Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38604478

ABSTRACT

Intestinal epithelial cell (IEC) responses to interferon (IFN) favor antiviral defense with minimal cytotoxicity, but IEC-specific factors that regulate these responses remain poorly understood. Interferon regulatory factors (IRFs) are a family of nine related transcription factors, and IRF6 is preferentially expressed by epithelial cells, but its roles in IEC immunity are unknown. In this study, Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) screens found that Irf6 deficiency enhanced IFN-stimulated antiviral responses in transformed mouse IECs but not macrophages. Furthermore, knockout (KO) of Irf6 in IEC organoids resulted in profound changes to homeostasis and immunity gene expression. Irf6 KO organoids grew more slowly, and single-cell ribonucleic acid sequencing indicated reduced expression of genes in epithelial differentiation and immunity pathways. IFN-stimulated gene expression was also significantly different in Irf6 KO organoids, with increased expression of stress and apoptosis-associated genes. Functionally, the transcriptional changes in Irf6 KO organoids were associated with increased cytotoxicity upon IFN treatment or inflammasome activation. These data indicate a previously unappreciated role for IRF6 in IEC biology, including regulation of epithelial development and moderation of innate immune responses to minimize cytotoxicity and maintain barrier function.

2.
Curr Opin Immunol ; 86: 102412, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38518661

ABSTRACT

Interferon (IFN) was discovered based on interference with virus production, and three types of IFN are now defined. Since its discovery, IFN's roles have expanded beyond viruses to diverse pathogen types, tissue homeostasis, and inflammatory disease. The gastrointestinal (GI) tract is arguably the tissue where the roles of IFN types are most distinct, with a particularly prominent role for type-III IFN in antiviral protection of the intestinal epithelium. Current studies continue to deepen our understanding of the type- and tissue-specific roles of IFN. This review highlights these advances within the GI tract, including discovery of protective roles for type-III IFNs against nonviral GI pathogens, and discovery of an antiviral homeostatic type-III IFN response within the intestinal epithelium.


Subject(s)
Interferon Type I , Viruses , Humans , Interferon Lambda , Interferons , Gastrointestinal Tract , Antiviral Agents
3.
Immunity ; 56(8): 1862-1875.e9, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37478853

ABSTRACT

Loss of oral tolerance (LOT) to gluten, driven by dendritic cell (DC) priming of gluten-specific T helper 1 (Th1) cell immune responses, is a hallmark of celiac disease (CeD) and can be triggered by enteric viral infections. Whether certain commensals can moderate virus-mediated LOT remains elusive. Here, using a mouse model of virus-mediated LOT, we discovered that the gut-colonizing protist Tritrichomonas (T.) arnold promotes oral tolerance and protects against reovirus- and murine norovirus-mediated LOT, independent of the microbiota. Protection was not attributable to antiviral host responses or T. arnold-mediated innate type 2 immunity. Mechanistically, T. arnold directly restrained the proinflammatory program in dietary antigen-presenting DCs, subsequently limiting Th1 and promoting regulatory T cell responses. Finally, analysis of fecal microbiomes showed that T. arnold-related Parabasalid strains are underrepresented in human CeD patients. Altogether, these findings will motivate further exploration of oral-tolerance-promoting protists in CeD and other immune-mediated food sensitivities.


Subject(s)
Antigens , Immunity, Innate , Animals , Mice , Humans , Diet , Glutens , Dendritic Cells , Immune Tolerance
4.
Cell Rep ; 42(5): 112407, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37083328

ABSTRACT

Poxvirus infections of the skin are a recent emerging public health concern, yet the mechanisms that mediate protective immunity against these viral infections remain largely unknown. Here, we show that T helper 1 (Th1) memory CD4+ T cells are necessary and sufficient to provide complete and broad protection against poxvirus skin infections, whereas memory CD8+ T cells are dispensable. Core 2 O-glycan-synthesizing Th1 effector memory CD4+ T cells rapidly infiltrate the poxvirus-infected skin microenvironment and produce interferon γ (IFNγ) in an antigen-dependent manner, causing global changes in gene expression to promote anti-viral immunity. Keratinocytes express IFN-stimulated genes, upregulate both major histocompatibility complex (MHC) class I and MHC class II antigen presentation in an IFNγ-dependent manner, and require IFNγ receptor (IFNγR) signaling and MHC class II expression for memory CD4+ T cells to protect the skin from poxvirus infection. Thus, Th1 effector memory CD4+ T cells exhibit potent anti-viral activity within the skin, and keratinocytes are the key targets of IFNγ necessary for preventing poxvirus infection of the epidermis.


