Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Immunol ; 9(92): eadf8776, 2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-38394230

RESUMEN

CD8+ T cells are classically recognized as adaptive lymphocytes based on their ability to recognize specific foreign antigens and mount memory responses. However, recent studies indicate that some antigen-inexperienced CD8+ T cells can respond to innate cytokines alone in the absence of cognate T cell receptor stimulation, a phenomenon referred to as bystander activation. Here, we demonstrate that neonatal CD8+ T cells undergo a robust and diverse program of bystander activation, which corresponds to enhanced innate-like protection against unrelated pathogens. Using a multi-omics approach, we found that the ability of neonatal CD8+ T cells to respond to innate cytokines derives from their capacity to undergo rapid chromatin remodeling, resulting in the usage of a distinct set of enhancers and transcription factors typically found in innate-like T cells. We observed that the switch between innate and adaptive functions in the CD8+ T cell compartment is mediated by changes in the abundance of distinct subsets of cells. The innate CD8+ T cell subset that predominates in early life was also present in adult mice and humans. Our findings provide support for the layered immune hypothesis and indicate that the CD8+ T cell compartment is more functionally diverse than previously thought.


Asunto(s)
Linfocitos T CD8-positivos , Inmunidad Innata , Humanos , Adulto , Ratones , Animales , Citocinas , Subgrupos de Linfocitos T , Antígenos
2.
Sci Immunol ; 8(86): eadf8161, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37566678

RESUMEN

Helminth endemic regions report lower COVID-19 morbidity and mortality. Here, we show that lung remodeling from a prior infection with a lung-migrating helminth, Nippostrongylus brasiliensis, enhances viral clearance and survival of human-ACE2 transgenic mice challenged with SARS-CoV-2 (SCV2). This protection is associated with a lymphocytic infiltrate, including increased accumulation of pulmonary SCV2-specific CD8+ T cells, and anti-CD8 antibody depletion abrogated the N. brasiliensis-mediated reduction in viral loads. Pulmonary macrophages with a type 2 transcriptional and epigenetic signature persist in the lungs of N. brasiliensis-exposed mice after clearance of the parasite and establish a primed environment for increased CD8+ T cell recruitment and activation. Accordingly, depletion of macrophages ablated the augmented viral clearance and accumulation of CD8+ T cells driven by prior N. brasiliensis infection. Together, these findings support the concept that lung-migrating helminths can limit disease severity during SCV2 infection through macrophage-dependent enhancement of antiviral CD8+ T cell responses.


Asunto(s)
Linfocitos T CD8-positivos , COVID-19 , Ratones , Humanos , Animales , COVID-19/metabolismo , SARS-CoV-2 , Macrófagos , Pulmón , Ratones Transgénicos
3.
Parasite Immunol ; 45(8): e12999, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37415265

RESUMEN

Intestinal helminth infection promotes a Type 2 inflammatory response in resistant C57BL/6 mice that is essential for worm clearance. The study of inbred mouse strains has revealed factors that are critical for parasite resistance and delineated the role of Type 1 versus Type 2 immune responses in worm clearance. In C57BL/6 mice, basophils are key innate immune cells that promote Type 2 inflammation and are programmed via the Notch signalling pathway during infection with the helminth Trichuris muris. However, how the host genetic background influences basophil responses and basophil expression of Notch receptors remains unclear. Here we use genetically susceptible inbred AKR/J mice that have a Type 1-skewed immune response during T. muris infection to investigate basophil responses in a susceptible host. Basophil population expansion occurred in AKR/J mice even in the absence of fulminant Type 2 inflammation during T. muris infection. However, basophils in AKR/J mice did not robustly upregulate expression of the Notch2 receptor in response to infection as occurred in C57BL/6 mice. Blockade of the Type 1 cytokine interferon-γ in infected AKR/J mice was not sufficient to elicit infection-induced basophil expression of the Notch2 receptor. These data suggest that the host genetic background, outside of the Type 1 skew, is important in regulating basophil responses during T. muris infection in susceptible AKR/J mice.


Asunto(s)
Parásitos , Tricuriasis , Animales , Ratones , Ratones Endogámicos AKR , Trichuris , Basófilos , Receptor Notch2 , Ratones Endogámicos C57BL , Susceptibilidad a Enfermedades , Inflamación
4.
bioRxiv ; 2022 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-36380767

RESUMEN

Helminth endemic regions report lower COVID-19 morbidity and mortality. Here, we show that lung remodeling from a prior infection with a lung migrating helminth, Nippostrongylus brasiliensis , enhances viral clearance and survival of human-ACE2 transgenic mice challenged with SARS-CoV-2 (SCV2). This protection is associated with a lymphocytic infiltrate including an increased accumulation of pulmonary SCV2-specific CD8+ T cells and anti-CD8 antibody depletion abrogated the N. brasiliensis -mediated reduction in viral loads. Pulmonary macrophages with a type-2 transcriptional signature persist in the lungs of N. brasiliensis exposed mice after clearance of the parasite and establish a primed environment for increased antigen presentation. Accordingly, depletion of macrophages ablated the augmented viral clearance and accumulation of CD8+ T cells driven by prior N. brasiliensis infection. Together, these findings support the concept that lung migrating helminths can limit disease severity during SCV2 infection through macrophage-dependent enhancement of anti-viral CD8+ T cell responses.

5.
Mucosal Immunol ; 15(6): 1224-1233, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35732819

RESUMEN

Helminths have evolved sophisticated immune regulating mechanisms to prevent rejection by their mammalian host. Our understanding of how the human immune system responds to these parasites remains poor compared to mouse models of infection and this limits our ability to develop vaccines as well as harness their unique properties as therapeutic strategies against inflammatory disorders. Here, we review how recent studies on human challenge infections, self-infected individuals, travelers, and endemic populations have improved our understanding of human type 2 immunity and its effects on the microbiome. The heterogeneity of responses between individuals and the limited access to tissue samples beyond the peripheral blood are challenges that limit human studies on helminths, but also provide opportunities to transform our understanding of human immunology. Organoids and single-cell sequencing are exciting new tools for immunological analysis that may aid this pursuit. Learning about the genetic and immunological basis of resistance, tolerance, and pathogenesis to helminth infections may thus uncover mechanisms that can be utilized for therapeutic purposes.


Asunto(s)
Helmintiasis , Helmintos , Microbiota , Ratones , Animales , Humanos , Tolerancia Inmunológica , Inmunidad , Interacciones Huésped-Parásitos , Mamíferos
6.
Front Immunol ; 13: 869163, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572520

RESUMEN

Helminth infection currently affect over 2 billion people worldwide, with those with the most pathologies and morbidities, living in regions with unequal and disproportionate access to effective healthcare solutions. Host genetics and environmental factors play critical roles in modulating and regulating immune responses following exposure to various pathogens and insults. However, the interplay of environment and genetic factors in influencing who gets infected and the establishment, persistence, and clearance of helminth parasites remains unclear. Inbred strains of mice have long been used to investigate the role of host genetic factors on pathogenesis and resistance to helminth infection in a laboratory setting. This review will discuss the use of ecological and environmental mouse models to study helminth infections and how this could be used in combination with host genetic variation to explore the relative contribution of these factors in influencing immune response to helminth infections. Improved understanding of interactions between genetics and the environment to helminth immune responses would be important for efforts to identify and develop new prophylactic and therapeutic options for the management of helminth infections and their pathogenesis.


Asunto(s)
Helmintiasis , Helmintos , Parásitos , Animales , Interacciones Huésped-Parásitos/genética , Humanos , Inmunidad , Ratones
7.
J Immunol ; 208(5): 1007-1020, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35181641

RESUMEN

E-protein transcription factors limit group 2 innate lymphoid cell (ILC2) development while promoting T cell differentiation from common lymphoid progenitors. Inhibitors of DNA binding (ID) proteins block E-protein DNA binding in common lymphoid progenitors to allow ILC2 development. However, whether E-proteins influence ILC2 function upon maturity and activation remains unclear. Mice that overexpress ID1 under control of the thymus-restricted proximal Lck promoter (ID1tg/WT) have a large pool of primarily thymus-derived ILC2s in the periphery that develop in the absence of E-protein activity. We used these mice to investigate how the absence of E-protein activity affects ILC2 function and the genomic landscape in response to house dust mite (HDM) allergens. ID1tg/WT mice had increased KLRG1- ILC2s in the lung compared with wild-type (WT; ID1WT/WT) mice in response to HDM, but ID1tg/WT ILC2s had an impaired capacity to produce type 2 cytokines. Analysis of WT ILC2 accessible chromatin suggested that AP-1 and C/EBP transcription factors but not E-proteins were associated with ILC2 inflammatory gene programs. Instead, E-protein binding sites were enriched at functional genes in ILC2s during development that were later dynamically regulated in allergic lung inflammation, including genes that control ILC2 response to cytokines and interactions with T cells. Finally, ILC2s from ID1tg/WT compared with WT mice had fewer regions of open chromatin near functional genes that were enriched for AP-1 factor binding sites following HDM treatment. These data show that E-proteins shape the chromatin landscape during ILC2 development to dictate the functional capacity of mature ILC2s during allergic inflammation in the lung.


Asunto(s)
Antígenos Dermatofagoides/inmunología , Asma/inmunología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Linfocitos T/inmunología , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Alérgenos/inmunología , Animales , Asma/patología , Diferenciación Celular/inmunología , Cromatina/metabolismo , Citocinas/inmunología , Proteínas de Unión al ADN/antagonistas & inhibidores , Femenino , Lectinas Tipo C/genética , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Pyroglyphidae/inmunología , Receptores Inmunológicos/genética , Células Madre/citología , Linfocitos T/citología , Factor de Transcripción AP-1/metabolismo
8.
Am J Physiol Gastrointest Liver Physiol ; 321(6): G668-G681, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34643097

RESUMEN

MicroRNA-mediated regulation is critical for the proper development and function of the small intestinal (SI) epithelium. However, it is not known which microRNAs are expressed in each of the cell types of the SI epithelium. To bridge this important knowledge gap, we performed comprehensive microRNA profiling in all major cell types of the mouse SI epithelium. We used flow cytometry and fluorescence-activated cell sorting with multiple reporter mouse models to isolate intestinal stem cells, enterocytes, goblet cells, Paneth cells, enteroendocrine cells, tuft cells, and secretory progenitors. We then subjected these cell populations to small RNA-sequencing. The resulting atlas revealed highly enriched microRNA markers for almost every major cell type (https://sethupathy-lab.shinyapps.io/SI_miRNA/). Several of these lineage-enriched microRNAs (LEMs) were observed to be embedded in annotated host genes. We used chromatin-run-on sequencing to determine which of these LEMs are likely cotranscribed with their host genes. We then performed single-cell RNA-sequencing to define the cell type specificity of the host genes and embedded LEMs. We observed that the two most enriched microRNAs in secretory progenitors are miR-1224 and miR-672, the latter of which we found is deleted in hominin species. Finally, using several in vivo models, we established that miR-152 is a Paneth cell-specific microRNA.NEW & NOTEWORTHY In this study, first, microRNA atlas (and searchable web server) across all major small intestinal epithelial cell types is presented. We have demonstrated microRNAs that uniquely mark several lineages, including enteroendocrine and tuft. Identification of a key marker of mouse secretory progenitor cells, miR-672, which we show is deleted in humans. We have used several in vivo models to establish miR-152 as a specific marker of Paneth cells, which are highly understudied in terms of microRNAs.


Asunto(s)
Linaje de la Célula , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , MicroARNs/genética , Transcriptoma , Animales , Biomarcadores/metabolismo , Separación Celular , Células Cultivadas , Biología Computacional , Perros , Femenino , Citometría de Flujo , Mucosa Intestinal/citología , Intestino Delgado/citología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Organoides , RNA-Seq , Análisis de la Célula Individual
9.
Eur J Immunol ; 51(10): 2399-2416, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34396535

RESUMEN

Type 2 immunity is critical for the protective and repair responses that mediate resistance to parasitic helminth infection. This immune response also drives aberrant inflammation during atopic diseases. Prostaglandins are a class of critical lipid mediators that are released during type 2 inflammation and are integral in controlling the initiation, activation, maintenance, effector functions, and resolution of Type 2 inflammation. In this review, we explore the roles of the different prostaglandin family members and the receptors they bind to during allergen- and helminth-induced Type 2 inflammation and the mechanism through which prostaglandins promote or suppress Type 2 inflammation. Furthermore, we discuss the potential role of prostaglandins produced by helminth parasites in the regulation of host-pathogen interactions, and how prostaglandins may regulate the inverse relationship between helminth infection and allergy. Finally, we discuss opportunities to capitalize on our understanding of prostaglandin pathways to develop new therapeutic options for humans experiencing Type 2 inflammatory disorders that have a significant prostaglandin-driven component including allergic rhinitis and asthma.


Asunto(s)
Biomarcadores , Susceptibilidad a Enfermedades , Inflamación/etiología , Inflamación/metabolismo , Prostaglandinas/metabolismo , Animales , Manejo de la Enfermedad , Metabolismo Energético , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación/diagnóstico , Inflamación/terapia , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Transducción de Señal
10.
J Exp Med ; 218(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34283207

RESUMEN

Type 2 inflammation is associated with epithelial cell responses, including goblet cell hyperplasia, that promote worm expulsion during intestinal helminth infection. How these epithelial responses are regulated remains incompletely understood. Here, we show that mice deficient in the prostaglandin D2 (PGD2) receptor CRTH2 and mice with CRTH2 deficiency only in nonhematopoietic cells exhibited enhanced worm clearance and intestinal goblet cell hyperplasia following infection with the helminth Nippostrongylus brasiliensis. Small intestinal stem, goblet, and tuft cells expressed CRTH2. CRTH2-deficient small intestinal organoids showed enhanced budding and terminal differentiation to the goblet cell lineage. During helminth infection or in organoids, PGD2 and CRTH2 down-regulated intestinal epithelial Il13ra1 expression and reversed Type 2 cytokine-mediated suppression of epithelial cell proliferation and promotion of goblet cell accumulation. These data show that the PGD2-CRTH2 pathway negatively regulates the Type 2 cytokine-driven epithelial program, revealing a mechanism that can temper the highly inflammatory effects of the anti-helminth response.


Asunto(s)
Citocinas/metabolismo , Mucosa Intestinal/parasitología , Prostaglandina D2/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Infecciones por Strongylida/parasitología , Animales , Femenino , Gastroenteritis/parasitología , Gastroenteritis/patología , Células Caliciformes/patología , Interacciones Huésped-Parásitos/fisiología , Mucosa Intestinal/patología , Masculino , Ratones Endogámicos C57BL , Nippostrongylus/patogenicidad , Organoides , Receptores Inmunológicos/genética , Receptores de Prostaglandina/genética , Infecciones por Strongylida/patología
11.
PLoS Pathog ; 16(10): e1009027, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33108405

RESUMEN

It is of great interest to understand how invading pathogens are sensed within the brain, a tissue with unique challenges to mounting an immune response. The eukaryotic parasite Toxoplasma gondii colonizes the brain of its hosts, and initiates robust immune cell recruitment, but little is known about pattern recognition of T. gondii within brain tissue. The host damage signal IL-33 is one protein that has been implicated in control of chronic T. gondii infection, but, like many other pattern recognition pathways, IL-33 can signal peripherally, and the specific impact of IL-33 signaling within the brain is unclear. Here, we show that IL-33 is expressed by oligodendrocytes and astrocytes during T. gondii infection, is released locally into the cerebrospinal fluid of T. gondii-infected animals, and is required for control of infection. IL-33 signaling promotes chemokine expression within brain tissue and is required for the recruitment and/or maintenance of blood-derived anti-parasitic immune cells, including proliferating, IFN-γ-expressing T cells and iNOS-expressing monocytes. Importantly, we find that the beneficial effects of IL-33 during chronic infection are not a result of signaling on infiltrating immune cells, but rather on radio-resistant responders, and specifically, astrocytes. Mice with IL-33 receptor-deficient astrocytes fail to mount an adequate adaptive immune response in the CNS to control parasite burden-demonstrating, genetically, that astrocytes can directly respond to IL-33 in vivo. Together, these results indicate a brain-specific mechanism by which IL-33 is released locally, and sensed locally, to engage the peripheral immune system in controlling a pathogen.


Asunto(s)
Astrocitos/inmunología , Interleucina-33/inmunología , Toxoplasmosis Cerebral/inmunología , Adulto , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Encéfalo/metabolismo , Femenino , Humanos , Inmunidad , Interferón gamma/inmunología , Interleucina-33/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Transducción de Señal , Toxoplasma/metabolismo , Toxoplasma/parasitología , Toxoplasmosis/metabolismo , Toxoplasmosis Cerebral/metabolismo
12.
Vet Immunol Immunopathol ; 221: 110015, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32058160

RESUMEN

Atopic dermatitis (AD) is an allergic skin disease that causes significant morbidity and affects multiple species. AD is highly prevalent in companion dogs, and the clinical management of the disease remains challenging. An improved understanding of the immunologic and genetic pathways that lead to disease could inform the development of novel treatments. In allergic humans and mouse models of AD, the disease is associated with Th2 and group 2 innate lymphoid cell (ILC2) activation that drives type 2 inflammation. Type 2 inflammation also appears to be associated with AD in dogs, but gaps remain in our understanding of how key type 2-associated cell types such as canine Th2 cells and ILC2s contribute to the pathogenesis of canine AD. Here, we describe previously uncharacterized canine ILC2-like cells and Th2 cells ex vivo that produced type 2 cytokines and expressed the transcription factor Gata3. Increased circulating Th2 cells were associated with chronic canine AD. Single-cell RNA sequencing revealed a unique gene expression signature in T cells in dogs with AD. These findings underline the importance of pro-allergic Th2 cells in orchestrating AD and provide new methods and pathways that can inform the development of improved therapies.


Asunto(s)
Dermatitis Atópica/veterinaria , Enfermedades de los Perros/inmunología , Inmunidad Innata , Linfocitos/inmunología , Células Th2/inmunología , Animales , Células Sanguíneas/inmunología , Dermatitis Atópica/inmunología , Perros , Femenino , Inflamación , Linfocitos/clasificación , Masculino , Análisis de Secuencia de ARN , Análisis de la Célula Individual
13.
J Immunol ; 204(4): 1001-1011, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31900341

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are rare innate immune cells that accumulate in tissues during allergy and helminth infection, performing critical effector functions that drive type 2 inflammation. ILC2s express ST2, the receptor for the cytokine IL-33, and chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2), a receptor for the bioactive lipid prostaglandin D2 (PGD2). The IL-33-ST2 and the PGD2-CRTH2 pathways have both been implicated in promoting ILC2 accumulation during type 2 inflammation. However, whether these two pathways coordinate to regulate ILC2 population size in the tissue in vivo remains undefined. In this study, we show that ILC2 accumulation in the murine lung in response to systemic IL-33 treatment was partially dependent on CRTH2. This effect was not a result of reduced ILC2 proliferation, increased apoptosis or cell death, or differences in expression of the ST2 receptor in the absence of CRTH2. Rather, data from adoptive transfer studies suggested that defective accumulation of CRTH2-deficient ILC2s in response to IL-33 was due to altered ILC2 migration patterns. Whereas donor wild-type ILC2s preferentially accumulated in the lungs compared with CRTH2-deficient ILC2s following transfer into IL-33-treated recipients, wild-type and CRTH2-deficient ILC2s accumulated equally in the recipient mediastinal lymph node. These data suggest that CRTH2-dependent effects lie downstream of IL-33, directly affecting the migration of ILC2s into inflamed lung tissues. A better understanding of the complex interactions between the IL-33 and PGD2-CRTH2 pathways that regulate ILC2 population size will be useful in understanding how these pathways could be targeted to treat diseases associated with type 2 inflammation.


Asunto(s)
Movimiento Celular/inmunología , Hipersensibilidad/inmunología , Interleucina-33/inmunología , Linfocitos/inmunología , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Infecciones por Strongylida/inmunología , Traslado Adoptivo , Animales , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Hipersensibilidad/patología , Inmunidad Innata , Interleucina-33/administración & dosificación , Pulmón/citología , Pulmón/inmunología , Pulmón/patología , Linfocitos/metabolismo , Ratones , Ratones Noqueados , Nippostrongylus/inmunología , Cultivo Primario de Células , Prostaglandina D2/inmunología , Prostaglandina D2/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología
14.
Cell Mol Gastroenterol Hepatol ; 9(3): 447-464, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31756561

RESUMEN

BACKGROUND & AIMS: The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis. METHODS: We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice. RESULTS: First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling. CONCLUSIONS: This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.


Asunto(s)
Células Enteroendocrinas/fisiología , Proteínas Inhibidoras de la Apoptosis/genética , Mucosa Intestinal/fisiología , MicroARNs/metabolismo , Células Madre/fisiología , Animales , Linaje de la Célula/genética , Proliferación Celular/genética , Células Cultivadas , Biología Computacional , Receptores ErbB/metabolismo , Conducta Alimentaria/fisiología , Femenino , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mucosa Intestinal/citología , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Organoides , Cultivo Primario de Células , RNA-Seq , Transducción de Señal/genética , Análisis de la Célula Individual
15.
Cytokine ; 133: 154527, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-30241895

RESUMEN

Parasitic helminth infection elicits a type 2 cytokine-mediated inflammatory response. During type 2 inflammation, damaged or stimulated epithelial cells exposed to helminths and their products produce alarmins and cytokines including IL-25, IL-33, and thymic stromal lymphopoietin. These factors promote innate immune cell activation that supports the polarization of CD4+ T helper type 2 (Th2) cells. Activated innate and Th2 cells produce the cytokines IL-4, -5, -9, and -13 that perpetuate immune activation and act back on the epithelium to cause goblet cell hyperplasia and increased epithelial cell turnover. Together, these events facilitate worm expulsion and wound healing processes. While the role of Th2 cells in this context has been heavily studied, recent work has revealed that epithelial cell-derived cytokines are drivers of key innate immune responses that are critical for type 2 anti-helminth responses. Cutting-edge studies have begun to fully assess how other factors and pathways, including lipid mediators, chemokines, Fc receptor signaling, danger-associated molecular pattern molecules, and direct cell-cell interactions, also participate in shaping innate cell-mediated type 2 inflammation. In this review, we discuss how these pathways intersect and synergize with pathways controlled by epithelial cell-derived cytokines to coordinate innate immune responses that drive helminth-induced type 2 inflammation.


Asunto(s)
Citocinas/inmunología , Helmintiasis/inmunología , Helmintos/inmunología , Inmunidad Innata/inmunología , Animales , Helmintiasis/parasitología , Humanos , Inflamación/inmunología , Inflamación/parasitología , Células Th2/inmunología
16.
J Virol ; 94(1)2019 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-31619551

RESUMEN

Canine parvovirus (CPV) is a highly successful pathogen that has sustained pandemic circulation in dogs for more than 40 years. Here, integrating full-genome and deep-sequencing analyses, structural information, and in vitro experimentation, we describe the macro- and microscale features that accompany CPV's evolutionary success. Despite 40 years of viral evolution, all CPV variants are more than ∼99% identical in nucleotide sequence, with only a limited number (<40) of substitutions becoming fixed or widespread during this time. Notably, most substitutions in the major capsid protein (VP2) gene are nonsynonymous, altering amino acid residues that fall within, or adjacent to, the overlapping receptor footprint or antigenic regions, suggesting that natural selection has channeled much of CPV evolution. Among the limited number of variable sites, CPV genomes exhibit complex patterns of variation that include parallel evolution, reversion, and recombination, compromising phylogenetic inference. At the intrahost level, deep sequencing of viral DNA in original clinical samples from dogs and other host species sampled between 1978 and 2018 revealed few subconsensus single nucleotide variants (SNVs) above ∼0.5%, and experimental passages demonstrate that substantial preexisting genetic variation is not necessarily required for rapid host receptor-driven adaptation. Together, these findings suggest that although CPV is capable of rapid host adaptation, a relatively low mutation rate, pleiotropy, and/or a lack of selective challenges since its initial emergence have inhibited the long-term accumulation of genetic diversity. Hence, continuously high levels of inter- and intrahost diversity are not necessarily required for virus host adaptation.IMPORTANCE Rapid mutation rates and correspondingly high levels of intra- and interhost diversity are often cited as key features of viruses with the capacity for emergence and sustained transmission in a new host species. However, most of this information comes from studies of RNA viruses, with relatively little known about evolutionary processes in viruses with single-stranded DNA (ssDNA) genomes. Here, we provide a unique model of virus evolution, integrating both long-term global-scale and short-term intrahost evolutionary processes of an ssDNA virus that emerged to cause a pandemic in a new host animal. Our analysis reveals that successful host jumping and sustained transmission does not necessarily depend on a high level of intrahost diversity nor result in the continued accumulation of high levels of long-term evolution change. These findings indicate that all aspects of the biology and ecology of a virus are relevant when considering their adaptability.


Asunto(s)
Proteínas de la Cápside/genética , ADN Viral/genética , Enfermedades de los Perros/epidemiología , Genoma Viral , Infecciones por Parvoviridae/veterinaria , Parvovirus Canino/genética , Proteínas no Estructurales Virales/genética , Adaptación Fisiológica/genética , Animales , Evolución Biológica , Proteínas de la Cápside/clasificación , Proteínas de la Cápside/metabolismo , ADN Viral/metabolismo , Enfermedades de los Perros/transmisión , Enfermedades de los Perros/virología , Perros , Zorros/virología , Especificidad del Huésped/genética , Modelos Moleculares , Mutación , Infecciones por Parvoviridae/epidemiología , Infecciones por Parvoviridae/transmisión , Infecciones por Parvoviridae/virología , Parvovirus Canino/clasificación , Parvovirus Canino/patogenicidad , Filogenia , Conformación Proteica , Perros Mapache/virología , Mapaches/virología , Proteínas no Estructurales Virales/clasificación , Proteínas no Estructurales Virales/metabolismo , Secuenciación Completa del Genoma
17.
J Exp Med ; 216(6): 1268-1279, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-30975892

RESUMEN

Type 2 inflammation drives the clearance of gastrointestinal helminth parasites, which infect over two billion people worldwide. Basophils are innate immune cells that support host-protective type 2 inflammation during murine infection with the helminth Trichuris muris However, the mechanisms required for basophil function and gene expression regulation in this context remain unclear. We show that during T. muris infection, basophils localized to the intestine and up-regulated Notch receptor expression, rendering them sensitive to Notch signals that rapidly regulate gene expression programs. In vitro, Notch inhibition limited basophil cytokine production in response to cytokine stimulation. Basophil-intrinsic Notch signaling was required for T. muris-elicited changes in genome-wide basophil transcriptional programs. Mice lacking basophil-intrinsic functional Notch signaling had impaired worm clearance, decreased intestinal type 2 inflammation, altered basophil localization in the intestine, and decreased CD4+ T helper 2 cell responses following infection. These findings demonstrate that Notch is required for basophil gene expression and effector function associated with helminth expulsion during type 2 inflammation.


Asunto(s)
Basófilos/inmunología , Inflamación/patología , Receptores Notch/metabolismo , Transducción de Señal , Animales , Ciego/parasitología , Femenino , Regulación de la Expresión Génica , Inflamación/complicaciones , Interleucinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Trichuris/fisiología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...