Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 731
Filter
1.
Cell Immunol ; 399-400: 104823, 2024.
Article in English | MEDLINE | ID: mdl-38520831

ABSTRACT

AAV-mediated gene transfer is a promising platform still plagued by potential host-derived, antagonistic immune responses to therapeutic components. CpG-mediated TLR9 stimulation activates innate immune cells and leads to cognate T cell activation and suppression of transgene expression. Here, we demonstrate that CpG depletion increased expression of an antibody transgene product by 2-3-fold as early as 24 h post-vector administration in mice. No significant differences were noted in anti-transgene product/ anti-AAV capsid antibody production or cytotoxic gene induction. Instead, CpG depletion significantly reduced the presence of a pDC-like myeloid cell population, which was able to directly bind the antibody transgene product via Fc-FcγR interactions. Thus, we extend the mechanisms of TLR9-mediated antagonism of transgene expression in AAV gene therapy to include the actions of a previously unreported pDC-like cell population.


Subject(s)
Dendritic Cells , Dependovirus , Genetic Therapy , Genetic Vectors , Mice, Inbred C57BL , Toll-Like Receptor 9 , Transgenes , Animals , Dendritic Cells/immunology , Dependovirus/genetics , Mice , Genetic Therapy/methods , Toll-Like Receptor 9/immunology , CpG Islands/genetics , CpG Islands/immunology , Receptors, IgG/immunology , Receptors, IgG/genetics , Receptors, IgG/metabolism
2.
Front Immunol ; 12: 758190, 2021.
Article in English | MEDLINE | ID: mdl-34867997

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are the key producers of type I interferons (IFNs), thus playing a central role in initiating antiviral immune response. Besides robust type I IFN production, pDCs also act as antigen presenting cells post immunogenic stimulation. Transcription factor Irf8 is indispensable for the development of both pDC and cDC1 subset. However, the mechanism underlying the differential regulation by IRF8 in cDC1- and pDC-specific genomic architecture of developmental pathways still remains to be fully elucidated. Previous studies indicated that the Irf8R294C mutation specifically abrogates development of cDC1 without affecting that of pDC. In the present study using RNA-seq based approach, we have found that though the point mutation Irf8R294C did not affect pDC development, it led to defective type I IFN production, thus resulting in inefficient antiviral response. This observation unraveled the distinctive roles of IRF8 in these two subpopulations-regulating the development of cDC1 whereas modulating the functionality of pDCs without affecting development. We have reported here that Irf8R294C mutation also caused defect in production of ISGs as well as defective upregulation of costimulatory molecules in pDCs in response to NDV infection (or CpG stimulation). Through in vivo studies, we demonstrated that abrogation of type I IFN production was concomitant with reduced upregulation of costimulatory molecules in pDCs and increased NDV burden in IRF8R294C mice in comparison with wild type, indicating inefficient viral clearance. Further, we have also shown that Irf8R294C mutation abolished the activation of type I IFN promoter by IRF8, justifying the low level of type I IFN production. Taken together, our study signifies that the single point mutation in Irf8, Irf8R294C severely compromised type I IFN-mediated immune response by murine pDCs, thereby causing impairment in antiviral immunity.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/genetics , Interferon Type I/immunology , Mutation, Missense , Newcastle Disease/immunology , Point Mutation , Animals , Bone Neoplasms/pathology , Cell Line, Tumor , CpG Islands/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factors/immunology , Interferon Type I/biosynthesis , Male , Mice , Mice, Inbred C57BL , Newcastle disease virus , Osteosarcoma/pathology , Transcriptome
3.
Sci Rep ; 11(1): 10696, 2021 05 21.
Article in English | MEDLINE | ID: mdl-34021237

ABSTRACT

A new pandemic caused by the betacoronavirus SARS-CoV-2 originated in China in late 2019. Although often asymptomatic, a relevant percentage of affected people can develop severe pneumonia. Initial evidence suggests that dysregulation of the immune response could contribute to the pathogenesis, as previously demonstrated for SARS-CoV. The presence of genome composition features involved in delaying viral recognition is herein investigated for human coronaviruses (HCoVs), with a special emphasis on SARS-CoV-2. A broad collection of HCoVs polyprotein, envelope, matrix, nucleocapsid and spike coding sequences was downloaded and several statistics representative of genome composition and codon bias were investigated. A model able to evaluate and test the presence of a significant under- or over-representation of dinucleotide pairs while accounting for the underlying codon bias and protein sequence was also implemented. The study revealed the significant under-representation of CpG dinucleotide pair in all HcoV, but especially in SARS-CoV and even more in SARS-CoV-2. The presence of forces acting to minimize CpG content was confirmed by relative synonymous codon usage pattern. Codons containing the CpG pair were severely under-represented, primarily in the polyprotein and spike coding sequences of SARS-CoV-2. Additionally, a significant under-representation of the TpA pair was observed in the N and S region of SARS-CoV and SARS-CoV-2. Increasing experimental evidence has proven that CpG and TpA are targeted by innate antiviral host defences, contributing both to RNA degradation and RIG-1 mediated interferon production. The low content of these dinucleotides could contribute to a delayed interferon production, dysregulated immune response, higher viral replication and poor outcome. Significantly, the RIG-1 signalling pathway was proven to be defective in elderlies, suggesting a likely interaction between limited viral recognition and lower responsiveness in interferon production that could justify the higher disease severity and mortality in older patients.


Subject(s)
COVID-19 , CpG Islands/immunology , Genome, Viral/immunology , RNA, Viral , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , COVID-19/genetics , COVID-19/immunology , Humans , RNA Stability/immunology , RNA, Viral/genetics , RNA, Viral/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology
4.
Sci Immunol ; 6(58)2021 04 02.
Article in English | MEDLINE | ID: mdl-33811059

ABSTRACT

Plasmacytoid dendritic cells (pDCs) can rapidly produce interferons and other soluble factors in response to extracellular viruses or virus mimics such as CpG-containing DNA. pDCs can also recognize live cells infected with certain RNA viruses, but the relevance and functional consequences of such recognition remain unclear. We studied the response of primary DCs to the prototypical persistent DNA virus, human cytomegalovirus (CMV). Human pDCs produced high amounts of type I interferon (IFN-I) when incubated with live CMV-infected fibroblasts but not with free CMV; the response involved integrin-mediated adhesion, transfer of DNA-containing virions to pDCs, and the recognition of DNA through TLR9. Compared with transient polyfunctional responses to CpG or free influenza virus, pDC response to CMV-infected cells was long-lasting, dominated by the production of IFN-I and IFN-III, and lacked diversification into functionally distinct populations. Similarly, pDC activation by influenza-infected lung epithelial cells was highly efficient, prolonged, and dominated by interferon production. Prolonged pDC activation by CMV-infected cells facilitated the activation of natural killer cells critical for CMV control. Last, patients with CMV viremia harbored phenotypically activated pDCs and increased circulating IFN-I and IFN-III. Thus, recognition of live infected cells is a mechanism of virus detection by pDCs that elicits a unique antiviral immune response.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/immunology , Interferon Type I/metabolism , Interferons/metabolism , Cells, Cultured , Coculture Techniques , CpG Islands/immunology , Cytomegalovirus/immunology , Dendritic Cells/metabolism , Fibroblasts/virology , Host Microbial Interactions/immunology , Humans , Primary Cell Culture , Interferon Lambda
5.
Clin Epigenetics ; 13(1): 29, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33541404

ABSTRACT

BACKGROUND: Pneumococcal infections are a major cause of morbidity and mortality in young children and immaturity of the immune system partly underlies poor vaccine responses seen in the young. Emerging evidence suggests a key role for epigenetics in the maturation and regulation of the immune system in health and disease. The study aimed to investigate epigenetic changes in early life and to understand the relationship between the epigenome and antigen-specific antibody responses to pneumococcal vaccination. METHODS: The epigenetic profiles from 24 healthy children were analyzed at 12 months prior to a booster dose of the 13-valent pneumococcal conjugate vaccine (PCV-13), and at 24 months of age, using the Illumina Methylation 450 K assay and assessed for differences over time and between high and low vaccine responders. RESULTS: Our analysis revealed 721 significantly differentially methylated positions between 12 and 24 months (FDR < 0.01), with significant enrichment in pathways involved in the regulation of cell-cell adhesion and T cell activation. Comparing high and low vaccine responders, we identified differentially methylated CpG sites (P value < 0.01) associated with HLA-DPB1 and IL6. CONCLUSION: These data imply that epigenetic changes that occur during early childhood may be associated with antigen-specific antibody responses to pneumococcal vaccines.


Subject(s)
Immune System/metabolism , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/genetics , Antigen-Antibody Reactions/immunology , Case-Control Studies , Cell Competition/immunology , Child, Preschool , CpG Islands/immunology , DNA Methylation , Epigenesis, Genetic , Female , HLA-DP beta-Chains/immunology , HLA-DP beta-Chains/metabolism , Humans , Immune System/immunology , Infant , Interleukin-6/immunology , Interleukin-6/metabolism , Male , Pneumococcal Infections/immunology , Pneumococcal Infections/mortality , Pneumococcal Vaccines/administration & dosage , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
6.
Mol Cell ; 81(7): 1469-1483.e8, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33609448

ABSTRACT

We demonstrate that DNA hypomethylating agent (HMA) treatment can directly modulate the anti-tumor response and effector function of CD8+ T cells. In vivo HMA treatment promotes CD8+ T cell tumor infiltration and suppresses tumor growth via CD8+ T cell-dependent activity. Ex vivo, HMAs enhance primary human CD8+ T cell activation markers, effector cytokine production, and anti-tumor cytolytic activity. Epigenomic and transcriptomic profiling shows that HMAs vastly regulate T cell activation-related transcriptional networks, culminating with over-activation of NFATc1 short isoforms. Mechanistically, demethylation of an intragenic CpG island immediately downstream to the 3' UTR of the short isoform was associated with antisense transcription and alternative polyadenylation of NFATc1 short isoforms. High-dimensional single-cell mass cytometry analyses reveal a selective effect of HMAs on a subset of human CD8+ T cell subpopulations, increasing both the number and abundance of a granzyme Bhigh, perforinhigh effector subpopulation. Overall, our findings support the use of HMAs as a therapeutic strategy to boost anti-tumor immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CpG Islands/immunology , DNA Methylation/drug effects , Decitabine/pharmacology , Granzymes/immunology , Lymphocyte Activation/drug effects , DNA Methylation/immunology , Humans , NFATC Transcription Factors/immunology , Perforin/immunology
7.
Allergy Asthma Proc ; 41(5): 372-385, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32867892

ABSTRACT

Background: Allergic and autoimmune diseases comprise a group of inflammatory disorders caused by aberrant immune responses in which CD25+ forkhead box P3-positive regulatory T cells (Treg) cells that normally suppress inflammatory events are often poorly functioning. This has stimulated an intensive investigative effort to find ways of increasing Tregs as a method of therapy for these conditions. Commensal microbiota known to have health benefits in humans include the lactic acid-producing, probiotic bacteria B. longum subsp. infantis and Lactobacillus rhamnosus. Mechanistically, several mechanisms have been proposed to explain how probiotics may favorably affect host immunity, including the induction of Tregs. Analysis of emerging data from several laboratories, including our own, suggest that DNA methylation may be an important determinant of immune reactivity responsible for Treg induction. Although methylated CpG moieties in normal mammalian DNA are both noninflammatory and lack immunogenicity, unmethylated CpGs, found largely in microbial DNA, are immunostimulatory and display proinflammatory properties. Objective: We hypothesize that microbiota with more DNA methylation may potentiate Treg induction to a greater degree than microbiota with a lower content of methylation. The purpose of the present study was to test this hypothesis by studying the methylation status of whole genomic DNA (gDNA) and the Treg-inducing capacity of purified gDNA in each of the probiotic bacteria B. longum subsp. infantis and L. rhamnosus, and a pathogenic Escherichia coli strain B. Results: We showed that gDNA from B. longum subsp. infantis is a potent Treg inducer that displays a dose-dependent response pattern at a dose threshold of 20 µg of gDNA. No similar Treg-inducing responses were observed with the gDNA from L. rhamnosus or E. coli. We identified a unique CpG methylated motif in the gDNA sequencing of B. longum subsp. infantis which was not found in L. rhamnosus or E. coli strain B. Conclusion: Although the literature indicates that both B. longum subsp. infantis and L. rhamnosus strains contribute to health, our data suggest that they do so by different mechanisms. Further, because of its small molecular size, low cost, ease of synthesis, and unique Treg-inducing feature, this methylated CpG oligodeoxynucleotide (ODN) from B. longum would offer many attractive features for an ideal novel therapeutic vaccine candidate for the treatment of immunologic diseases, such as the allergic and autoimmune disorders, in which Treg populations are diminished.


Subject(s)
Bifidobacterium longum subspecies infantis/immunology , CpG Islands/immunology , DNA, Bacterial/immunology , Microbiota/immunology , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , DNA Methylation , Forkhead Transcription Factors/metabolism , Genome , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lacticaseibacillus rhamnosus/immunology , Lymphocyte Activation , Probiotics
8.
Cells ; 9(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32752173

ABSTRACT

The analysis of tumours using biomarkers in blood is transforming cancer diagnosis and therapy. Cancers are characterised by evolving genetic alterations, making it difficult to develop reliable and broadly applicable DNA-based biomarkers for liquid biopsy. In contrast to the variability in gene mutations, the methylation pattern remains generally constant during carcinogenesis. Thus, methylation more than mutation analysis may be exploited to recognise tumour features in the blood of patients. In this work, we investigated the possibility of using global CpG (CpG means a CG motif in the context of methylation. The p represents the phosphate. This is used to distinguish CG sites meant for methylation from other CG motifs or from mentions of CG content) island methylation profiles as a basis for the prediction of cancer state of patients utilising liquid biopsy samples. We retrieved existing GEO methylation datasets on hepatocellular carcinoma (HCC) and cell-free DNA (cfDNA) from HCC patients and healthy donors, as well as healthy whole blood and purified peripheral blood mononuclear cell (PBMC) samples, and used a random forest classifier as a predictor. Additionally, we tested three different feature selection techniques in combination. When using cfDNA samples together with solid tumour samples and healthy blood samples of different origin, we could achieve an average accuracy of 0.98 in a 10-fold cross-validation. In this setting, all the feature selection methods we tested in this work showed promising results. We could also show that it is possible to use solid tumour samples and purified PBMCs as a training set and correctly predict a cfDNA sample as cancerous or healthy. In contrast to the complete set of samples, the feature selections led to varying results of the respective random forests. ANOVA feature selection worked well with this training set, and the selected features allowed the random forest to predict all cfDNA samples correctly. Feature selection based on mutual information could also lead to better than random results, but LASSO feature selection would not lead to a confident prediction. Our results show the relevance of CpG islands as tumour markers in blood.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/immunology , CpG Islands/immunology , Liquid Biopsy/methods , Liver Neoplasms/immunology , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology
9.
Int Arch Allergy Immunol ; 181(12): 947-955, 2020.
Article in English | MEDLINE | ID: mdl-32799212

ABSTRACT

AIM: The role of CD4+ Treg in immune responses has been well established. More recently, a role of CD8+ T regulatory cells (CD8 Treg) in the regulation of immune responses in health and autoimmune diseases has been investigated. Furthermore, different investigators have used different markers to define CD8 Treg. Finally, regulatory effects of CD8 Treg have been studied against T-cell responses; however, their role in regulating B-cell proliferation and immunoglobulin production has not been evaluated. Therefore, in this study we examined the effect of two types of CD8 Treg on B-cell proliferation and immunoglobulin production. METHODS: Purified CD8+ T cells were activated with anti-CD3/CD28 for 48 h and then sorted into two different types of CD8 Treg as defined by two different sets of markers, CD8+CD183+CD197+CD45RA- and CD8+CD183+CD25highCD278+. Purified B cells were cocultured with sorted CD8 Treg at 1:1, 1:1/2, and 1:1/4 ratios and activated with anti-CD40 and CpG. B-cell proliferation was assessed by the CFSE dye dilution assay and immunoglobulin production by the ELISA assay. RESULTS: Our data show CD183+CD197+CD45RA-CD8 Treg significantly inhibited B-cell proliferation and inhibited IgM and IgG production but not IgA production at 1:1 ratio only. However, CD183+CD25highCD278+CD8 Treg inhibited significantly B-cell proliferation at 1:1 and 1:1/2 ratios and IgM, IgG, and IgA production at all ratios. CONCLUSION: CD8 Treg regulate B-cell responses, and CD183+CD25highCD278+CD8 Treg are more powerful regulators of B-cell proliferation and immunoglobulin production than CD183+CD197+CD45RA-CD8 Treg and, therefore, may be used as preferred markers for CD8 Treg.


Subject(s)
B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , T-Lymphocyte Subsets/immunology , Adult , Antibody Formation , Cell Proliferation , Cells, Cultured , CpG Islands/immunology , Female , Humans , Immunoglobulins/metabolism , Lymphocyte Activation , Male , Middle Aged , Young Adult
10.
J Leukoc Biol ; 108(4): 1329-1337, 2020 10.
Article in English | MEDLINE | ID: mdl-32421902

ABSTRACT

CD8 T cells play a crucial role in immune responses to virus infections and tumors. Naïve CD8 T lymphocytes after TCR stimulation undergo differentiation into CTLs and memory cells, which are essential sources of IFN-γ. We investigated IFN-γ production by CD8 T cell subsets found in nonimmune mice. A minor fraction of in vitro TCR-stimulated CD8 T cells produce IFN-γ, and it is regulated at the transcriptional level. Antigen inexperienced C57BL/6 mice present the coexistence of 2 populations. The main population exhibits a CD44low CD122low profile, which is compatible with naïve lymphocytes. The minor expresses a phenotype of immunologic memory, CD44hi CD122hi . Both subsets are able to produce IL-2 in response to TCR activation, but only the memory-like population is responsible for IFN-γ production. Similar to memory CD8 T cells, CD44hi CD8+ T cells also present a higher level of the transcriptional factor Eomes and a lower level of T-bet (Tbx21) mRNA than CD44low CD8+ T cells. The presence of the CD44hi CD8+ T cell population in nonimmune OT-I transgenic mice reveals that the population is generated independently of antigenic stimulation. CpG methylation is an efficient epigenetic mechanism for gene silencing. DNA methylation at posttranscriptional CpG sites in the Ifng promoter is higher in CD44low CD8+ T cells than in CD44hi CD8+ T cells. Thus, memory-like CD8 T cells have a distinct epigenetic pattern in the Ifng promoter and can rapidly produce IFN-γ in response to TCR stimulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CpG Islands/immunology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Interferon-gamma/genetics , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
11.
Proc Natl Acad Sci U S A ; 117(14): 7971-7980, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32205444

ABSTRACT

Timely resolution of bacterial infections critically depends on phagocytosis of invading pathogens by polymorphonuclear neutrophil granulocytes (PMNs), followed by PMN apoptosis and efferocytosis. Here we report that bacterial DNA (CpG DNA) and mitochondrial DNA impair phagocytosis and attenuate phagocytosis-induced apoptosis in human PMNs through Toll-like receptor 9 (TLR9)-mediated release of neutrophil elastase and proteinase 3 and subsequent down-regulation of the complement receptor C5aR. Consistently, CpG DNA delays pulmonary clearance of Escherichia coli in mice and suppresses PMN apoptosis, efferocytosis, and generation of proresolving lipid mediators, thereby prolonging lung inflammation evoked by E. coli Genetic deletion of TLR9 renders mice unresponsive to CpG DNA. We also show that aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 (17-epi-RvD1) through the receptor ALX/FPR2 antagonize cues from CpG DNA, preserve C5aR expression, restore impaired phagocytosis, and redirect human PMNs to apoptosis. Treatment of mice with 15-epi-LXA4 or 17-epi-RvD1 at the peak of inflammation accelerates clearance of bacteria, blunts PMN accumulation, and promotes PMN apoptosis and efferocytosis, thereby facilitating resolution of E. coli-evoked lung injury. Collectively, these results uncover a TLR9-mediated endogenous mechanism that impairs PMN phagocytosis and prolongs inflammation, and demonstrate both endogenous and therapeutic potential for 15-epi-LXA4 and 17-epi-RvD1 to restore impaired bacterial clearance and facilitate resolution of acute lung inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Escherichia coli Infections/immunology , Neutrophils/immunology , Phagocytosis/immunology , Pneumonia/immunology , Toll-Like Receptor 9/metabolism , Adult , Aged , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cells, Cultured , CpG Islands/immunology , DNA, Bacterial/immunology , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Female , Healthy Volunteers , Humans , Lipoxins/pharmacology , Lipoxins/therapeutic use , Lung/microbiology , Lung/pathology , Male , Mice , Middle Aged , Neutrophils/metabolism , Phagocytosis/drug effects , Pneumonia/drug therapy , Pneumonia/microbiology , Pneumonia/pathology , Primary Cell Culture , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Receptors, Lipoxin/immunology , Receptors, Lipoxin/metabolism
12.
Genes (Basel) ; 11(1)2020 01 11.
Article in English | MEDLINE | ID: mdl-31940818

ABSTRACT

During the last few years, mitochondrial DNA has attained much attention as a modulator of immune responses. Due to common evolutionary origin, mitochondrial DNA shares various characteristic features with DNA of bacteria, as it consists of a remarkable number of unmethylated DNA as 2'-deoxyribose cytidine-phosphate-guanosine (CpG) islands. Due to this particular feature, mitochondrial DNA seems to be recognized as a pathogen-associated molecular pattern by the innate immune system. Under the normal physiological situation, mitochondrial DNA is enclosed in the double membrane structure of mitochondria. However, upon pathological conditions, it is usually released into the cytoplasm. Growing evidence suggests that this cytosolic mitochondrial DNA induces various innate immune signaling pathways involving NLRP3, toll-like receptor 9, and stimulator of interferon genes (STING) signaling, which participate in triggering downstream cascade and stimulating to produce effector molecules. Mitochondrial DNA is responsible for inflammatory diseases after stress and cellular damage. In addition, it is also involved in the anti-viral and anti-bacterial innate immunity. Thus, instead of entire mitochondrial importance in cellular metabolism and energy production, mitochondrial DNA seems to be essential in triggering innate anti-microbial immunity. Here, we describe existing knowledge on the involvement of mitochondrial DNA in the anti-microbial immunity by modulating the various immune signaling pathways.