Subject(s)
CD4-Positive T-Lymphocytes , Poxviridae Infections , Humans , CD8-Positive T-Lymphocytes , Skin/metabolism , Histocompatibility Antigens Class II , Histocompatibility Antigens Class I , Interferon-gamma
5.
Immunohorizons ; 6(7): 416-429, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35790340

ABSTRACT

The three types of IFN have roles in antimicrobial immunity and inflammation that must be properly balanced to maintain tissue homeostasis. For example, IFNs are elevated in the context of inflammatory bowel disease and may synergize with inflammatory cytokines such as TNF-α to promote tissue damage. Prior studies suggest that in mouse intestinal epithelial cells (IECs), type III IFNs are preferentially produced during viral infections and are less cytotoxic than type I IFN. In this study, we generated human IEC organoid lines from biopsies of ileum, ascending colon, and sigmoid colon of three healthy subjects to establish the baseline responses of normal human IECs to types I, II, and III IFN. We found that all IFN types elicited responses that were qualitatively consistent across intestinal biopsy sites. However, IFN types differed in magnitude of STAT1 phosphorylation and identity of genes in their downstream transcriptional programs. Specifically, there was a core transcriptional module shared by IFN types, but types I and II IFN stimulated unique transcriptional modules beyond this core gene signature. The transcriptional modules of type I and II IFN included proapoptotic genes, and expression of these genes correlated with potentiation of TNF-α cytotoxicity. These data define the response profiles of healthy human IEC organoids across IFN types, and they suggest that cytotoxic effects mediated by TNF-α in inflamed tissues may be amplified by a simultaneous high-magnitude IFN response.


Subject(s)
Organoids , Tumor Necrosis Factor-alpha , Animals , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Intestines , Mice , Organoids/metabolism
6.
Infect Immun ; 90(7): e0066321, 2022 07 21.
Article in English | MEDLINE | ID: mdl-35678562

ABSTRACT

Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that causes diseases ranging from gastroenteritis to systemic infection and sepsis. Salmonella uses type III secretion systems (T3SS) to inject effectors into host cells. While these effectors are necessary for bacterial invasion and intracellular survival, intracellular delivery of T3SS products also enables detection of translocated Salmonella ligands by cytosolic immune sensors. Some of these sensors form multimeric complexes called inflammasomes, which activate caspases that lead to interleukin-1 (IL-1) family cytokine release and pyroptosis. In particular, the Salmonella T3SS needle, inner rod, and flagellin proteins activate the NAIP/NLRC4 inflammasome in murine intestinal epithelial cells (IECs), which leads to restriction of bacterial replication and extrusion of infected IECs into the intestinal lumen, thereby preventing systemic dissemination of Salmonella. While these processes are quite well studied in mice, the role of the NAIP/NLRC4 inflammasome in human IECs remains unknown. Unexpectedly, we found the NAIP/NLRC4 inflammasome is dispensable for early inflammasome responses to Salmonella in both human IEC lines and enteroids. Additionally, NLRP3 and the adaptor protein ASC are not required for inflammasome activation in Caco-2 cells. Instead, we observed a necessity for caspase-4 and gasdermin D pore-forming activity in mediating inflammasome responses to Salmonella in Caco-2 cells. These findings suggest that unlike murine IECs, human IECs do not rely on NAIP/NLRC4 or NLRP3/ASC inflammasomes and instead primarily use caspase-4 to mediate inflammasome responses to Salmonella pathogenicity island 1 (SPI-1)-expressing Salmonella.


Subject(s)
Inflammasomes , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Caco-2 Cells , Calcium-Binding Proteins , Caspases, Initiator , Epithelial Cells/metabolism , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuronal Apoptosis-Inhibitory Protein , Salmonella typhimurium , Serogroup
7.
Elife ; 112022 02 09.
Article in English | MEDLINE | ID: mdl-35137688

ABSTRACT

Interferon-lambda (IFN-λ) protects intestinal epithelial cells (IECs) from enteric viruses by inducing expression of antiviral IFN-stimulated genes (ISGs). Here, we find that bacterial microbiota stimulate a homeostatic ISG signature in the intestine of specific pathogen-free mice. This homeostatic ISG expression is restricted to IECs, depends on IEC-intrinsic expression of IFN-λ receptor (Ifnlr1), and is associated with IFN-λ production by leukocytes. Strikingly, imaging of these homeostatic ISGs reveals localization to pockets of the epithelium and concentration in mature IECs. Correspondingly, a minority of mature IECs express these ISGs in public single-cell RNA sequencing datasets from mice and humans. Furthermore, we assessed the ability of orally administered bacterial components to restore localized ISGs in mice lacking bacterial microbiota. Lastly, we find that IECs lacking Ifnlr1 are hyper-susceptible to initiation of murine rotavirus infection. These observations indicate that bacterial microbiota stimulate ISGs in localized regions of the intestinal epithelium at homeostasis, thereby preemptively activating antiviral defenses in vulnerable IECs to improve host defense against enteric viruses.


Subject(s)
Enterovirus/physiology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/immunology , Receptors, Interferon/genetics , Animals , Bacterial Physiological Phenomena , Female , Homeostasis , Male , Mice , Receptors, Interferon/metabolism
8.
Front Immunol ; 12: 767505, 2021.
Article in English | MEDLINE | ID: mdl-34712246

ABSTRACT

Interferon λ (IFN-λ) is critical for host viral defense at mucosal surfaces and stimulates immunomodulatory signals, acting on epithelial cells and few other cell types due to restricted IFN-λ receptor expression. Epithelial cells of the intestine play a critical role in the pathogenesis of Inflammatory Bowel Disease (IBD), and the related type II interferons (IFN-γ) have been extensively studied in the context of IBD. However, a role for IFN-λ in IBD onset and progression remains unclear. Recent investigations of IFN-λ in IBD are beginning to uncover complex and sometimes opposing actions, including pro-healing roles in colonic epithelial tissues and potentiation of epithelial cell death in the small intestine. Additionally, IFN-λ has been shown to act through non-epithelial cell types, such as neutrophils, to protect against excessive inflammation. In most cases IFN-λ demonstrates an ability to coordinate the host antiviral response without inducing collateral hyperinflammation, suggesting that IFN-λ signaling pathways could be a therapeutic target in IBD. This mini review discusses existing data on the role of IFN-λ in the pathogenesis of inflammatory bowel disease, current gaps in the research, and therapeutic potential of modulating the IFN-λ-stimulated response.


Subject(s)
Epithelial Cells/immunology , Immunity, Innate/immunology , Inflammatory Bowel Diseases/immunology , Interferons/immunology , Intestinal Mucosa/immunology , Signal Transduction/immunology , Animals , Apoptosis/immunology , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Interferons/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Models, Immunological , Protein Isoforms/immunology , Protein Isoforms/metabolism , STAT Transcription Factors/immunology , STAT Transcription Factors/metabolism , Tight Junctions/immunology , Tight Junctions/metabolism , Interferon Lambda
9.
Microbiology (Reading) ; 167(10)2021 10.
Article in English | MEDLINE | ID: mdl-34698627

ABSTRACT

Bacterial small RNAs (sRNAs) are important regulators of gene expression; however, the impact of natural mutations on sRNA functions has not been studied extensively. Here we show that the sRNA MgrR contains a unique 53 bp insertion in Escherichia fergusonii, a close relative of Escherichia coli and Salmonella enterica. The insertion is a repetitive extragenic palindromic (REP) sequence that could block transcription, but full-length MgrR is produced in E. fergusonii, showing that the insertion has not affected sRNA production. Additionally, despite containing the large insertion, the sRNA appears to be functional because deletion of mgrR made E. fergusonii more susceptible to H2O2. The molecular details of MgrR's roles in H2O2defence are yet to be defined, but our results suggest that having an alternative function allowed the sRNA to be retained in E. fergusonii despite it sustaining a large, potentially disruptive mutation.


Subject(s)
Escherichia/genetics , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Enterobacteriaceae/classification , Enterobacteriaceae/genetics , Escherichia/classification , Escherichia/metabolism , Gene Expression Regulation, Bacterial , Hydrogen Peroxide/metabolism , Magnesium/metabolism , Mutation , Phylogeny , RNA, Bacterial/metabolism , RNA, Small Untranslated/metabolism
10.
Curr Opin Immunol ; 73: 1-8, 2021 12.
Article in English | MEDLINE | ID: mdl-34392232

ABSTRACT

Epithelial cells in barrier tissues perform a critical immune function by detecting, restricting, and often directly eliminating extrinsic pathogens. Membrane-bound and cytosolic pattern recognition receptors in epithelial cells bind to diverse ligands, detecting pathogen components and behaviors and stimulating cell-autonomous immunity. In addition to directly acting as first-responders to pathogens, epithelial cells detect commensal-derived and diet-derived products to promote homeostasis. Recent advances have clarified the array of molecular sensors expressed by epithelial cells, and how epithelial cells responses are wired to promote homeostatic balance while simultaneously allowing elimination of pathogens. These new studies emphatically position epithelial cells as central to an effective innate immune response.


Subject(s)
Epithelial Cells/immunology , Immunity, Innate , Receptors, Pattern Recognition/metabolism , Animals , Homeostasis , Humans , Pathogen-Associated Molecular Pattern Molecules/immunology , Symbiosis
11.
PLoS Pathog ; 17(3): e1009402, 2021 03.
Article in English | MEDLINE | ID: mdl-33705489

ABSTRACT

Interferons (IFNs) are key controllers of viral replication, with intact IFN responses suppressing virus growth and spread. Using the murine norovirus (MNoV) system, we show that IFNs exert selective pressure to limit the pathogenic evolutionary potential of this enteric virus. In animals lacking type I IFN signaling, the nonlethal MNoV strain CR6 rapidly acquired enhanced virulence via conversion of a single nucleotide. This nucleotide change resulted in amino acid substitution F514I in the viral capsid, which led to >10,000-fold higher replication in systemic organs including the brain. Pathogenicity was mediated by enhanced recruitment and infection of intestinal myeloid cells and increased extraintestinal dissemination of virus. Interestingly, the trade-off for this mutation was reduced fitness in an IFN-competent host, in which CR6 bearing F514I exhibited decreased intestinal replication and shedding. In an immunodeficient context, a spontaneous amino acid change can thus convert a relatively avirulent viral strain into a lethal pathogen.


Subject(s)
Caliciviridae Infections/virology , Capsid Proteins/genetics , Norovirus/genetics , Norovirus/pathogenicity , Virulence/genetics , Animals , Caliciviridae Infections/genetics , Caliciviridae Infections/immunology , Genetic Fitness/genetics , Immunity, Innate/immunology , Mice , Norovirus/immunology , Polymorphism, Single Nucleotide , Virulence/immunology , Virus Replication
12.
J Virol ; 95(3)2021 01 13.
Article in English | MEDLINE | ID: mdl-33177207

ABSTRACT

Noroviruses are a leading cause of gastrointestinal infection in humans and mice. Understanding human norovirus (HuNoV) cell tropism has important implications for our understanding of viral pathogenesis. Murine norovirus (MNoV) is extensively used as a surrogate model for HuNoV. We previously identified CD300lf as the receptor for MNoV. Here, we generated a Cd300lf conditional knockout (CD300lfF/F ) mouse to elucidate the cell tropism of persistent and nonpersistent strains of murine norovirus. Using this mouse model, we demonstrated that CD300lf expression on intestinal epithelial cells (IECs), and on tuft cells in particular, is essential for transmission of the persistent MNoV strain CR6 (MNoVCR6) in vivo In contrast, the nonpersistent MNoV strain CW3 (MNoVCW3) does not require CD300lf expression on IECs for infection. However, deletion of CD300lf in myelomonocytic cells (LysM Cre+) partially reduces CW3 viral load in lymphoid and intestinal tissues. Disruption of CD300lf expression on B cells (CD19 Cre), neutrophils (Mrp8 Cre), and dendritic cells (CD11c Cre) did not affect MNoVCW3 viral RNA levels. Finally, we show that the transcription factor STAT1, which is critical for the innate immune response, partially restricts the cell tropism of MNoVCW3 to LysM+ cells. Taken together, these data demonstrate that CD300lf expression on tuft cells is essential for MNoVCR6; that myelomonocytic cells are a major, but not exclusive, target cell of MNoVCW3; and that STAT1 signaling restricts the cellular tropism of MNoVCW3 This study provides the first genetic system for studying the cell type-specific role of CD300lf in norovirus pathogenesis.IMPORTANCE Human noroviruses (HuNoVs) are a leading cause of gastroenteritis resulting in up to 200,000 deaths each year. The receptor and cell tropism of HuNoV in immunocompetent humans are unclear. We use murine norovirus (MNoV) as a model for HuNoV. We recently identified CD300lf as the sole physiologic receptor for MNoV. Here, we leverage this finding to generate a Cd300lf conditional knockout mouse to decipher the contributions of specific cell types to MNoV infection. We demonstrate that persistent MNoVCR6 requires CD300lf expression on tuft cells. In contrast, multiple CD300lf+ cell types, dominated by myelomonocytic cells, are sufficient for nonpersistent MNoVCW3 infection. CD300lf expression on epithelial cells, B cells, neutrophils, and dendritic cells is not critical for MNoVCW3 infection. Mortality associated with the MNoVCW3 strain in Stat1-/- mice does not require CD300lf expression on LysM+ cells, highlighting that both CD300lf receptor expression and innate immunity regulate MNoV cell tropism in vivo.


Subject(s)
Epithelial Cells/immunology , Host-Pathogen Interactions , Immunity, Innate/immunology , Intestines/immunology , Norovirus/physiology , Receptors, Immunologic/physiology , Viral Tropism , Animals , Caliciviridae Infections/immunology , Caliciviridae Infections/metabolism , Caliciviridae Infections/virology , Epithelial Cells/virology , Female , Intestines/virology , Male , Mice , Mice, Knockout
13.
J Virol ; 94(21)2020 10 14.
Article in English | MEDLINE | ID: mdl-32847859

ABSTRACT

Interferon (IFN) family cytokines stimulate genes (interferon-stimulated genes [ISGs]) that are integral to antiviral host defense. Type I IFNs act systemically, whereas type III IFNs act preferentially at epithelial barriers. Among barrier cells, intestinal epithelial cells (IECs) are particularly dependent on type III IFN for the control and clearance of virus infection, but the physiological basis of this selective IFN response is not well understood. Here, we confirm that type III IFN treatment elicits robust and uniform ISG expression in neonatal mouse IECs and inhibits the replication of IEC-tropic rotavirus. In contrast, type I IFN elicits a marginal ISG response in neonatal mouse IECs and does not inhibit rotavirus replication. In vitro treatment of IEC organoids with type III IFN results in ISG expression that mirrors the in vivo type III IFN response. However, IEC organoids have increased expression of the type I IFN receptor relative to neonate IECs, and the response of IEC organoids to type I IFN is strikingly increased in magnitude and scope relative to type III IFN. The expanded type I IFN-specific response includes proapoptotic genes and potentiates toxicity triggered by tumor necrosis factor alpha (TNF-α). The ISGs stimulated in common by type I and III IFNs have strong interferon-stimulated response element (ISRE) promoter motifs, whereas the expanded set of type I IFN-specific ISGs, including proapoptotic genes, have weak ISRE motifs. Thus, the preferential responsiveness of IECs to type III IFN in vivo enables selective ISG expression during infection that confers antiviral protection but minimizes disruption of intestinal homeostasis.IMPORTANCE Enteric viral infections are a major cause of gastroenteritis worldwide and have the potential to trigger or exacerbate intestinal inflammatory diseases. Prior studies have identified specialized innate immune responses that are active in the intestinal epithelium following viral infection, but our understanding of the benefits of such an epithelium-specific response is incomplete. Here, we show that the intestinal epithelial antiviral response is programmed to enable protection while minimizing epithelial cytotoxicity that can often accompany an inflammatory response. Our findings offer new insight into the benefits of a tailored innate immune response at the intestinal barrier and suggest how dysregulation of this response could promote inflammatory disease.


Subject(s)
Cytokines/immunology , Intestinal Mucosa/immunology , Rotavirus Infections/immunology , STAT1 Transcription Factor/immunology , STAT2 Transcription Factor/immunology , Tumor Necrosis Factor-alpha/toxicity , Animals , Animals, Newborn , Cytokines/genetics , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Regulation , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Organoids/drug effects , Organoids/immunology , Organoids/virology , Response Elements , Rotavirus/drug effects , Rotavirus/growth & development , Rotavirus/pathogenicity , Rotavirus Infections/genetics , Rotavirus Infections/pathology , Rotavirus Infections/virology , STAT1 Transcription Factor/genetics , STAT2 Transcription Factor/genetics , Signal Transduction , Virus Replication
14.
Cell ; 179(3): 644-658.e13, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31607511

ABSTRACT

Rotavirus (RV) encounters intestinal epithelial cells amidst diverse microbiota, opening possibilities of microbes influencing RV infection. Although RV clearance typically requires adaptive immunity, we unintentionally generated RV-resistant immunodeficient mice, which, we hypothesized, reflected select microbes protecting against RV. Accordingly, such RV resistance was transferred by co-housing and fecal transplant. RV-protecting microbiota were interrogated by heat, filtration, and antimicrobial agents, followed by limiting dilution transplant to germ-free mice and microbiome analysis. This approach revealed that segmented filamentous bacteria (SFB) were sufficient to protect mice against RV infection and associated diarrhea. Such protection was independent of previously defined RV-impeding factors, including interferon, IL-17, and IL-22. Colonization of the ileum by SFB induced changes in host gene expression and accelerated epithelial cell turnover. Incubation of RV with SFB-containing feces reduced infectivity in vitro, suggesting direct neutralization of RV. Thus, independent of immune cells, SFB confer protection against certain enteric viral infections and associated diarrheal disease.


Subject(s)
Adaptive Immunity/genetics , Diarrhea/microbiology , Intestinal Mucosa/microbiology , Rotavirus Infections/microbiology , Animals , Anti-Infective Agents/pharmacology , Bacteria/genetics , Bacteria/metabolism , Diarrhea/prevention & control , Diarrhea/virology , Feces/microbiology , Gene Expression Regulation/genetics , Humans , Ileum/microbiology , Ileum/pathology , Ileum/virology , Interferons/genetics , Interleukin-17/genetics , Interleukins/genetics , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Mice , Microbiota/genetics , Rotavirus/pathogenicity , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Interleukin-22
15.
PLoS Pathog ; 15(7): e1007940, 2019 07.
Article in English | MEDLINE | ID: mdl-31329638

ABSTRACT

Human norovirus (HNoV) is the leading cause of acute gastroenteritis and is spread by fecal shedding that can often persist for weeks to months after the resolution of symptoms. Elimination of persistent viral reservoirs has the potential to prevent outbreaks. Similar to HNoV, murine norovirus (MNV) is spread by persistent shedding in the feces and provides a tractable model to study molecular mechanisms of enteric persistence. Previous studies have identified non-structural protein 1 (NS1) from the persistent MNV strain CR6 as critical for persistent infection in intestinal epithelial cells (IECs), but its mechanism of action remains unclear. We now find that the function of CR6 NS1 is regulated by apoptotic caspase cleavage. Following induction of apoptosis in infected cells, caspases cleave the precursor NS1/2 protein, and this cleavage is prevented by mutation of caspase target motifs. These mutations profoundly compromise CR6 infection of IECs and persistence in the intestine. Conversely, NS1/2 cleavage is not strictly required for acute replication in extra-intestinal tissues or in cultured myeloid cells, suggesting an IEC-centric role. Intriguingly, we find that caspase cleavage of CR6 NS1/2 reciprocally promotes caspase activity, potentiates cell death, and amplifies spread among cultured IEC monolayers. Together, these data indicate that the function of CR6 NS1 is regulated by apoptotic caspases, and suggest that apoptotic cell death enables epithelial spread and persistent shedding.


Subject(s)
Intestinal Mucosa/virology , Norovirus/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , Apoptosis , Caliciviridae Infections/etiology , Caliciviridae Infections/pathology , Caliciviridae Infections/virology , Caspases/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Gastroenteritis/etiology , Gastroenteritis/pathology , Gastroenteritis/virology , Host Microbial Interactions , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Models, Biological , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloid Cells/virology , Norovirus/genetics , Norovirus/physiology , Viral Nonstructural Proteins/genetics , Virus Replication , Virus Shedding
16.
Nat Microbiol ; 4(10): 1737-1749, 2019 10.
Article in English | MEDLINE | ID: mdl-31182797

ABSTRACT

Products derived from bacterial members of the gut microbiota evoke immune signalling pathways of the host that promote immunity and barrier function in the intestine. How immune reactions to enteric viruses support intestinal homeostasis is unknown. We recently demonstrated that infection by murine norovirus (MNV) reverses intestinal abnormalities following depletion of bacteria, indicating that an intestinal animal virus can provide cues to the host that are typically attributed to the microbiota. Here, we elucidate mechanisms by which MNV evokes protective responses from the host. We identify an important role for the viral protein NS1/2 in establishing local replication and a type I interferon (IFN-I) response in the colon. We further show that IFN-I acts on intestinal epithelial cells to increase the proportion of CCR2-dependent macrophages and interleukin (IL)-22-producing innate lymphoid cells, which in turn promote pSTAT3 signalling in intestinal epithelial cells and protection from intestinal injury. In addition, we demonstrate that MNV provides a striking IL-22-dependent protection against early-life lethal infection by Citrobacter rodentium. These findings demonstrate novel ways in which a viral member of the microbiota fortifies the intestinal barrier during chemical injury and infectious challenges.


Subject(s)
Gastrointestinal Microbiome/immunology , Interferon Type I/metabolism , Interleukins/metabolism , Intestines/immunology , Intestines/virology , Animals , Anti-Bacterial Agents/toxicity , Cell Proliferation , Citrobacter rodentium/physiology , Colon/cytology , Colon/immunology , Colon/metabolism , Colon/virology , Dextran Sulfate/toxicity , Enterobacteriaceae Infections/prevention & control , Interleukins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestines/cytology , Intestines/drug effects , Lymphocytes/cytology , Lymphocytes/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Norovirus/immunology , Norovirus/physiology , Signal Transduction/genetics , Specific Pathogen-Free Organisms , Viral Nonstructural Proteins/genetics , Virus Replication , Interleukin-22
17.
Cell Host Microbe ; 25(6): 845-857.e5, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31130511

ABSTRACT

Murine norovirus (MNoV) infects a low percentage of enteric tuft cells and can persist in these cells for months following acute infection. Both tuft-cell tropism and resistance to interferon-λ (IFN-λ)-mediated clearance during persistent infection requires the viral nonstructural protein 1/2 (NS1/2). We show that processing of NS1/2 yields NS1, an unconventionally secreted viral protein that is central for IFN-λ resistance. MNoV infection globally suppresses intestinal IFN-λ responses, which is attributable to secreted NS1. MNoV NS1 secretion is triggered by caspase-3 cleavage of NS1/2, and a secreted form of human NoV NS1 is also observed. NS1 secretion is essential for intestinal infection and resistance to IFN-λ in vivo. NS1 vaccination alone protects against MNoV challenge, despite the lack of induction of neutralizing anti-capsid antibodies previously shown to confer protection. Thus, despite infecting a low number of tuft cells, NS1 secretion allows MNoV to globally suppress IFN responses and promote persistence.


Subject(s)
Caliciviridae Infections/pathology , Caliciviridae Infections/virology , Cytokines/antagonists & inhibitors , Immune Evasion , Norovirus/growth & development , Norovirus/pathogenicity , Viral Nonstructural Proteins/metabolism , Animals , Disease Models, Animal , Gastroenteritis/pathology , Gastroenteritis/virology , Humans , Mice , Virulence Factors/metabolism
18.
Cell Host Microbe ; 24(5): 665-676.e4, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30392829

ABSTRACT

Viral persistence can contribute to chronic disease and promote virus dissemination. Prior work demonstrated that timely clearance of systemic murine norovirus (MNV) infection depends on cell-intrinsic type I interferon responses and adaptive immunity. We now find that the capsid of the systemically replicating MNV strain CW3 promotes lytic cell death, release of interleukin-1α, and increased inflammatory cytokine release. Correspondingly, inflammatory monocytes and neutrophils are recruited to sites of infection in a CW3-capsid-dependent manner. Recruited monocytes and neutrophils are subsequently infected, representing a majority of infected cells in vivo. Systemic depletion of inflammatory monocytes or neutrophils from persistently infected Rag1-/- mice reduces viral titers in a tissue-specific manner. These data indicate that the CW3 capsid facilitates lytic cell death, inflammation, and recruitment of susceptible cells to promote persistence. Infection of continuously recruited inflammatory cells may be a mechanism of persistence broadly utilized by lytic viruses incapable of establishing latency.


Subject(s)
Caliciviridae Infections/immunology , Gastroenteritis/immunology , Myeloid Cells/immunology , Myeloid Cells/virology , Norovirus/immunology , Norovirus/pathogenicity , Adaptive Immunity , Animals , Caliciviridae Infections/virology , Capsid/immunology , Cell Death , Cytokines/metabolism , Disease Models, Animal , Female , Gastroenteritis/virology , Genes, Viral/genetics , HEK293 Cells , Homeodomain Proteins/genetics , Host-Pathogen Interactions , Humans , Inflammation/immunology , Interferon Type I/immunology , Interleukin-1alpha/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/virology , Neutrophils/immunology , Neutrophils/virology , Norovirus/genetics , Viral Load
19.
J Virol ; 92(23)2018 12 01.
Article in English | MEDLINE | ID: mdl-30209176

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC), composed of heme-oxidized IRP2 ubiquitin ligase 1 (HOIL1), HOIL1-interacting protein (HOIP), and SHANK-associated RH domain-interacting protein (SHARPIN), is a crucial regulator of multiple immune signaling pathways. In humans, HOIL1 or HOIP deficiency is associated with an immune disorder involving autoinflammation, immunodeficiency, and inflammatory bowel disease (IBD)-like symptoms. During viral infection, LUBAC is reported to inhibit the induction of interferon (IFN) by the cytosolic RNA sensor retinoic acid-inducible gene I (RIG-I). Surprisingly, we found that HOIL1 is essential for the induction of both type I and type III IFNs, as well as the phosphorylation of IFN regulatory factor 3 (IRF3), during murine norovirus (MNoV) infection in cultured dendritic cells. The RIG-I-like receptor, melanoma differentiation-associated protein 5 (MDA5), is also required for IFN induction and IRF3 phosphorylation during MNoV infection. Furthermore, HOIL1 and MDA5 were required for IFN induction after Theiler's murine encephalomyelitis virus infection and poly(I·C) transfection, but not Sendai virus or vesicular stomatitis virus infection, indicating that HOIL1 and LUBAC are required selectively for MDA5 signaling. Moreover, Hoil1-/- mice exhibited defective control of acute and persistent murine norovirus infection and defective regulation of MNoV persistence by the microbiome as also observed previously for mice deficient in interferon lambda (IFN-λ) receptor, signal transducer and activator of transcription factor 1 (STAT1), and IRF3. These data indicate that LUBAC plays a critical role in IFN induction to control RNA viruses sensed by MDA5.IMPORTANCE Human noroviruses are a leading cause of gastroenteritis throughout the world but are challenging to study in vivo and in vitro Murine norovirus (MNoV) provides a tractable genetic and small-animal model to study norovirus biology and immune responses. Interferons are critical mediators of antiviral immunity, but excessive expression can dysregulate the immune system. IFN-λ plays an important role at mucosal surfaces, including the gastrointestinal tract, and both IFN-λ and commensal enteric bacteria are important modulators of persistent MNoV infection. LUBAC, of which HOIL1 is a component, is reported to inhibit type I IFN induction after RIG-I stimulation. We show, in contrast, that HOIL1 is critical for type I and III IFN induction during infection with MNoV, a virus that preferentially activates MDA5. Moreover, HOIL1 regulates MNoV infection in vivo These data reveal distinct functions for LUBAC in these closely related signaling pathways and in modulation of IFN expression.


Subject(s)
Caliciviridae Infections/virology , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/metabolism , Norovirus/pathogenicity , Ubiquitin-Protein Ligases/physiology , Animals , Caliciviridae Infections/genetics , Caliciviridae Infections/metabolism , Caliciviridae Infections/microbiology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Dendritic Cells/virology , Fibroblasts/metabolism , Fibroblasts/microbiology , Fibroblasts/virology , Genome, Viral , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Type I/genetics , Interferon-Induced Helicase, IFIH1/genetics , Interferons/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Norovirus/genetics , Phosphorylation , Interferon Lambda
20.
Trends Microbiol ; 26(6): 510-524, 2018 06.
Article in English | MEDLINE | ID: mdl-29157967

ABSTRACT

Persistent viral infections result from evasion or avoidance of sterilizing immunity, extend the timeframe of virus transmission, and can trigger disease. Prior studies in mouse models of persistent infection have suggested that ineffective adaptive immune responses are necessary for persistent viral infection. However, recent work in the murine norovirus (MNV) model of persistent infection demonstrates that innate immunity can control both early and persistent viral replication independently of adaptive immune effector functions. Interferons (IFNs) are central to the innate control of persistent MNV, apart from a role in modulating adaptive immunity. Furthermore, subtypes of IFN play distinct tissue-specific roles in innate control of persistent MNV infection. Type I IFN (IFN-α/ß) controls systemic replication, and type III IFN (IFN-λ) controls MNV persistence in the intestinal epithelium. In this article, we review recent findings in the MNV model, highlighting the role of IFNs and innate immunity in clearing persistent viral infection, and discussing the broader implications of these findings for control of persistent human infections.


Subject(s)
Adaptive Immunity , Caliciviridae Infections/immunology , Immunity, Innate , Interferons/immunology , Animals , Disease Models, Animal , Host Microbial Interactions/immunology , Humans , Interferon Type I/immunology , Mice , Norovirus/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...