Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 35: 501-532, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28226227

ABSTRACT

Respiratory syncytial virus (RSV) is an exceptional mucosal pathogen. It specializes in infection of the ciliated respiratory epithelium, causing disease of variable severity with little or no direct systemic effects. It infects virtually all children by the age of three years and then repeatedly infects throughout life; this it does despite relatively slight variations in antigenicity, apparently by inducing selective immunological amnesia. Inappropriate or dysregulated responses to RSV can be pathogenic, causing disease-enhancing inflammation that contributes to short- and long-term effects. In addition, RSV's importance as a largely unrecognized pathogen of debilitated older people is increasingly evident. Vaccines that induce nonpathogenic protective immunity may soon be available, and it is possible that different vaccines will be optimal for infants; older children; young to middle-age adults (including pregnant women); and elderly persons. At the dawn of RSV vaccination, it is timely to review what is known (and unknown) about immune responses to this fascinating virus.


Subject(s)
Respiratory Mucosa/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Viral Vaccines/immunology , Adult , Aged , Animals , Child , Humans , Immune Evasion , Immunomodulation , Respiratory Mucosa/virology
2.
Nature ; 615(7952): 490-498, 2023 03.
Article in English | MEDLINE | ID: mdl-36890227

ABSTRACT

Metabolic rewiring underlies the effector functions of macrophages1-3, but the mechanisms involved remain incompletely defined. Here, using unbiased metabolomics and stable isotope-assisted tracing, we show that an inflammatory aspartate-argininosuccinate shunt is induced following lipopolysaccharide stimulation. The shunt, supported by increased argininosuccinate synthase (ASS1) expression, also leads to increased cytosolic fumarate levels and fumarate-mediated protein succination. Pharmacological inhibition and genetic ablation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) further increases intracellular fumarate levels. Mitochondrial respiration is also suppressed and mitochondrial membrane potential increased. RNA sequencing and proteomics analyses demonstrate that there are strong inflammatory effects resulting from FH inhibition. Notably, acute FH inhibition suppresses interleukin-10 expression, which leads to increased tumour necrosis factor secretion, an effect recapitulated by fumarate esters. Moreover, FH inhibition, but not fumarate esters, increases interferon-ß production through mechanisms that are driven by mitochondrial RNA (mtRNA) release and activation of the RNA sensors TLR7, RIG-I and MDA5. This effect is recapitulated endogenously when FH is suppressed following prolonged lipopolysaccharide stimulation. Furthermore, cells from patients with systemic lupus erythematosus also exhibit FH suppression, which indicates a potential pathogenic role for this process in human disease. We therefore identify a protective role for FH in maintaining appropriate macrophage cytokine and interferon responses.


Subject(s)
Fumarate Hydratase , Interferon-beta , Macrophages , Mitochondria , RNA, Mitochondrial , Humans , Argininosuccinate Synthase/metabolism , Argininosuccinic Acid/metabolism , Aspartic Acid/metabolism , Cell Respiration , Cytosol/metabolism , Fumarate Hydratase/antagonists & inhibitors , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Fumarates/metabolism , Interferon-beta/biosynthesis , Interferon-beta/immunology , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Lupus Erythematosus, Systemic/enzymology , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Membrane Potential, Mitochondrial , Metabolomics , Mitochondria/genetics , Mitochondria/metabolism , RNA, Mitochondrial/metabolism
3.
PLoS Pathog ; 18(2): e1010272, 2022 02.
Article in English | MEDLINE | ID: mdl-35108347

ABSTRACT

Respiratory syncytial virus (RSV) can cause bronchiolitis and viral pneumonia in young children and the elderly. Lack of vaccines and recurrence of RSV infection indicate the difficulty in eliciting protective memory immune responses. Tissue resident memory T cells (TRM) can confer protection from pathogen re-infection and, in human experimental RSV infection, the presence of lung CD8+ TRM cells correlates with a better outcome. However, the requirements for generating and maintaining lung TRM cells during RSV infection are not fully understood. Here, we use mouse models to assess the impact of innate immune response determinants in the generation and subsequent expansion of the TRM cell pool during RSV infection. We show that CD8+ TRM cells expand independently from systemic CD8+ T cells after RSV re-infection. Re-infected MAVS and MyD88/TRIF deficient mice, lacking key components involved in innate immune recognition of RSV and induction of type I interferons (IFN-α/ß), display impaired expansion of CD8+ TRM cells and reduction in antigen specific production of granzyme B and IFN-γ. IFN-α treatment of MAVS deficient mice during primary RSV infection restored TRM cell expansion upon re-challenge but failed to recover TRM cell functionality. Our data reveal how innate immunity, including the axis controlling type I IFN induction, instructs and regulates CD8+ TRM cell responses to RSV infection, suggesting possible mechanisms for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD8-Positive T-Lymphocytes/immunology , Interferon Type I/immunology , Memory T Cells/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Granzymes/immunology , Granzymes/metabolism , Immunity, Innate , Immunologic Memory , Interferon Type I/metabolism , Lung/immunology , Mice , Mice, Inbred C57BL , Respiratory Syncytial Virus Infections/virology , Signal Transduction
4.
Eur J Immunol ; 52(11): 1768-1775, 2022 11.
Article in English | MEDLINE | ID: mdl-36106692

ABSTRACT

SARS-CoV-2 is a newly emerged coronavirus, causing the global pandemic of respiratory coronavirus disease (COVID-19). The type I interferon (IFN) pathway is of particular importance for anti-viral defense and recent studies identified that type I IFNs drive early inflammatory responses to SARS-CoV-2. Here, we use a mouse model of SARS-CoV-2 infection, facilitating viral entry by intranasal recombinant Adeno-Associated Virus (rAAV) transduction of hACE2 in wildtype (WT) and type I IFN receptor-1 deficient (Ifnar1-/- ) mice, to study the role of type I IFN signalling and innate immune responses during SARS-CoV-2 infection. Our data show that type I IFN signalling is essential for inducing anti-viral effector responses to SARS-CoV-2, control of virus replication, and to prevent enhanced disease. Furthermore, hACE2-Ifnar1-/- mice had increased gene expression of the chemokine Cxcl1 and airway infiltration of neutrophils as well as reduced and delayed production of monocyte-recruiting chemokine CCL2. hACE2-Ifnar1-/- mice showed altered recruitment of inflammatory myeloid cells to the lung upon SARS-CoV-2 infection, with a shift from Ly6C+ to Ly6C- expressing cells. Together, our findings suggest that type I IFN signalling deficiency results in a dysregulated innate immune response to SARS-CoV-2 infection.


Subject(s)
COVID-19 , Immunity, Innate , Receptor, Interferon alpha-beta , Animals , Mice , COVID-19/immunology , Interferon Type I , Pandemics , Receptor, Interferon alpha-beta/genetics , SARS-CoV-2
5.
J Intern Med ; 294(5): 605-615, 2023 11.
Article in English | MEDLINE | ID: mdl-37387643

ABSTRACT

BACKGROUND: Previous studies have shown an increased risk for atrial fibrillation and atrial flutter (AF) in people with type 2 diabetes and prediabetes. It is unclear whether this increase in AF risk is independent of other risk factors for AF. OBJECTIVE: To investigate the association between diabetes and different prediabetic states, as independent risk factors for the onset of AF. METHODS: We performed a population-based cohort study in Northern Sweden, including data on fasting plasma glucose, oral glucose tolerance test, major cardiovascular risk factors, medical history, and lifestyle factors. Participants were divided into six groups depending on glycemic status and followed through national registers for AF diagnosis. Cox proportional hazard model was used to assess the association between glycemic status and AF, using normoglycemia as reference. RESULTS: The cohort consisted of 88,889 participants who underwent a total of 139,661 health examinations. In the model adjusted for age and sex, there was a significant association between glycemic status and development of AF in all groups except the impaired glucose tolerance group, with the strongest association for the group with known diabetes (p-value <0.001). In a model adjusted for sex, age, systolic blood pressure, body mass index, antihypertensive drugs, cholesterol, alcohol, smoking, education level, marital status, and physical activity, there was no significant association between glycemic status and AF. CONCLUSIONS/INTERPRETATION: The association between glycemic status and AF disappears upon adjustment for potential confounders. Diabetes and prediabetes do not appear to be independent risk factors for AF.


Subject(s)
Atrial Fibrillation , Diabetes Mellitus, Type 2 , Prediabetic State , Humans , Prediabetic State/complications , Prediabetic State/epidemiology , Atrial Fibrillation/epidemiology , Atrial Fibrillation/etiology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/epidemiology , Cohort Studies , Blood Glucose , Risk Factors
6.
J Virol ; 96(21): e0117822, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36226985

ABSTRACT

Defective viral genomes (DVGs), which are generated by the viral polymerase in error during RNA replication, can trigger innate immunity and are implicated in altering the clinical outcome of infection. Here, we investigated the impact of DVGs on innate immunity and pathogenicity in a BALB/c mouse model of influenza virus infection. We generated stocks of influenza viruses containing the internal genes of an H5N1 virus that contained different levels of DVGs (indicated by different genome-to-PFU ratios). In lung epithelial cells, the high-DVG stock was immunostimulatory at early time points postinfection. DVGs were amplified during virus replication in myeloid immune cells and triggered proinflammatory cytokine production. In the mouse model, infection with the different virus stocks produced divergent outcomes. The high-DVG stock induced an early type I interferon (IFN) response that limited viral replication in the lungs, resulting in minimal weight loss. In contrast, the virus stock with low levels of DVGs replicated to high titers and amplified DVGs over time, resulting in elevated levels of proinflammatory cytokines accompanied by rapid weight loss and increased morbidity and mortality. Our results suggest that the timing and levels of immunostimulatory DVGs generated during infection contribute to H5N1 pathogenesis. IMPORTANCE Mammalian infections with highly pathogenic avian influenza viruses (HPAIVs) cause severe disease associated with excessive proinflammatory cytokine production. Aberrant replication products, such as defective viral genomes (DVGs), can stimulate the antiviral response, and cytokine induction is associated with their emergence in vivo. We show that stocks of a recombinant virus containing HPAIV internal genes that differ in their amounts of DVGs have vastly diverse outcomes in a mouse model. The high-DVG stock resulted in extremely mild disease due to suppression of viral replication. Conversely, the stock that contained low DVGs but rapidly accumulated DVGs over the course of infection led to severe disease. Therefore, the timing of DVG amplification and proinflammatory cytokine production impact disease outcome, and these findings demonstrate that not all DVG generation reduces viral virulence. This study also emphasizes the crucial requirement to examine the quality of virus preparations regarding DVG content to ensure reproducible research.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Mice , Animals , Defective Viruses/genetics , Influenza A virus/genetics , Mice, Inbred BALB C , Influenza A Virus, H5N1 Subtype/genetics , Genome, Viral , Virus Replication/genetics , Cytokines/genetics , Weight Loss/genetics , Mammals/genetics
7.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28716804

ABSTRACT

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN-I) in enabling this process. An IFN-I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN-I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1-/- mice were incapable of initiating Th2 responses in vivo These data demonstrate for the first time that the influence of IFN-I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/metabolism , Th2 Cells/immunology , Allergens/immunology , Animals , Mice , Mice, Knockout , Pyroglyphidae/immunology , Receptor, Interferon alpha-beta/deficiency , Schistosoma mansoni/immunology
8.
PLoS Pathog ; 14(1): e1006821, 2018 01.
Article in English | MEDLINE | ID: mdl-29300777

ABSTRACT

The highly pathogenic avian influenza (HPAI) H5N1 influenza virus has been a public health concern for more than a decade because of its frequent zoonoses and the high case fatality rate associated with human infections. Severe disease following H5N1 influenza infection is often associated with dysregulated host innate immune response also known as cytokine storm but the virological and cellular basis of these responses has not been clearly described. We rescued a series of 6:2 reassortant viruses that combined a PR8 HA/NA pairing with the internal gene segments from human adapted H1N1, H3N2, or avian H5N1 viruses and found that mice infected with the virus with H5N1 internal genes suffered severe weight loss associated with increased lung cytokines but not high viral load. This phenotype did not map to the NS gene segment, and NS1 protein of H5N1 virus functioned as a type I IFN antagonist as efficient as NS1 of H1N1 or H3N2 viruses. Instead we discovered that the internal genes of H5N1 virus supported a much higher level of replication of viral RNAs in myeloid cells in vitro, but not in epithelial cells and that this was associated with high induction of type I IFN in myeloid cells. We also found that in vivo during H5N1 recombinant virus infection cells of haematopoetic origin were infected and produced type I IFN and proinflammatory cytokines. Taken together our data infer that human and avian influenza viruses are differently controlled by host factors in alternative cell types; internal gene segments of avian H5N1 virus uniquely drove high viral replication in myeloid cells, which triggered an excessive cytokine production, resulting in severe immunopathology.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/physiology , Myeloid Cells/virology , Orthomyxoviridae Infections/genetics , Virus Replication/genetics , A549 Cells , Animals , Cells, Cultured , Dogs , Female , Genes, Viral/physiology , HEK293 Cells , Humans , Immunity, Innate/physiology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Severity of Illness Index
9.
Appl Environ Microbiol ; 85(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30578266

ABSTRACT

Campylobacter bacteria are major human enteropathogens. Campylobacter coli shows less genetic diversity than C. jejuni and clusters into three clades, of which clade 1 includes most human and farm animal isolates, while environmental C. coli isolates mainly belong to clades 2 and 3. Recently, we sequenced the whole genomes of eight C. coli clade 2 and 3 isolates cultivated from water, and here we studied their interaction with human HT-29 colon cancer cells compared to that of clinical clade 1 isolates. All C. coli clade 3 isolates already caused cell necrosis 1 to 2 h after inoculation, whereas none of the clade 1 and 2 isolates analyzed induced cell death. Isolates from clades 2 and 3 adhered to epithelial cells better than clade 1 isolates, but all isolates induced similar levels of interleukin-8 (IL-8). Alignment and phylogenetic analysis of the translated putative virulence genes cadF, flpA, iamA, ciaB, and ceuE revealed clade-specific protein sequence variations, with clade 1 and 2 sequences being more closely related and clade 3 sequences being further apart, in general. Moreover, when RNA levels were measured, clade 3 isolates showed significantly lower levels of expression of cadF, iamA, and ceuE than clade 2 isolates, while flpA expression levels were higher in clade 3 isolates. The cytolethal distending toxin genes were also expressed in clades 2 and 3, although there was no difference between clades. Our findings demonstrate differences between the effects of C. coli clade 1, 2, and 3 isolates on human cells and suggest that C. coli clade 3 might be more virulent than clade 2 due to the observed cytotoxicity.IMPORTANCECampylobacter coli is a common zoonotic cause of gastroenteritis in humans worldwide. The majority of infections are caused by C. coli clade 1 isolates, whereas infections due to clade 2 and 3 isolates are rare. Whether this depends on a low prevalence of clade 2 and 3 isolates in reservoirs important for human infections or their lower ability to cause human disease is unknown. Here, we studied the effects of C. coli clade 2 and 3 isolates on a human cell line. These isolates adhered to human cells to a higher degree than clinical clade 1 isolates. Furthermore, we could show that C. coli clade 3 isolates rapidly induced cell death, suggesting differences in the virulence of C. coli The exact mechanism of cell death remains to be revealed, but selected genes showed interesting clade-specific expression patterns.


Subject(s)
Campylobacter coli/isolation & purification , Campylobacter coli/metabolism , Cell Death , Phylogeny , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Base Sequence , Campylobacter Infections/microbiology , Campylobacter coli/genetics , Campylobacter coli/pathogenicity , Gastroenteritis/microbiology , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Genome, Bacterial , HT29 Cells , Humans , Interleukin-8/metabolism , Necrosis , Sequence Analysis , Virulence/genetics , Whole Genome Sequencing
11.
J Biol Chem ; 290(21): 13354-71, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25878250

ABSTRACT

In order to identify cellular factors that regulate human papillomavirus type 16 (HPV16) gene expression, cervical cancer cells permissive for HPV16 late gene expression were identified and characterized. These cells either contained a novel spliced variant of the L1 mRNAs that bypassed the suppressed HPV16 late, 5'-splice site SD3632; produced elevated levels of RNA-binding proteins SRSF1 (ASF/SF2), SRSF9 (SRp30c), and HuR that are known to regulate HPV16 late gene expression; or were shown by a gene expression array analysis to overexpress the RALYL RNA-binding protein of the heterogeneous nuclear ribonucleoprotein C (hnRNP C) family. Overexpression of RALYL or hnRNP C1 induced HPV16 late gene expression from HPV16 subgenomic plasmids and from episomal forms of the full-length HPV16 genome. This induction was dependent on the HPV16 early untranslated region. Binding of hnRNP C1 to the HPV16 early, untranslated region activated HPV16 late 5'-splice site SD3632 and resulted in production of HPV16 L1 mRNAs. Our results suggested that hnRNP C1 controls HPV16 late gene expression.


Subject(s)
3' Untranslated Regions/genetics , Capsid Proteins/metabolism , Gene Expression Regulation, Viral , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Oncogene Proteins, Viral/metabolism , RNA Splicing/genetics , RNA, Messenger/genetics , Uterine Cervical Neoplasms/metabolism , Blotting, Western , Capsid Proteins/genetics , Epidermal Cells , Epidermis/metabolism , Epidermis/virology , Female , Fluorescent Antibody Technique , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Human papillomavirus 16/physiology , Humans , Immunoprecipitation , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/virology , Microarray Analysis , Oncogene Proteins, Viral/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
12.
EMBO J ; 31(14): 3212-27, 2012 May 22.
Article in English | MEDLINE | ID: mdl-22617423

ABSTRACT

We provide evidence that the human papillomavirus (HPV) E2 protein regulates HPV late gene expression. High levels of E2 caused a read-through at the early polyadenylation signal pAE into the late region of the HPV genome, thereby inducing expression of L1 and L2 mRNAs. This is a conserved property of E2 of both mucosal and cutaneous HPV types. Induction could be reversed by high levels of HPV-16 E1 protein, or by the polyadenylation factor CPSF30. HPV-16 E2 inhibited polyadenylation in vitro by preventing the assembly of the CPSF complex. Both the N-terminal and hinge domains of E2 were required for induction of HPV late gene expression in transfected cells as well as for inhibition of polyadenylation in vitro. Finally, overexpression of HPV-16 E2 induced late gene expression from a full-length genomic clone of HPV-16. We speculate that the accumulation of high levels of E2 during the viral life cycle, not only turns off the expression of the pro-mitotic viral E6 and E7 genes, but also induces the expression of the late HPV genes L1 and L2.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral/physiology , Human papillomavirus 16/physiology , Oncogene Proteins, Viral/metabolism , Polyadenylation/physiology , RNA 3' Polyadenylation Signals/physiology , RNA, Viral/metabolism , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/genetics , RNA, Viral/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 110(8): 2987-92, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382205

ABSTRACT

Human trials of formaldehyde-inactivated respiratory syncytial virus (FI-RSV) vaccine in 1966-1967 caused disastrous worsening of disease and death in infants during subsequent natural respiratory syncytial virus (RSV) infection. The reasons behind vaccine-induced augmentation are only partially understood, and fear of augmentation continues to hold back vaccine development. We now show that mice vaccinated with FI-RSV show enhanced local recruitment of conventional CD4(+) T cells accompanied by a profound loss of regulatory T cells (Tregs) in the airways. This loss of Tregs was so complete that additional depletion of Tregs (in transgenic depletion of regulatory T-cell mice) produced no additional disease enhancement. Transfer of conventional CD4(+) T cells from FI-RSV-vaccinated mice into naive RSV-infected recipients also caused a reduction in airway Treg responses; boosting Tregs with IL-2 immune complexes failed to restore normal levels of Tregs or to ameliorate disease. However, delivery of chemokine ligands (CCL) 17/22 via the airway selectively recruited airway Tregs and attenuated vaccine-augmented disease, reducing weight loss and inhibiting local recruitment of pathogenic CD4(+) T cells. These findings reveal an unexpected mechanism of vaccine-induced disease augmentation and indicate that selective chemoattraction of Tregs into diseased sites may offer a novel approach to the modulation of tissue-specific inflammation.


Subject(s)
Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/immunology , Viral Vaccines/immunology , Adoptive Transfer , Animals , Bronchoalveolar Lavage Fluid , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Respiratory Syncytial Virus Infections/immunology
14.
Proc Natl Acad Sci U S A ; 110(14): 5576-81, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23509276

ABSTRACT

Respiratory syncytial virus (RSV) infects most children in the first year of life and is a major single cause of hospitalization in infants and young children. There is no effective vaccine, and antibody generated by primary neonatal infection is poorly protective against reinfection even with antigenically homologous viral strains. Studying the immunological basis of these observations in neonatal mice, we found that antibody responses to infection were low and unaffected by CD4 depletion, in contrast with adult mice, which had stronger CD4-dependent antibody responses. Natural killer cell depletion or codepletion of CD4(+) and CD8(+) cells during neonatal RSV infection caused a striking increase in anti-RSV antibody titer. These cells are major sources of the cytokine IFN-γ, and blocking IFN-γ also enhanced RSV-specific antibody responses in neonates. In addition, infection with a recombinant RSV engineered to produce IFN-γ reduced antibody titer, confirming that IFN-γ plays a pivotal role in inhibition of antibody responses after neonatal infection. These unexpected findings show that the induction of a strong cellular immune response may limit antibody responses in early life and that vaccines that induce IFN-γ-secreting cells might, in some situations, be less protective than those that do not.


Subject(s)
Animals, Newborn , Antibodies, Viral/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Respiratory Syncytial Virus Infections/immunology , T-Lymphocytes/immunology , Analysis of Variance , Animals , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy
15.
Eur J Immunol ; 44(8): 2340-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777856

ABSTRACT

During respiratory syncytial virus (RSV) infection CD8(+) T cells both assist in viral clearance and contribute to immunopathology. CD8(+) T cells recognize viral peptides presented by dendritic cells (DCs), which can directly present viral antigens when infected or, alternatively, "cross-present" antigens after endocytosis of dead or dying infected cells. Mouse CD8α(+) and CD103(+) DCs excel at cross-presentation, in part because they express the receptor DNGR-1 that detects dead cells by binding to exposed F-actin and routes internalized cell debris into the cross-presentation pathway. As RSV causes death in infected epithelial cells, we tested whether cross-presentation via DNGR-1 is necessary for CD8(+) T-cell responses to the virus. DNGR-1-deficient or wild-type mice were intranasally inoculated with RSV and the magnitude of RSV-specific CD8(+) T-cell induction was measured. We found that during live RSV infection, cross-presentation via DNGR-1 did not have a major role in the generation of RSV-specific CD8(+) T-cell responses. However, after intranasal immunization with dead cells infected with RSV, a dependence on DNGR-1 for RSV-specific CD8(+) T-cell responses was observed, confirming the ascribed role of the receptor. Thus, direct presentation by DCs may be the major pathway initiating CD8(+) T-cell responses to RSV, while DNGR-1-dependent cross-presentation has no detectable role.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lectins, C-Type/immunology , Receptors, Immunologic/immunology , Respiratory Syncytial Virus Infections/immunology , Actins/immunology , Animals , Antigen Presentation/immunology , Antigens, Viral/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Epithelial Cells/immunology , Epithelial Cells/virology , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Respiratory Syncytial Viruses/immunology , Viral Load/immunology
16.
J Virol ; 88(11): 6128-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24648449

ABSTRACT

UNLABELLED: Type I interferons (IFNs) are produced early upon virus infection and signal through the alpha/beta interferon (IFN-α/ß) receptor (IFNAR) to induce genes that encode proteins important for limiting viral replication and directing immune responses. To investigate the extent to which type I IFNs play a role in the local regulation of inflammation in the airways, we examined their importance in early lung responses to infection with respiratory syncytial virus (RSV). IFNAR1-deficient (IFNAR1(-/-)) mice displayed increased lung viral load and weight loss during RSV infection. As expected, expression of IFN-inducible genes was markedly reduced in the lungs of IFNAR1(-/-) mice. Surprisingly, we found that the levels of proinflammatory cytokines and chemokines in the lungs of RSV-infected mice were also greatly reduced in the absence of IFNAR signaling. Furthermore, low levels of proinflammatory cytokines were also detected in the lungs of IFNAR1(-/-) mice challenged with noninfectious innate immune stimuli such as selected Toll-like receptor (TLR) agonists. Finally, recombinant IFN-α was sufficient to potentiate the production of inflammatory mediators in the lungs of wild-type mice challenged with innate immune stimuli. Thus, in addition to its well-known role in antiviral resistance, type I IFN receptor signaling acts as a central driver of early proinflammatory responses in the lung. Inhibiting the effects of type I IFNs may therefore be useful in dampening inflammation in lung diseases characterized by enhanced inflammatory cytokine production. IMPORTANCE: The initial response to viral infection is characterized by the production of interferons (IFNs). One group of IFNs, the type I IFNs, are produced early upon virus infection and signal through the IFN-α/ß receptor (IFNAR) to induce proteins important for limiting viral replication and directing immune responses. Here we examined the importance of type I IFNs in early responses to respiratory syncytial virus (RSV). Our data suggest that type I IFN production and IFNAR receptor signaling not only induce an antiviral state but also serve to amplify proinflammatory responses in the respiratory tract. We also confirm this conclusion in another model of acute inflammation induced by noninfectious stimuli. Our findings are of relevance to human disease, as RSV is a major cause of infant bronchiolitis and polymorphisms in the IFN system are known to impact disease severity.


Subject(s)
Cytokines/biosynthesis , Gene Expression Regulation/immunology , Lung/metabolism , Respiratory Syncytial Virus Infections/immunology , Signal Transduction/physiology , Animals , DNA Primers/genetics , Lung/virology , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Statistics, Nonparametric , Viral Load
17.
Nucleic Acids Res ; 41(22): 10488-508, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24013563

ABSTRACT

Human papillomavirus type 16 (HPV-16) 5'-splice site SD3632 is used exclusively to produce late L1 mRNAs. We identified a 34-nt splicing inhibitory element located immediately upstream of HPV-16 late 5'-splice site SD3632. Two AUAGUA motifs located in these 34 nt inhibited SD3632. Two nucleotide substitutions in each of the HPV-16 specific AUAGUA motifs alleviated splicing inhibition and induced late L1 mRNA production from episomal forms of the HPV-16 genome in primary human keratinocytes. The AUAGUA motifs bind specifically not only to the heterogeneous nuclear RNP (hnRNP) D family of RNA-binding proteins including hnRNP D/AUF, hnRNP DL and hnRNP AB but also to hnRNP A2/B1. Knock-down of these proteins induced HPV-16 late L1 mRNA expression, and overexpression of hnRNP A2/B1, hnRNP AB, hnRNP DL and the two hnRNP D isoforms hnRNP D37 and hnRNP D40 further suppressed L1 mRNA expression. This inhibition may allow HPV-16 to hide from the immune system and establish long-term persistent infections with enhanced risk at progressing to cancer. There is an inverse correlation between expression of hnRNP D proteins and hnRNP A2/B1 and HPV-16 L1 production in the cervical epithelium, as well as in cervical cancer, supporting the conclusion that hnRNP D proteins and A2/B1 inhibit HPV-16 L1 mRNA production.


Subject(s)
Capsid Proteins/genetics , Gene Expression Regulation, Viral , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Human papillomavirus 16/genetics , Oncogene Proteins, Viral/genetics , RNA Splice Sites , RNA, Viral/chemistry , Binding Sites , Capsid Proteins/biosynthesis , Cell Line , HeLa Cells , Humans , Keratinocytes/virology , Nucleotide Motifs , Oncogene Proteins, Viral/biosynthesis , RNA Splicing , RNA, Messenger/biosynthesis , Regulatory Sequences, Ribonucleic Acid , Sequence Deletion
19.
J Virol ; 87(20): 10946-54, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23926350

ABSTRACT

During viral infection, inflammation and recovery are tightly controlled by competing proinflammatory and regulatory immune pathways. Respiratory syncytial virus (RSV) is the leading global cause of infantile bronchiolitis, which is associated with recurrent wheeze and asthma diagnosis in later life. Th2-driven disease has been well described under some conditions for RSV-infected mice. In the present studies, we used the Foxp3(DTR) mice (which allow specific conditional depletion of Foxp3(+) T cells) to investigate the functional effects of regulatory T cells (Tregs) during A2-strain RSV infection. Infected Treg-depleted mice lost significantly more weight than wild-type mice, indicating enhanced disease. This enhancement was characterized by increased cellularity in the bronchoalveolar lavage (BAL) fluid and notable lung eosinophilia not seen in control mice. This was accompanied by abundant CD4(+) and CD8(+) T cells exhibiting an activated phenotype and induction of interleukin 13 (IL-13)- and GATA3-expressing Th2-type CD4(+) T cells that remained present in the airways even 14 days after infection. Therefore, Treg cells perform vital anti-inflammatory functions during RSV infection, suppressing pathogenic T cell responses and inhibiting lung eosinophilia. These findings provide additional evidence that dysregulation of normal immune responses to viral infection may contribute to severe RSV disease.


Subject(s)
Pulmonary Eosinophilia/pathology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/pathology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Forkhead Transcription Factors/biosynthesis , Gene Knockdown Techniques , Interleukin-13/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Scand J Prim Health Care ; 32(4): 226-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25464863

ABSTRACT

OBJECTIVE: An underuse of oral anticoagulants (OAC) in patients with atrial fibrillation (AF) has been suggested, as only 50% of all patients with AF receive OAC treatment. Whether this is due to contraindications, lack of an indication to treat, or an expression of underuse is sparsely investigated. This study therefore aimed to characterize individuals without OAC treatment in a real-life population of patients with AF. DESIGN: Retrospective cross-sectional study. The medical records were scrutinized in order to identify the type of AF, risk factors for embolism and bleeding, and other factors of importance for OAC treatment. SETTING: The municipalities of Skellefteå and Norsjö, northern Sweden. SUBJECTS: A total of 2274 living residents with at least one verified episode of AF on or before December 31, 2010. MAIN OUTCOME MEASURES: Prevalence of treatment with OAC and documented reasons to withhold OAC treatment. RESULTS: Among all 2274 patients with AF, 1187 (52%) were not treated with OAC. Of the untreated patients, 19% had no indication or had declined or had experienced adverse effects other than bleeding on warfarin treatment. The most common reason to withhold OAC was presence of risk factors for bleeding, found in 38% of all untreated patients. Furthermore, a documented reason could be identified to withhold OAC in 75%. CONCLUSIONS: Among patients with AF without OAC treatment a reason could be identified to withhold OAC in 75%. The underuse of OAC is estimated to be 25%.


Subject(s)
Anticoagulants/administration & dosage , Atrial Fibrillation/drug therapy , Atrial Fibrillation/epidemiology , Cardiovascular Diseases/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Anticoagulants/adverse effects , Anticoagulants/therapeutic use , Child , Child, Preschool , Cross-Sectional Studies , Female , Guideline Adherence , Humans , Infant , Infant, Newborn , Male , Middle Aged , Retrospective Studies , Risk Factors , Sweden/epidemiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL