Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 611(7937): 787-793, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36323781

RESUMEN

Emerging studies indicate that cooperation between neurons and immune cells regulates antimicrobial immunity, inflammation and tissue homeostasis. For example, a neuronal rheostat provides excitatory or inhibitory signals that control the functions of tissue-resident group 2 innate lymphoid cells (ILC2s) at mucosal barrier surfaces1-4. ILC2s express NMUR1, a receptor for neuromedin U (NMU), which is a prominent cholinergic neuropeptide that promotes ILC2 responses5-7. However, many functions of ILC2s are shared with adaptive lymphocytes, including the production of type 2 cytokines8,9 and the release of tissue-protective amphiregulin (AREG)10-12. Consequently, there is controversy regarding whether innate lymphoid cells and adaptive lymphocytes perform redundant or non-redundant functions13-15. Here we generate a new genetic tool to target ILC2s for depletion or gene deletion in the presence of an intact adaptive immune system. Transgenic expression of iCre recombinase under the control of the mouse Nmur1 promoter enabled ILC2-specific deletion of AREG. This revealed that ILC2-derived AREG promotes non-redundant functions in the context of antiparasite immunity and tissue protection following intestinal damage and inflammation. Notably, NMU expression levels increased in inflamed intestinal tissues from both mice and humans, and NMU induced AREG production in mouse and human ILC2s. These results indicate that neuropeptide-mediated regulation of non-redundant functions of ILC2s is an evolutionarily conserved mechanism that integrates immunity and tissue protection.


Asunto(s)
Inmunidad Innata , Mucosa Intestinal , Linfocitos , Neuropéptidos , Animales , Humanos , Ratones , Citocinas/inmunología , Citocinas/metabolismo , Inmunidad Innata/inmunología , Inflamación/inmunología , Inflamación/parasitología , Inflamación/patología , Linfocitos/inmunología , Neuropéptidos/metabolismo , Neuropéptidos/fisiología , Anfirregulina , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Mucosa Intestinal/patología
2.
Front Immunol ; 13: 900624, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341337

RESUMEN

Influenza A virus (IAV) infections are a significant recurrent threat to public health and a significant burden on global economy, highlighting the need for developing more effective therapies. Natural killer (NK) cells play a pivotal role in the control of pulmonary IAV infection, however, little is known about the therapeutic potential of adoptively transferred NK cells for viral infections. Here, we investigated the antiviral activity of CYNK, human placental hematopoietic stem cell-derived NK cells, against IAV infection in vitro. Virus infection induced the expression of NK cell activating ligands on respiratory epithelial cells, resulting in enhanced recognition by CYNK cells. Upon co-culture with IAV-infected epithelial cells, CYNK exhibited elevated degranulation and increased production of IFN-γ, TNF-α and GM-CSF in a virus dose-dependent manner. Furthermore, CYNK showed virus dose-dependent cytotoxicity against IAV-infected cells. The antiviral activity of CYNK was mediated by NKp46 and NKG2D. Together, these data demonstrate that CYNK possesses potent antiviral function against IAV and warrant clinical investigations for adoptive NK cell therapies against viral infections.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Embarazo , Humanos , Femenino , Placenta , Células Asesinas Naturales/metabolismo , Gripe Humana/metabolismo , Células Madre Hematopoyéticas , Antivirales/metabolismo
3.
PLoS One ; 16(8): e0247738, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34383769

RESUMEN

The commensal microbiota regulates susceptibility to enteric pathogens by fine-tuning mucosal innate immune responses, but how susceptibility to enteric viruses is shaped by the microbiota remains incompletely understood. Past reports have indicated that commensal bacteria may either promote or repress rotavirus replication in the small intestine of mice. We now report that rotavirus replicated more efficiently in the intestines of germ-free and antibiotic-treated mice compared to animals with an unmodified microbiota. Antibiotic treatment also facilitated rotavirus replication in type I and type III interferon (IFN) receptor-deficient mice, revealing IFN-independent proviral effects. Expression of interleukin-22 (IL-22) was strongly diminished in the intestine of antibiotic-treated mice. Treatment with exogenous IL-22 blocked rotavirus replication in microbiota-depleted wild-type and Stat1-/- mice, demonstrating that the antiviral effect of IL-22 in animals with altered microbiome is not dependent on IFN signaling. In antibiotic-treated animals, IL-22-induced a specific set of genes including Fut2, encoding fucosyl-transferase 2 that participates in the biosynthesis of fucosylated glycans which can mediate rotavirus binding. Interestingly, IL-22 also blocked rotavirus replication in antibiotic-treated Fut2-/- mice. Furthermore, IL-22 inhibited rotavirus replication in antibiotic-treated mice lacking key molecules of the necroptosis or pyroptosis pathways of programmed cell death. Taken together, our results demonstrate that IL-22 determines rotavirus susceptibility of antibiotic-treated mice, yet the IL-22-induced effector molecules conferring rotavirus resistance remain elusive.


Asunto(s)
Antibacterianos/efectos adversos , Interleucinas/metabolismo , Infecciones por Rotavirus/etiología , Animales , Antibacterianos/farmacología , Susceptibilidad a Enfermedades , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Perfilación de la Expresión Génica , Interleucinas/fisiología , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Rotavirus/fisiología , Interleucina-22
4.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33741730

RESUMEN

BACKGROUND: Tumors often develop resistance to surveillance by endogenous immune cells, which include natural killer (NK) cells. Ex vivo activated and/or expanded NK cells demonstrate cytotoxicity against various tumor cells and are promising therapeutics for adoptive cancer immunotherapy. Genetic modification can further enhance NK effector cell activity or activation sensitization. Here, we evaluated the effect of the genetic deletion of ubiquitin ligase Casitas B-lineage lymphoma pro-oncogene-b (CBLB), a negative regulator of lymphocyte activity, on placental CD34+ cell-derived NK (PNK) cell cytotoxicity against tumor cells. METHODS: Using CRISPR/Cas9 technology, CBLB was knocked out in placenta-derived CD34+ hematopoietic stem cells, followed by differentiation into PNK cells. Cell expansion, phenotype and cytotoxicity against tumor cells were characterized in vitro. The antitumor efficacy of CBLB knockout (KO) PNK cells was tested in an acute myeloid leukemia (HL-60) tumor model in NOD-scid IL2R gammanull (NSG) mice. PNK cell persistence, biodistribution, proliferation, phenotype and antitumor activity were evaluated. RESULTS: 94% of CBLB KO efficacy was achieved using CRISPR/Cas9 gene editing technology. CBLB KO placental CD34+ cells differentiated into PNK cells with high cell yield and >90% purity determined by CD56+ CD3- cell identity. Ablation of CBLB did not impact cell proliferation, NK cell differentiation or phenotypical characteristics of PNK cells. When compared with the unmodified PNK control, CBLB KO PNK cells exhibited higher cytotoxicity against a range of liquid and solid tumor cell lines in vitro. On infusion into busulfan-conditioned NSG mice, CBLB KO PNK cells showed in vivo proliferation and maturation as evidenced by increased expression of CD16, killer Ig-like receptors and NKG2A over 3 weeks. Additionally, CBLB KO PNK cells showed greater antitumor activity in a disseminated HL60-luciferase mouse model compared with unmodified PNK cells. CONCLUSION: CBLB ablation increased PNK cell effector function and proliferative capacity compared with non-modified PNK cells. These data suggest that targeting CBLB may offer therapeutic advantages via enhancing antitumor activities of NK cell therapies.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas , Citotoxicidad Inmunológica , Inmunoterapia Adoptiva , Células Asesinas Naturales/trasplante , Neoplasias/terapia , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Células Madre , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD34/metabolismo , Proteína 9 Asociada a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Cocultivo , Femenino , Proteínas Ligadas a GPI/metabolismo , Técnicas de Inactivación de Genes , Células HL-60 , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones Endogámicos NOD , Ratones SCID , Subfamília C de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Placenta/citología , Embarazo , Proteínas Proto-Oncogénicas c-cbl/genética , Receptores de IgG/metabolismo , Células Madre/inmunología , Células Madre/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32160524

RESUMEN

Group 2 innate lymphoid cells (ILC2s) regulate immunity, inflammation, and tissue homeostasis. Two distinct subsets of ILC2s have been described: steady-state natural ILC2s and inflammatory ILC2s, which are elicited following helminth infection. However, how tissue-specific cues regulate these two subsets of ILC2s and their effector functions remains elusive. Here, we report that interleukin-33 (IL-33) promotes the generation of inflammatory ILC2s (ILC2INFLAM) via induction of the enzyme tryptophan hydroxylase 1 (Tph1). Tph1 expression was upregulated in ILC2s upon activation with IL-33 or following helminth infection in an IL-33-dependent manner. Conditional deletion of Tph1 in lymphocytes resulted in selective impairment of ILC2INFLAM responses and increased susceptibility to helminth infection. Further, RNA sequencing analysis revealed altered gene expression in Tph1 deficient ILC2s including inducible T cell co-stimulator (Icos). Collectively, these data reveal a previously unrecognized function for IL-33, Tph1, and ICOS in promoting inflammatory ILC2 responses and type 2 immunity at mucosal barriers.


Asunto(s)
Inmunidad Celular , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-33/inmunología , Nippostrongylus/inmunología , Infecciones por Strongylida/inmunología , Subgrupos de Linfocitos T/inmunología , Triptófano Hidroxilasa/inmunología , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Inmunidad Mucosa , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-33/genética , Larva/crecimiento & desarrollo , Larva/inmunología , Larva/patogenicidad , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nippostrongylus/crecimiento & desarrollo , Nippostrongylus/patogenicidad , Cultivo Primario de Células , Transducción de Señal , Infecciones por Strongylida/genética , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología , Subgrupos de Linfocitos T/clasificación , Subgrupos de Linfocitos T/parasitología , Triptófano Hidroxilasa/genética
6.
Mucosal Immunol ; 13(4): 626-636, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32066836

RESUMEN

Thymic stromal lymphopoietin (TSLP) is a critical upstream cytokine inducing type 2 inflammation in various diseases, including asthma and atopic dermatitis. Accumulating evidence suggests that TSLP can directly stimulate a variety of immune cells, such as dendritic cells (DCs), basophils, T cells, and group 2 innate lymphoid cells (ILC2s). However, which cell types directly respond to TSLP in vivo and how TSLP initiates type 2 inflammation has remained controversial. To define the precise role of TSLP in vivo, for the first time we generated multiple cell lineage-specific TSLP receptor-deficient mice to systematically dissect the cell-intrinsic requirements for TSLP responsiveness in type 2 inflammation in the lung. In papain-induced innate immune-mediated type 2 airway inflammation, TSLP directly stimulated ILC2s, but not basophils, leading to enhanced type 2 inflammation. On the other hand, in OVA-induced adaptive immune-mediated type 2 airway inflammation, TSLP principally acted on DCs and CD4 + T cells during the sensitization phase, but not basophils or ILC2s, and facilitated the development of Th2 cell-mediated airway inflammation. Together, these findings reveal that TSLP activates distinct immune cell cascades in the context of innate and adaptive immune-mediated type 2 inflammation.


Asunto(s)
Susceptibilidad a Enfermedades , Técnicas de Silenciamiento del Gen , Inmunoglobulinas/genética , Inflamación/etiología , Inflamación/metabolismo , Receptores de Citocinas/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Inmunidad Adaptativa , Animales , Biomarcadores , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Marcación de Gen , Inmunidad Innata , Inmunohistoquímica , Inflamación/patología , Ratones , Ratones Noqueados , Mucosa Respiratoria/patología
7.
Immunity ; 51(4): 682-695.e6, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31353223

RESUMEN

Innate lymphocytes maintain tissue homeostasis at mucosal barriers, with group 2 innate lymphoid cells (ILC2s) producing type 2 cytokines and controlling helminth infection. While the molecular understanding of ILC2 responses has advanced, the complexity of microenvironmental factors impacting ILC2s is becoming increasingly apparent. Herein, we used single-cell analysis to explore the diversity of gene expression among lung lymphocytes during helminth infection. Following infection, we identified a subset of ILC2s that preferentially expressed Il5-encoding interleukin (IL)-5, together with Calca-encoding calcitonin gene-related peptide (CGRP) and its cognate receptor components. CGRP in concert with IL-33 and neuromedin U (NMU) supported IL-5 but constrained IL-13 expression and ILC2 proliferation. Without CGRP signaling, ILC2 responses and worm expulsion were enhanced. Collectively, these data point to CGRP as a context-dependent negative regulatory factor that shapes innate lymphocyte responses to alarmins and neuropeptides during type 2 innate immune responses.


Asunto(s)
Inflamación/inmunología , Linfocitos/inmunología , Nippostrongylus/fisiología , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Infecciones por Strongylida/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Inmunidad Innata , Interleucina-33/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/metabolismo , Receptores de Péptido Relacionado con el Gen de Calcitonina/genética , Análisis de la Célula Individual , Células Th2/inmunología , Quimera por Trasplante
8.
Elife ; 72018 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-29651984

RESUMEN

Host factors restricting the transmission of respiratory viruses are poorly characterized. We analyzed the contribution of type I and type III interferon (IFN) using a mouse model in which the virus is selectively administered to the upper airways, mimicking a natural respiratory virus infection. Mice lacking functional IFN-λ receptors (Ifnlr1-/-) no longer restricted virus dissemination from the upper airways to the lungs. Ifnlr1-/- mice shed significantly more infectious virus particles via the nostrils and transmitted the virus much more efficiently to naïve contacts compared with wild-type mice or mice lacking functional type I IFN receptors. Prophylactic treatment with IFN-α or IFN-λ inhibited initial virus replication in all parts of the respiratory tract, but only IFN-λ conferred long-lasting antiviral protection in the upper airways and blocked virus transmission. Thus, IFN-λ has a decisive and non-redundant function in the upper airways that greatly limits transmission of respiratory viruses to naïve contacts.


Asunto(s)
Antivirales/farmacología , Interferón gamma/farmacología , Pulmón/efectos de los fármacos , Infecciones por Orthomyxoviridae/prevención & control , Orthomyxoviridae/efectos de los fármacos , Receptores de Interferón/fisiología , Sistema Respiratorio/efectos de los fármacos , Animales , Citocinas/metabolismo , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , Replicación Viral
10.
Nature ; 549(7671): 282-286, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28869965

RESUMEN

The type 2 cytokines interleukin (IL)-4, IL-5, IL-9 and IL-13 have important roles in stimulating innate and adaptive immune responses that are required for resistance to helminth infection, promotion of allergic inflammation, metabolic homeostasis and tissue repair. Group 2 innate lymphoid cells (ILC2s) produce type 2 cytokines, and although advances have been made in understanding the cytokine milieu that promotes ILC2 responses, how ILC2 responses are regulated by other stimuli remains poorly understood. Here we demonstrate that ILC2s in the mouse gastrointestinal tract co-localize with cholinergic neurons that express the neuropeptide neuromedin U (NMU). In contrast to other haematopoietic cells, ILC2s selectively express the NMU receptor 1 (NMUR1). In vitro stimulation of ILC2s with NMU induced rapid cell activation, proliferation, and secretion of the type 2 cytokines IL-5, IL-9 and IL-13 that was dependent on cell-intrinsic expression of NMUR1 and Gαq protein. In vivo administration of NMU triggered potent type 2 cytokine responses characterized by ILC2 activation, proliferation and eosinophil recruitment that was associated with accelerated expulsion of the gastrointestinal nematode Nippostrongylus brasiliensis or induction of lung inflammation. Conversely, worm burden was higher in Nmur1-/- mice than in control mice. Furthermore, use of gene-deficient mice and adoptive cell transfer experiments revealed that ILC2s were necessary and sufficient to mount NMU-elicited type 2 cytokine responses. Together, these data indicate that the NMU-NMUR1 neuronal signalling circuit provides a selective mechanism through which the enteric nervous system and innate immune system integrate to promote rapid type 2 cytokine responses that can induce anti-microbial, inflammatory and tissue-protective type 2 responses at mucosal sites.


Asunto(s)
Citocinas/inmunología , Inmunidad Innata , Inflamación/inmunología , Linfocitos/inmunología , Neuropéptidos/metabolismo , Traslado Adoptivo , Animales , Neuronas Colinérgicas/efectos de los fármacos , Neuronas Colinérgicas/metabolismo , Citocinas/metabolismo , Eosinófilos/citología , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/inervación , Inmunidad Innata/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/patología , Interleucina-13/inmunología , Interleucina-13/metabolismo , Interleucina-5/inmunología , Interleucina-5/metabolismo , Interleucina-9/inmunología , Interleucina-9/metabolismo , Linfocitos/citología , Linfocitos/efectos de los fármacos , Masculino , Ratones , Neuropéptidos/farmacología , Nippostrongylus/inmunología , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/patología , Receptores de Neurotransmisores/deficiencia , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Nature ; 549(7672): 351-356, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28902842

RESUMEN

Type 2 innate lymphoid cells (ILC2s) both contribute to mucosal homeostasis and initiate pathologic inflammation in allergic asthma. However, the signals that direct ILC2s to promote homeostasis versus inflammation are unclear. To identify such molecular cues, we profiled mouse lung-resident ILCs using single-cell RNA sequencing at steady state and after in vivo stimulation with the alarmin cytokines IL-25 and IL-33. ILC2s were transcriptionally heterogeneous after activation, with subpopulations distinguished by expression of proliferative, homeostatic and effector genes. The neuropeptide receptor Nmur1 was preferentially expressed by ILC2s at steady state and after IL-25 stimulation. Neuromedin U (NMU), the ligand of NMUR1, activated ILC2s in vitro, and in vivo co-administration of NMU with IL-25 strongly amplified allergic inflammation. Loss of NMU-NMUR1 signalling reduced ILC2 frequency and effector function, and altered transcriptional programs following allergen challenge in vivo. Thus, NMUR1 signalling promotes inflammatory ILC2 responses, highlighting the importance of neuro-immune crosstalk in allergic inflammation at mucosal surfaces.


Asunto(s)
Hipersensibilidad/inmunología , Hipersensibilidad/patología , Inflamación/inmunología , Inflamación/patología , Pulmón/patología , Linfocitos/inmunología , Neuropéptidos/metabolismo , Animales , Femenino , Regulación de la Expresión Génica , Inmunidad Innata/inmunología , Interleucina-17/inmunología , Interleucina-33/inmunología , Ligandos , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Neurotransmisores/biosíntesis , Receptores de Neurotransmisores/genética , Receptores de Neurotransmisores/metabolismo , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Transducción de Señal , Transcripción Genética
12.
Immunity ; 45(6): 1188-1190, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28002727

RESUMEN

It is known that young children exposed to allergens are prone to develop asthma later in life. In this issue of Immunity, de Kleer et al. (2016) identify IL-33 as a key player in the developing lung for sensitization to environmental allergens and airway hyperreactivity.


Asunto(s)
Alérgenos/inmunología , Hiperreactividad Bronquial , Asma/inmunología , Exposición a Riesgos Ambientales , Humanos , Interleucina-33 , Pulmón/inmunología
13.
Nat Neurosci ; 18(7): 965-77, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26030851

RESUMEN

As the tissue macrophages of the CNS, microglia are critically involved in diseases of the CNS. However, it remains unknown what controls their maturation and activation under homeostatic conditions. We observed substantial contributions of the host microbiota to microglia homeostasis, as germ-free (GF) mice displayed global defects in microglia with altered cell proportions and an immature phenotype, leading to impaired innate immune responses. Temporal eradication of host microbiota severely changed microglia properties. Limited microbiota complexity also resulted in defective microglia. In contrast, recolonization with a complex microbiota partially restored microglia features. We determined that short-chain fatty acids (SCFA), microbiota-derived bacterial fermentation products, regulated microglia homeostasis. Accordingly, mice deficient for the SCFA receptor FFAR2 mirrored microglia defects found under GF conditions. These findings suggest that host bacteria vitally regulate microglia maturation and function, whereas microglia impairment can be rectified to some extent by complex microbiota.


Asunto(s)
Sistema Nervioso Central/fisiología , Ácidos Grasos Volátiles/metabolismo , Homeostasis/fisiología , Inmunidad Innata/fisiología , Microbiota/fisiología , Microglía/fisiología , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Ácidos Grasos Volátiles/inmunología , Femenino , Homeostasis/inmunología , Inmunidad Innata/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/inmunología , Microglía/inmunología , Microglía/metabolismo , Receptores Acoplados a Proteínas G/deficiencia
14.
Nat Immunol ; 16(7): 698-707, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26006013

RESUMEN

The epithelium is the main entry point for many viruses, but the processes that protect barrier surfaces against viral infections are incompletely understood. Here we identified interleukin 22 (IL-22) produced by innate lymphoid cell group 3 (ILC3) as an amplifier of signaling via interferon-λ (IFN-λ), a synergism needed to curtail the replication of rotavirus, the leading cause of childhood gastroenteritis. Cooperation between the receptor for IL-22 and the receptor for IFN-λ, both of which were 'preferentially' expressed by intestinal epithelial cells (IECs), was required for optimal activation of the transcription factor STAT1 and expression of interferon-stimulated genes (ISGs). These data suggested that epithelial cells are protected against viral replication by co-option of two evolutionarily related cytokine networks. These data may inform the design of novel immunotherapy for viral infections that are sensitive to interferons.


Asunto(s)
Citocinas/inmunología , Expresión Génica/inmunología , Interleucinas/inmunología , Infecciones por Rotavirus/inmunología , Animales , Células CACO-2 , Línea Celular , Chlorocebus aethiops , Citocinas/genética , Citocinas/farmacología , Perros , Sinergismo Farmacológico , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Immunoblotting , Interleucinas/genética , Interleucinas/farmacología , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Rotavirus/genética , Infecciones por Rotavirus/virología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Células Vero , Interleucina-22
15.
Nat Commun ; 6: 7090, 2015 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-25987506

RESUMEN

Unrelenting environmental challenges to the gut epithelium place particular demands on the local immune system. In this context, intestinal intraepithelial lymphocytes (IEL) compose a large, highly conserved T cell compartment, hypothesized to provide a first line of defence via cytolysis of dysregulated intestinal epithelial cells (IEC) and cytokine-mediated re-growth of healthy IEC. Here we show that one of the most conspicuous impacts of activated IEL on IEC is the functional upregulation of antiviral interferon (IFN)-responsive genes, mediated by the collective actions of IFNs with other cytokines. Indeed, IEL activation in vivo rapidly provoked type I/III IFN receptor-dependent upregulation of IFN-responsive genes in the villus epithelium. Consistent with this, activated IEL mediators protected cells against virus infection in vitro, and pre-activation of IEL in vivo profoundly limited norovirus infection. Hence, intraepithelial T cell activation offers an overt means to promote the innate antiviral potential of the intestinal epithelium.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Gastroenteritis/inmunología , Activación de Linfocitos , Norovirus/inmunología , Animales , Citocinas/metabolismo , Células Epiteliales/inmunología , Femenino , Gastroenteritis/virología , Inmunidad Innata , Interferón-alfa/metabolismo , Interferón gamma/metabolismo , Interferones/metabolismo , Intestino Delgado/metabolismo , Linfocitos/inmunología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología
16.
PLoS Pathog ; 11(4): e1004782, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25849543

RESUMEN

Epithelial cells are a major port of entry for many viruses, but the molecular networks which protect barrier surfaces against viral infections are incompletely understood. Viral infections induce simultaneous production of type I (IFN-α/ß) and type III (IFN-λ) interferons. All nucleated cells are believed to respond to IFN-α/ß, whereas IFN-λ responses are largely confined to epithelial cells. We observed that intestinal epithelial cells, unlike hematopoietic cells of this organ, express only very low levels of functional IFN-α/ß receptors. Accordingly, after oral infection of IFN-α/ß receptor-deficient mice, human reovirus type 3 specifically infected cells in the lamina propria but, strikingly, did not productively replicate in gut epithelial cells. By contrast, reovirus replicated almost exclusively in gut epithelial cells of IFN-λ receptor-deficient mice, suggesting that the gut mucosa is equipped with a compartmentalized IFN system in which epithelial cells mainly respond to IFN-λ that they produce after viral infection, whereas other cells of the gut mostly rely on IFN-α/ß for antiviral defense. In suckling mice with IFN-λ receptor deficiency, reovirus replicated in the gut epithelium and additionally infected epithelial cells lining the bile ducts, indicating that infants may use IFN-λ for the control of virus infections in various epithelia-rich tissues. Thus, IFN-λ should be regarded as an autonomous virus defense system of the gut mucosa and other epithelial barriers that may have evolved to avoid unnecessarily frequent triggering of the IFN-α/ß system which would induce exacerbated inflammation.


Asunto(s)
Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Leucocitos/inmunología , Infecciones por Reoviridae/inmunología , Animales , Separación Celular , Citometría de Flujo , Humanos , Inmunohistoquímica , Interferón-alfa/inmunología , Interferón beta/inmunología , Interferón gamma/inmunología , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
17.
PLoS One ; 9(6): e96360, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24914933

RESUMEN

Ebola virus (EBOV) is a highly virulent human pathogen. Recovery of infected patients is associated with efficient EBOV-specific immunoglobulin G (IgG) responses, whereas fatal outcome is associated with defective humoral immunity. As B-cell epitopes on EBOV are poorly defined, we sought to identify specific epitopes in four EBOV proteins (Glycoprotein (GP), Nucleoprotein (NP), and matrix Viral Protein (VP)40 and VP35). For the first time, we tested EBOV IgG+ sera from asymptomatic individuals and symptomatic Gabonese survivors, collected during the early humoral response (seven days after the end of symptoms) and the late memory phase (7-12 years post-infection). We also tested sera from EBOV-seropositive patients who had never had clinical signs of hemorrhagic fever or who lived in non-epidemic areas (asymptomatic subjects). We found that serum from asymptomatic individuals was more strongly reactive to VP40 peptides than to GP, NP or VP35. Interestingly, anti-EBOV IgG from asymptomatic patients targeted three immunodominant regions of VP40 reported to play a crucial role in virus assembly and budding. In contrast, serum from most survivors of the three outbreaks, collected a few days after the end of symptoms, reacted mainly with GP peptides. However, in asymptomatic subjects the longest immunodominant domains were identified in GP, and analysis of the GP crystal structure revealed that these domains covered a larger surface area of the chalice bowl formed by three GP1 subunits. The B-cell epitopes we identified in the EBOV VP35, VP40, NP and GP proteins may represent important tools for understanding the humoral response to this virus and for developing new antibody-based therapeutics or detection methods.


Asunto(s)
Linfocitos B/inmunología , Ebolavirus/genética , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/virología , Nucleoproteínas/inmunología , Serogrupo , Proteínas del Núcleo Viral/inmunología , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Brotes de Enfermedades , Ebolavirus/inmunología , Epítopos/genética , Epítopos/inmunología , Femenino , Gabón , Glicoproteínas/química , Glicoproteínas/genética , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Masculino , Datos de Secuencia Molecular , Proteínas de la Nucleocápside , Nucleoproteínas/genética , Proteínas del Núcleo Viral/genética
18.
Mol Cell Biol ; 34(12): 2235-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24710278

RESUMEN

The transcription factor STAT1 is essential for interferon (IFN)-mediated immunity in humans and mice. STAT1 function is tightly regulated, and both loss- and gain-of-function mutations result in severe immune diseases. The two alternatively spliced isoforms, STAT1α and STAT1ß, differ with regard to a C-terminal transactivation domain, which is absent in STAT1ß. STAT1ß is considered to be transcriptionally inactive and to be a competitive inhibitor of STAT1α. To investigate the functions of the STAT1 isoforms in vivo, we generated mice deficient for either STAT1α or STAT1ß. As expected, the functions of STAT1α and STAT1ß in IFN-α/ß- and IFN-λ-dependent antiviral activity are largely redundant. In contrast to the current dogma, however, we found that STAT1ß is transcriptionally active in response to IFN-γ. In the absence of STAT1α, STAT1ß shows more prolonged IFN-γ-induced phosphorylation and promoter binding. Both isoforms mediate protective, IFN-γ-dependent immunity against the bacterium Listeria monocytogenes, although with remarkably different efficiencies. Our data shed new light on the potential contributions of the individual STAT1 isoforms to STAT1-dependent immune responses. Knowledge of STAT1ß's function will help fine-tune diagnostic approaches and help design more specific strategies to interfere with STAT1 activity.


Asunto(s)
Genes Dominantes , Inmunidad Innata/efectos de los fármacos , Interferón gamma/farmacología , Factor de Transcripción STAT1/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/patología , Interferón beta/farmacología , Listeria/efectos de los fármacos , Listeria/fisiología , Listeriosis/inmunología , Listeriosis/patología , Ratones , Muromegalovirus/efectos de los fármacos , Muromegalovirus/fisiología , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Isoformas de Proteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Factor de Transcripción STAT1/deficiencia , Transcripción Genética/efectos de los fármacos
19.
PLoS One ; 9(1): e87906, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498220

RESUMEN

The type III interferon (IFN) receptor is preferentially expressed by epithelial cells. It is made of two subunits: IFNLR1, which is specific to IFN-lambda (IFN-λ) and IL10RB, which is shared by other cytokine receptors. Human hepatocytes express IFNLR1 and respond to IFN-λ. In contrast, the IFN-λ response of the mouse liver is very weak and IFNLR1 expression is hardly detectable in this organ. Here we investigated the IFN-λ response at the cellular level in the mouse liver and we tested whether human and mouse hepatocytes truly differ in responsiveness to IFN-λ. When monitoring expression of the IFN-responsive Mx genes by immunohistofluorescence, we observed that the IFN-λ response in mouse livers was restricted to cholangiocytes, which form the bile ducts, and that mouse hepatocytes were indeed not responsive to IFN-λ. The lack of mouse hepatocyte response to IFN-λ was observed in different experimental settings, including the infection with a hepatotropic strain of influenza A virus which triggered a strong local production of IFN-λ. With the help of chimeric mice containing transplanted human hepatocytes, we show that hepatocytes of human origin readily responded to IFN-λ in a murine environment. Thus, our data suggest that human but not mouse hepatocytes are responsive to IFN-λ in vivo. The non-responsiveness is an intrinsic property of mouse hepatocytes and is not due to the mouse liver micro-environment.


Asunto(s)
Hepatocitos/metabolismo , Interleucinas/metabolismo , Hígado/metabolismo , Receptores de Interferón/metabolismo , Animales , Hepatocitos/citología , Humanos , Interferones , Interleucinas/genética , Hígado/citología , Ratones , Ratones SCID , Ratones Transgénicos , Receptores de Citocinas , Receptores de Interferón/genética , Especificidad de la Especie
20.
PLoS Pathog ; 9(11): e1003773, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278020

RESUMEN

Interferons (IFNs) are a group of cytokines with a well-established antiviral function. They can be induced by viral infection, are secreted and bind to specific receptors on the same or neighbouring cells to activate the expression of hundreds of IFN stimulated genes (ISGs) with antiviral function. Type I IFN has been known for more than half a century. However, more recently, type III IFN (IFNλ, IL-28/29) was shown to play a similar role and to be particularly important at epithelial surfaces. Here we show that airway epithelia, the primary target of influenza A virus, produce both IFN I and III upon infection, and that induction of both depends on the RIG-I/MAVS pathway. While IRF3 is generally regarded as the transcription factor required for initiation of IFN transcription and the so-called "priming loop", we find that IRF3 deficiency has little impact on IFN expression. In contrast, lack of IRF7 reduced IFN production significantly, and only IRF3(-/-)IRF7(-/-) double deficiency completely abolished it. The transcriptional response to influenza infection was largely dependent on IFNs, as it was reduced to a few upregulated genes in epithelia lacking receptors for both type I and III IFN (IFNAR1(-/-)IL-28Rα(-/-)). Wild-type epithelia and epithelia deficient in either the type I IFN receptor or the type III IFN receptor exhibit similar transcriptional profiles in response to virus, indicating that none of the induced genes depends selectively on only one IFN system. In chimeric mice, the lack of both IFN I and III signalling in the stromal compartment alone significantly increased the susceptibility to influenza infection. In conclusion, virus infection of airway epithelia induces, via a RIG-I/MAVS/IRF7 dependent pathway, both type I and III IFNs which drive two completely overlapping and redundant amplification loops to upregulate ISGs and protect from influenza infection.


Asunto(s)
Células Epiteliales/metabolismo , Virus de la Influenza A/metabolismo , Interferón Tipo I/metabolismo , Interleucinas/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Mucosa Respiratoria/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Epiteliales/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interleucinas/genética , Interleucinas/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Receptores de Superficie Celular , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...