Subject(s)
Bacteria/immunology , DNA Methylation/immunology , DNA, Bacterial/immunology , DNA, Mitochondrial/immunology , Immunity, Innate , Mitochondria/immunology , Signal Transduction/immunology , Animals , CpG Islands/immunology , Humans
13.
J Allergy Clin Immunol ; 145(6): 1655-1663, 2020 06.
Article in English | MEDLINE | ID: mdl-31953105

ABSTRACT

BACKGROUND: Epigenetic signatures in the nasal epithelium, which is a primary interface with the environment and an accessible proxy for the bronchial epithelium, might provide insights into mechanisms of allergic disease. OBJECTIVE: We aimed to identify and interpret methylation signatures in nasal epithelial brushes associated with rhinitis and asthma. METHODS: Nasal epithelial brushes were obtained from 455 children at the 16-year follow-up of the Dutch Prevention and Incidence of Asthma and Mite Allergy birth cohort study. Epigenome-wide association studies were performed on children with asthma, rhinitis, and asthma and/or rhinitis (AsRh) by using logistic regression, and the top results were replicated in 2 independent cohorts of African American and Puerto Rican children. Significant CpG sites were related to environmental exposures (pets, active and passive smoking, and molds) during secondary school and were correlated with gene expression by RNA-sequencing (n = 244). RESULTS: The epigenome-wide association studies identified CpG sites significantly associated with rhinitis (n = 81) and AsRh (n = 75), but not with asthma. We significantly replicated 62 of 81 CpG sites with rhinitis and 60 of 75 with AsRh, as well as 1 CpG site with asthma. Methylation of cg03565274 was negatively associated with AsRh and positively associated with exposure to pets during secondary school. DNA methylation signals associated with AsRh were mainly driven by specific IgE-positive subjects. DNA methylation related to gene transcripts that were enriched for immune pathways and expressed in immune and epithelial cells. Nasal CpG sites performed well in predicting AsRh. CONCLUSIONS: We identified replicable DNA methylation profiles of asthma and rhinitis in nasal brushes. Exposure to pets may affect nasal epithelial methylation in relation to asthma and rhinitis.


Subject(s)
Asthma/genetics , DNA Methylation/genetics , Nasal Mucosa/immunology , Rhinitis/genetics , Adolescent , Black or African American/genetics , Asthma/immunology , Child , Cohort Studies , CpG Islands/genetics , CpG Islands/immunology , DNA Methylation/immunology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Epigenome/genetics , Epigenome/immunology , Epigenomics/methods , Epithelial Cells/immunology , Female , Genome-Wide Association Study/methods , Humans , Immunoglobulin E/genetics , Male , Respiratory Mucosa/immunology , Rhinitis/immunology
14.
Int Immunopharmacol ; 78: 106013, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31806571

ABSTRACT

There is an urgent need for efficient vaccines against the highly pathogenic avian influenza A viral strain H7N9. The duration and intensity of the immune response to H7N9 critically impacts the epidemiology of influenza viral infection at the population level. However, the insufficient immunogenicity of H7N9 raises concerns about vaccine efficacy. In this study, we evaluated the impact of immunization routes and the adjuvant CpG on the immune response to a split H7N9 vaccine in mice. Determination of humoral and cellular responses to the vaccine revealed that after four vaccine doses, high titers of H7N9-specific serum IgG, determined by the influenza hemagglutination inhibition (HI) assay, were induced through the intramuscular (i.m.) route and lasted for at least 40 weeks. CpG-adjuvanted immunization increased the levels of long-lived IFN-γ+ T cells and raised the Th1-biased IgG2a/IgG1 response ratio. In addition, aside from mucosal IgA, CpG-adjuvanted intranasal (i.n.) immunization elicited serum IgG and cellular responses of a similar duration and intensity to CpG-adjuvanted i.m. immunization. Mouse challenge assays demonstrated that 24 weeks following i.m. immunization without CpG or CpG-adjuvanted immunization through the i.m. or i.n. routes, both offered a high level of protection against H7N9 infection. These results indicate that efficient long-term protection against H7N9 can be achieved via the optimization of vaccination strategies, such as immunization doses, routes, and adjuvants.


Subject(s)
Antibodies, Viral/blood , CpG Islands/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Humans , Immunogenicity, Vaccine , Influenza Vaccines/administration & dosage , Influenza, Human/blood , Influenza, Human/immunology , Influenza, Human/virology , Injections, Intramuscular , Mice , Th1 Cells/immunology , Time Factors , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
16.
J Autoimmun ; 104: 102347, 2019 11.
Article in English | MEDLINE | ID: mdl-31607428

ABSTRACT

Thrombosis in the context of Cardiovascular disease (CVD) affects mainly the blood vessels supplying the heart, brain and peripheries and it is the leading cause of death worldwide. The pathophysiological thrombotic mechanisms are largely unknown. Heritability contributes to a 30% of the incidence of CVD. The remaining variation can be explained by life style factors such as smoking, dietary and exercise habits, environmental exposure to toxins, and drug usage and other comorbidities. Epigenetic variation can be acquired or inherited and constitutes an interaction between genes and the environment. Epigenetics have been implicated in atherosclerosis, ischemia/reperfusion damage and the cardiovascular response to hypoxia. Epigenetic regulators of gene expression are mainly the methylation of CpG islands, histone post translational modifications (PTMs) and microRNAs (miRNAs). These epigenetic regulators control gene expression either through activation or silencing. Epigenetic control is mostly dynamic and can potentially be manipulated to prevent or reverse the uncontrolled expression of genes, a trait that renders them putative therapeutic targets. In the current review, we systematically studied and present available data on epigenetic alterations implicated in thrombosis derived from human studies. Evidence of epigenetic alterations is observed in several thrombotic diseases such as Coronary Artery Disease and Cerebrovascular Disease, Preeclampsia and Antiphospholipid Syndrome. Differential CpG methylation and specific histone PTMs that control transcription of prothrombotic and proinflammatory genes have also been associated with predisposing factors of thrombosis and CVD, such us smoking, air pollution, hypertriglyceridemia, occupational exposure to particulate matter and comorbidities including cancer, Chronic Obstructive Pulmonary Disease and Chronic Kidney Disease. These clinical observations are further supported by in vitro experiments and indicate that epigenetic regulation affects the pathophysiology of thrombotic disorders with potential diagnostic or therapeutic utility.


Subject(s)
CpG Islands/immunology , DNA Methylation/immunology , Epigenesis, Genetic/immunology , Thrombosis/immunology , Histones/immunology , Humans , MicroRNAs/immunology , Protein Processing, Post-Translational/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Thrombosis/pathology
17.
J Control Release ; 315: 76-84, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31639342

ABSTRACT

Nucleic acids have been used as building blocks to assemble nanostructures by their sequence specific self-recognition properties, and resulting DNA architectures were applied as potential multifunctional drug carriers. Here, we report an amphiphilic lipid-DNA aggregate hybridized with pharmaceutically active DNA and peptide segments for cancer immunotherapy. The facile formulation of the CpG sequence and antigen peptide-bearing peptide nucleic acid representing immune-adjuvant and antigen, respectively, enabled the highly efficacious induction of antigen-specific immune activation. This immunotherapeutic formulation was evaluated in terms of multiple types of tumor growth and metastasis in vivo.


Subject(s)
DNA/administration & dosage , Immunotherapy/methods , Nanoparticles , Neoplasms/therapy , Adjuvants, Immunologic/administration & dosage , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , CpG Islands/immunology , DNA/immunology , Drug Carriers/chemistry , Lipids/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Peptide Nucleic Acids/administration & dosage , Peptide Nucleic Acids/immunology
18.
Vaccine ; 37(47): 6933-6941, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31383491

ABSTRACT

The novel H7N9 avian influenza A virus has caused human infections in China since 2013; some isolates from the fifth wave of infections have emerged as highly pathogenic avian influenza viruses. Recombinant hemagglutinin proteins of H7N9 viruses can be rapidly and efficiently produced with low-level biocontainment facilities. In this study, recombinant H7 antigen was obtained from engineered stable clones of Chinese Hamster Ovary (CHO) cells for subsequent large-scale production. The stable CHO cell clones were also adapted to grow in serum-free suspension cultures. To improve the immunogenicity of the recombinant H7 antigens, we evaluated the use of a novel combination adjuvant of PELC and CpG (PELC/CpG) to augment the anti-H7N9 immune responses in mice. We compared the effects with other adjuvants such as alum, AddaVax (MF59-like), and several Toll-like receptor ligands such as R848, CpG, and poly (I:C). With the PELC/CpG combination adjuvant, CHO cell-expressed rH7 antigens containing terminally sialylated complex type N-glycans were able to induce high titers of neutralizing antibodies in sera and conferred protection following live virus challenges. These data indicate that the CHO cell-expressed recombinant H7 antigens and a PELC/CpG combination adjuvant can be used for H7N9 subunit vaccine development.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CpG Islands/immunology , Hemagglutinins/immunology , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Recombinant Proteins/immunology , Alum Compounds/chemistry , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , CHO Cells , Cell Line , Cricetulus , Female , Hemagglutination Inhibition Tests/methods , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Mice , Mice, Inbred BALB C , Polysorbates/chemistry , Squalene/chemistry , Vaccines, Subunit/immunology
19.
Front Immunol ; 10: 1686, 2019.
Article in English | MEDLINE | ID: mdl-31428085

ABSTRACT

Objectives: To perform a cross-comparative analysis of DNA methylation in patients with systemic lupus erythematosus (SLE), patients with primary Sjögren's syndrome (pSS), and healthy controls addressing the question of epigenetic sharing and aiming to detect disease-specific alterations. Methods: DNA extracted from peripheral blood from 347 cases with SLE, 100 cases with pSS, and 400 healthy controls were analyzed on the Human Methylation 450k array, targeting 485,000 CpG sites across the genome. A linear regression model including age, sex, and blood cell type distribution as covariates was fitted, and association p-values were Bonferroni corrected. A random forest machine learning classifier was designed for prediction of disease status based on DNA methylation data. Results: We established a combined set of 4,945 shared differentially methylated CpG sites (DMCs) in SLE and pSS compared to controls. In pSS, hypomethylation at type I interferon induced genes was mainly driven by patients who were positive for Ro/SSA and/or La/SSB autoantibodies. Analysis of differential methylation between SLE and pSS identified 2,244 DMCs with a majority of sites showing decreased methylation in SLE compared to pSS. The random forest classifier demonstrated good performance in discerning between disease status with an area under the curve (AUC) between 0.83 and 0.96. Conclusions: The majority of differential DNA methylation is shared between SLE and pSS, however, important quantitative differences exist. Our data highlight neutrophil dysregulation as a shared mechanism, emphasizing the role of neutrophils in the pathogenesis of systemic autoimmune diseases. The current study provides evidence for genes and molecular pathways driving common and disease-specific pathogenic mechanisms.


Subject(s)
CpG Islands/immunology , DNA Methylation/immunology , Genome, Human , Lupus Erythematosus, Systemic , Sjogren's Syndrome , Adult , Aged , Female , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Male , Middle Aged , Sjogren's Syndrome/genetics , Sjogren's Syndrome/immunology
20.
Front Immunol ; 10: 1525, 2019.
Article in English | MEDLINE | ID: mdl-31333659

ABSTRACT

Historically, systemic self-inflammatory conditions were classified as either autoinflammatory and caused by the innate immune system or autoimmune and driven by adaptive immune responses. However, it became clear that reality is much more complex and that autoimmune/inflammatory conditions range along an "inflammatory spectrum" with primarily autoinflammatory vs. autoimmune conditions resembling extremes at either end. Epigenetic modifications influence gene expression and alter cellular functions without modifying the genomic sequence. Methylation of CpG DNA dinucleotides and/or their hydroxymethylation, post-translational modifications to amino termini of histone proteins, and non-coding RNA expression are main epigenetic events. The pathophysiology of autoimmune/inflammatory diseases has been closely linked with disease causing gene mutations (rare) or a combination of genetic susceptibility and epigenetic modifications arising from exposure to the environment (more common). Over recent years, progress has been made in understanding molecular mechanisms involved in systemic inflammation and the contribution of innate and adaptive immune responses. Epigenetic events have been identified as (i) central pathophysiological factors in addition to genetic disease predisposition and (ii) as co-factors determining clinical pictures and outcomes in individuals with monogenic disease. Thus, a complete understanding of epigenetic contributors to autoimmune/inflammatory disease will result in approaches to predict individual disease outcomes and the introduction of effective, target-directed, and tolerable therapies. Here, we summarize recent findings that signify the importance of epigenetic modifications in autoimmune/inflammatory disorders along the inflammatory spectrum choosing three examples: the autoinflammatory bone condition chronic nonbacterial osteomyelitis (CNO), the "mixed pattern" disorder psoriasis, and the autoimmune disease systemic lupus erythematosus (SLE).


Subject(s)
Autoimmune Diseases , DNA Methylation/immunology , Epigenesis, Genetic/immunology , Epigenomics , Protein Processing, Post-Translational/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CpG Islands/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology
SELECTION OF CITATIONS
SEARCH DETAIL