Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Immunity ; 52(4): 606-619.e6, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32160524

ABSTRACT

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.


Subject(s)
Immunity, Cellular , Inducible T-Cell Co-Stimulator Protein/immunology , Interleukin-33/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , T-Lymphocyte Subsets/immunology , Tryptophan Hydroxylase/immunology , Animals , Cell Lineage/genetics , Cell Lineage/immunology , Disease Susceptibility , Gene Expression Regulation/immunology , Immunity, Innate , Immunity, Mucosal , Inducible T-Cell Co-Stimulator Protein/genetics , Interleukin-33/genetics , Larva/growth & development , Larva/immunology , Larva/pathogenicity , Lymph Nodes/immunology , Lymph Nodes/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/growth & development , Nippostrongylus/pathogenicity , Primary Cell Culture , Signal Transduction , Strongylida Infections/genetics , Strongylida Infections/parasitology , Strongylida Infections/pathology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/parasitology , Tryptophan Hydroxylase/genetics
2.
Immunity ; 51(4): 682-695.e6, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31353223

ABSTRACT

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.


Subject(s)
Inflammation/immunology , Lymphocytes/immunology , Nippostrongylus/physiology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Strongylida Infections/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Interleukin-33/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/metabolism , Receptors, Calcitonin Gene-Related Peptide/genetics , Single-Cell Analysis , Th2 Cells/immunology , Transplantation Chimera
3.
Nature ; 611(7937): 787-793, 2022 11.
Article in English | MEDLINE | ID: mdl-36323781

ABSTRACT

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.


Subject(s)
Immunity, Innate , Intestinal Mucosa , Lymphocytes , Neuropeptides , Animals , Humans , Mice , Cytokines/immunology , Cytokines/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/parasitology , Inflammation/pathology , Lymphocytes/immunology , Neuropeptides/metabolism , Neuropeptides/physiology , Amphiregulin , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology
4.
Nat Immunol ; 16(7): 698-707, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26006013

ABSTRACT

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.


Subject(s)
Cytokines/immunology , Gene Expression/immunology , Interleukins/immunology , Rotavirus Infections/immunology , Animals , Caco-2 Cells , Cell Line , Chlorocebus aethiops , Cytokines/genetics , Cytokines/pharmacology , Dogs , Drug Synergism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression/drug effects , HT29 Cells , Humans , Immunoblotting , Interleukins/genetics , Interleukins/pharmacology , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/virology , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Reverse Transcriptase Polymerase Chain Reaction , Rotavirus Infections/genetics , Rotavirus Infections/virology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism , Vero Cells , Interleukin-22
5.
Immunity ; 45(6): 1188-1190, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28002727

ABSTRACT

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.


Subject(s)
Allergens/immunology , Bronchial Hyperreactivity , Asthma/immunology , Environmental Exposure , Humans , Interleukin-33 , Lung/immunology
6.
Nature ; 549(7671): 282-286, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28869965

ABSTRACT

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.


Subject(s)
Cytokines/immunology , Immunity, Innate , Inflammation/immunology , Lymphocytes/immunology , Neuropeptides/metabolism , Adoptive Transfer , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cytokines/metabolism , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/immunology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/innervation , Immunity, Innate/drug effects , Inflammation/chemically induced , Inflammation/pathology , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Interleukin-9/immunology , Interleukin-9/metabolism , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Mice , Neuropeptides/pharmacology , Nippostrongylus/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/pathology , Receptors, Neurotransmitter/deficiency , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Signal Transduction/drug effects
8.
Nature ; 549(7672): 351-356, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28902842

ABSTRACT

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.


Subject(s)
Hypersensitivity/immunology , Hypersensitivity/pathology , Inflammation/immunology , Inflammation/pathology , Lung/pathology , Lymphocytes/immunology , Neuropeptides/metabolism , Animals , Female , Gene Expression Regulation , Immunity, Innate/immunology , Interleukin-17/immunology , Interleukin-33/immunology , Ligands , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Neurotransmitter/biosynthesis , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Signal Transduction , Transcription, Genetic
9.
PLoS Pathog ; 11(4): e1004782, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25849543

ABSTRACT

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.


Subject(s)
Epithelial Cells/immunology , Intestinal Mucosa/immunology , Leukocytes/immunology , Reoviridae Infections/immunology , Animals , Cell Separation , Flow Cytometry , Humans , Immunohistochemistry , Interferon-alpha/immunology , Interferon-beta/immunology , Interferon-gamma/immunology , Mammalian orthoreovirus 3/immunology , Mice , Mice, Knockout , Polymerase Chain Reaction
10.
PLoS Pathog ; 9(11): e1003773, 2013.
Article in English | MEDLINE | ID: mdl-24278020

ABSTRACT

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.


Subject(s)
Epithelial Cells/metabolism , Influenza A virus/metabolism , Interferon Type I/metabolism , Interleukins/metabolism , Orthomyxoviridae Infections/metabolism , Respiratory Mucosa/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Influenza A virus/genetics , Influenza A virus/immunology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferon Type I/genetics , Interferon Type I/immunology , Interleukins/genetics , Interleukins/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Receptors, Cell Surface , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
11.
Proc Natl Acad Sci U S A ; 108(19): 7944-9, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518880

ABSTRACT

Type I and type III IFNs bind to different cell-surface receptors but induce identical signal transduction pathways, leading to the expression of antiviral host effector molecules. Despite the fact that type III IFN (IFN-λ) has been shown to predominantly act on mucosal organs, in vivo infection studies have failed to attribute a specific, nonredundant function. Instead, a predominant role of type I IFN was observed, which was explained by the ubiquitous expression of the type I IFN receptor. Here we comparatively analyzed the role of functional IFN-λ and type I IFN receptor signaling in the innate immune response to intestinal rotavirus infection in vivo, and determined viral replication and antiviral gene expression on the cellular level. We observed that both suckling and adult mice lacking functional receptors for IFN-λ were impaired in the control of oral rotavirus infection, whereas animals lacking functional receptors for type I IFN were similar to wild-type mice. Using Mx1 protein accumulation as marker for IFN responsiveness of individual cells, we demonstrate that intestinal epithelial cells, which are the prime target cells of rotavirus, strongly responded to IFN-λ but only marginally to type I IFN in vivo. Systemic treatment of suckling mice with IFN-λ repressed rotavirus replication in the gut, whereas treatment with type I IFN was not effective. These results are unique in identifying a critical role of IFN-λ in the epithelial antiviral host defense.


Subject(s)
Cytokines/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Animals , Immunity, Innate , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Rotavirus/immunology , Rotavirus/physiology , Rotavirus Infections/immunology , Rotavirus Infections/pathology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Signal Transduction/immunology , Virus Replication
12.
J Gen Virol ; 93(Pt 12): 2601-2605, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22956738

ABSTRACT

STAT1-deficient mice are more susceptible to infection with severe acute respiratory syndrome coronavirus (SARS-CoV) than type I interferon (IFN) receptor-deficient mice. We used mice lacking functional receptors for both type I and type III IFN (double knockout, dKO) to evaluate the possibility that type III IFN plays a decisive role in SARS-CoV protection. We found that viral peak titres in lungs of dKO and STAT1-deficient mice were similar, but significantly higher than in wild-type mice. The kinetics of viral clearance from the lung were also comparable in dKO and STAT1-deficient mice. Surprisingly, however, infected dKO mice remained healthy, whereas infected STAT1-deficient mice developed liver pathology and eventually succumbed to neurological disease. Our data suggest that the failure of STAT1-deficient mice to control initial SARS-CoV replication efficiently in the lung is due to impaired type I and type III IFN signalling, whereas the failure to control subsequent systemic viral spread is due to unrelated defects in STAT1-deficient mice.


Subject(s)
Interferon Type I/immunology , Interferons/immunology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Animals , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Severe acute respiratory syndrome-related coronavirus/physiology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Virus Replication/immunology
13.
Front Immunol ; 13: 900624, 2022.
Article in English | MEDLINE | ID: mdl-36341337

ABSTRACT

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.


Subject(s)
Influenza A virus , Influenza, Human , Pregnancy , Humans , Female , Placenta , Killer Cells, Natural/metabolism , Influenza, Human/metabolism , Hematopoietic Stem Cells , Antiviral Agents/metabolism
14.
PLoS One ; 16(8): e0247738, 2021.
Article in English | MEDLINE | ID: mdl-34383769

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/adverse effects , Interleukins/metabolism , Rotavirus Infections/etiology , Animals , Anti-Bacterial Agents/pharmacology , Disease Susceptibility , Female , Gastrointestinal Microbiome/drug effects , Gene Expression Profiling , Interleukins/physiology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Rotavirus/physiology , Interleukin-22
15.
J Immunother Cancer ; 9(3)2021 03.
Article in English | MEDLINE | ID: mdl-33741730

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Cytotoxicity, Immunologic , Immunotherapy, Adoptive , Killer Cells, Natural/transplantation , Neoplasms/therapy , Proto-Oncogene Proteins c-cbl/deficiency , Stem Cells , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD34/metabolism , CRISPR-Associated Protein 9/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Coculture Techniques , Female , GPI-Linked Proteins/metabolism , Gene Knockout Techniques , HL-60 Cells , Humans , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred NOD , Mice, SCID , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Phenotype , Placenta/cytology , Pregnancy , Proto-Oncogene Proteins c-cbl/genetics , Receptors, IgG/metabolism , Stem Cells/immunology , Stem Cells/metabolism , Xenograft Model Antitumor Assays
16.
Mucosal Immunol ; 13(4): 626-636, 2020 07.
Article in English | MEDLINE | ID: mdl-32066836

ABSTRACT

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.


Subject(s)
Disease Susceptibility , Gene Knockdown Techniques , Immunoglobulins/genetics , Inflammation/etiology , Inflammation/metabolism , Receptors, Cytokine/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Adaptive Immunity , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Gene Targeting , Immunity, Innate , Immunohistochemistry , Inflammation/pathology , Mice , Mice, Knockout , Respiratory Mucosa/pathology
17.
Elife ; 72018 04 13.
Article in English | MEDLINE | ID: mdl-29651984

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , Interferon-gamma/pharmacology , Lung/drug effects , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae/drug effects , Receptors, Interferon/physiology , Respiratory System/drug effects , Animals , Cytokines/metabolism , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Respiratory System/immunology , Respiratory System/virology , Virus Replication
18.
Nat Commun ; 6: 7090, 2015 05 19.
Article in English | MEDLINE | ID: mdl-25987506

ABSTRACT

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.


Subject(s)
Caliciviridae Infections/immunology , Gastroenteritis/immunology , Lymphocyte Activation , Norovirus/immunology , Animals , Cytokines/metabolism , Epithelial Cells/immunology , Female , Gastroenteritis/virology , Immunity, Innate , Interferon-alpha/metabolism , Interferon-gamma/metabolism , Interferons/metabolism , Intestine, Small/metabolism , Lymphocytes/immunology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
19.
Nat Neurosci ; 18(7): 965-77, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26030851

ABSTRACT

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.


Subject(s)
Central Nervous System/physiology , Fatty Acids, Volatile/metabolism , Homeostasis/physiology , Immunity, Innate/physiology , Microbiota/physiology , Microglia/physiology , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Fatty Acids, Volatile/immunology , Female , Homeostasis/immunology , Immunity, Innate/immunology , Male , Mice , Mice, Inbred C57BL , Microbiota/immunology , Microglia/immunology , Microglia/metabolism , Receptors, G-Protein-Coupled/deficiency
20.
PLoS One ; 9(6): e96360, 2014.
Article in English | MEDLINE | ID: mdl-24914933

ABSTRACT

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.


Subject(s)
B-Lymphocytes/immunology , Ebolavirus/genetics , Glycoproteins/immunology , Hemorrhagic Fever, Ebola/virology , Nucleoproteins/immunology , Serogroup , Viral Core Proteins/immunology , Adolescent , Adult , Amino Acid Sequence , Child , Disease Outbreaks , Ebolavirus/immunology , Epitopes/genetics , Epitopes/immunology , Female , Gabon , Glycoproteins/chemistry , Glycoproteins/genetics , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/epidemiology , Humans , Male , Molecular Sequence Data , Nucleocapsid Proteins , Nucleoproteins/genetics , Viral Core Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL