolismo , Inhibidores Enzimáticos/metabolismo , Humanos , Enlace de Hidrógeno , Cinética , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Unión Proteica , Dominios Proteicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Succinatos/química , Succinatos/metabolismo
10.
Mucosal Immunol ; 17(2): 238-256, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38336020

RESUMEN

Host defense at the mucosal interface requires collaborative interactions between diverse cell lineages. Epithelial cells damaged by microbial invaders release reparative proteins such as the Trefoil factor family (TFF) peptides that functionally restore barrier integrity. However, whether TFF peptides and their receptors also serve instructive roles for immune cell function during infection is incompletely understood. Here, we demonstrate that the intestinal trefoil factor, TFF3, restrains (T cell helper) TH1 cell proliferation and promotes host-protective type 2 immunity against the gastrointestinal parasitic nematode Trichuris muris. Accordingly, T cell-specific deletion of the TFF3 receptor, leucine-rich repeat and immunoglobulin containing nogo receptor 2 (LINGO2), impairs TH2 cell commitment, allows proliferative expansion of interferon (IFN)g+ cluster of differentiation (CD)4+ TH1 cells and blocks normal worm expulsion through an IFNg-dependent mechanism. This study indicates that TFF3, in addition to its known tissue reparative functions, drives anti-helminth immunity by controlling the balance between TH1/TH2 subsets.


Asunto(s)
Enfermedades Transmisibles , Enfermedades Gastrointestinales , Nematodos , Infecciones por Nematodos , Tricuriasis , Animales , Factor Trefoil-3 , Células TH1 , Linfocitos T Colaboradores-Inductores
11.
Front Immunol ; 14: 997376, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36960049

RESUMEN

At homeostasis, a substantial proportion of Foxp3+ T regulatory cells (Tregs) have an activated phenotype associated with enhanced TCR signals and these effector Treg cells (eTregs) co-express elevated levels of PD-1 and CTLA-4. Short term in vivo blockade of the PD-1 or CTLA-4 pathways results in increased eTreg populations, while combination blockade of both pathways had an additive effect. Mechanistically, combination blockade resulted in a reduction of suppressive phospho-SHP2 Y580 in eTreg cells which was associated with increased proliferation, enhanced production of IL-10, and reduced dendritic cell and macrophage expression of CD80 and MHC-II. Thus, at homeostasis, PD-1 and CTLA-4 function additively to regulate eTreg function and the ability to target these pathways in Treg cells may be useful to modulate inflammation.


Asunto(s)
Receptor de Muerte Celular Programada 1 , Linfocitos T Reguladores , Linfocitos T Reguladores/metabolismo , Antígeno CTLA-4/genética , Receptor de Muerte Celular Programada 1/metabolismo , Antígeno B7-1/metabolismo , Homeostasis
12.
Sci Immunol ; 5(53)2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33188058

RESUMEN

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


Asunto(s)
Células Dendríticas/inmunología , Interleucina-33/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Strongylida/inmunología , Linfocitos T Reguladores/inmunología , Animales , Membrana Celular/metabolismo , Enfermedad Crónica , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Inmunidad Mucosa , Interleucina-33/análisis , Interleucina-33/genética , Masculino , Ratones , Ratones Transgénicos , Mucosa Nasal/inmunología , Mucosa Nasal/patología , Pólipos Nasales/inmunología , Pólipos Nasales/patología , Nematospiroides dubius/inmunología , Nippostrongylus/inmunología , Proteínas Citotóxicas Formadoras de Poros , Rinitis/inmunología , Rinitis/patología , Sinusitis/inmunología , Sinusitis/patología , Infecciones por Strongylida/parasitología , Linfocitos T Reguladores/metabolismo
13.
Biochem Mol Biol Educ ; 45(5): 403-410, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28294503

RESUMEN

Here, we present a 10-week project-oriented laboratory module designed to provide a course-based undergraduate research experience in biochemistry that emphasizes the importance of biomolecular structure and dynamics in enzyme function. This module explores the impact of mutagenesis on an important active site loop for a biomedically-relevant human enzyme, protein tyrosine phosphatase 1B (PTP1B). Over the course of the semester students guide their own mutant of PTP1B from conception to characterization in a cost-effective manner and gain exposure to fundamental techniques in biochemistry, including site-directed DNA mutagenesis, bacterial recombinant protein expression, affinity column purification, protein quantitation, SDS-PAGE, and enzyme kinetics. This project-based approach allows an instructor to simulate a research setting and prepare students for productive research beyond the classroom. Potential modifications to expand or contract this module are also provided. © 2017 by The International Union of Biochemistry and Molecular Biology, 45(5):403-410, 2017.


Asunto(s)
Bioquímica/educación , Laboratorios , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Investigación/educación , Humanos , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 1/aislamiento & purificación , Estudiantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA