Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 555
Filter
1.
Front Cell Infect Microbiol ; 14: 1356542, 2024.
Article in English | MEDLINE | ID: mdl-38741892

ABSTRACT

Background and objectives: This study aimed to investigate the possible prognostic significance of interferon alpha-beta receptor subunit 2 (IFNAR2) and tyrosine kinase 2 (TYK2) expressions. Methods: We conducted a retrospective study including COVID-19 adult patients. All blood samples were collected before any interventions. The expressions of IFNAR2 and TYK2 were assessed using real-time PCR in venous blood samples of 54 cases and 56 controls. The transcript quantities of IFNAR2 and TYK2 genes were assessed using a Delta-Ct method. Results: Our findings show no significant differences in gene expression levels for IFNAR2 and TYK2 between patients who required oxygen (O2) therapy and those who did not (p-value = 0.732 and p-value = 0.629, respectively). Likewise, there were no significant differences in IFNAR2 and TYK2 expressions between patients hospitalized for less than 7 days and those hospitalized for 7 days or more (p-value = 0.455 and p-value = 0.626, respectively). We also observed a weak correlation between IFNAR2 expression and CRP (p-value = 0.045, r = 0.192). There was a negative correlation between the expression levels of IFNAR2 and TYK2 transcripts in COVID-19 patients (p-value = 0.044; partial correlation coefficient = -0.283). Additionally, IFNAR2 and TYK2 were significantly downregulated in the COVID-19 group compared to healthy subjects (p-value = 0.002 and p-value = 0.028, respectively). However, neither IFNAR2 nor TYK2 expression was significantly different between the case subgroups based on COVID-19 severity. The IFNAR2 ΔΔCt (B = -0.184, 95% CI: -0.524-0.157, p-value = 0.275) and the TYK2 ΔΔCt (B = 0.114, 95% CI: -0.268-0.496, p-value = 0.543) were not found to be significant predictors of hospitalization duration. The area under the curve (AUC) for IFNAR2 expression is 0.655 (p-value = 0.005, 95% CI: 0.554-0.757), suggesting its poor discriminative value. Conclusion: We were unable to comment definitively on the prognostic power of IFNAR2 and TYK2 expressions in COVID-19 patients, and larger-scale studies are needed. The principal limitations of this study included the lack of longitudinal analysis and limited sample size.


Subject(s)
COVID-19 , Receptor, Interferon alpha-beta , SARS-CoV-2 , TYK2 Kinase , Humans , COVID-19/genetics , TYK2 Kinase/genetics , TYK2 Kinase/metabolism , Retrospective Studies , Male , Female , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Prognosis , Middle Aged , Adult , SARS-CoV-2/genetics , Aged
2.
J Innate Immun ; 16(1): 226-247, 2024.
Article in English | MEDLINE | ID: mdl-38527452

ABSTRACT

INTRODUCTION: While TLR ligands derived from microbial flora and pathogens are important activators of the innate immune system, a variety of factors such as intracellular bacteria, viruses, and parasites can induce a state of hyperreactivity, causing a dysregulated and potentially life-threatening cytokine over-response upon TLR ligand exposure. Type I interferon (IFN-αß) is a central mediator in the induction of hypersensitivity and is strongly expressed in splenic conventional dendritic cells (cDC) and marginal zone macrophages (MZM) when mice are infected with adenovirus. This study investigates the ability of adenoviral infection to influence the activation state of the immune system and underlines the importance of considering this state when planning the treatment of patients. METHODS: Infection with adenovirus-based vectors (Ad) or pretreatment with recombinant IFN-ß was used as a model to study hypersensitivity to lipopolysaccharide (LPS) in mice, murine macrophages, and human blood samples. The TNF-α, IL-6, IFN-αß, and IL-10 responses induced by LPS after pretreatment were measured. Mouse knockout models for MARCO, IFN-αßR, CD14, IRF3, and IRF7 were used to probe the mechanisms of the hypersensitive reaction. RESULTS: We show that, similar to TNF-α and IL-6 but not IL-10, the induction of IFN-αß by LPS increases strongly after Ad infection. This is true both in mice and in human blood samples ex vivo, suggesting that the regulatory mechanisms seen in the mouse are also present in humans. In mice, the scavenger receptor MARCO on IFN-αß-producing cDC and splenic marginal zone macrophages is important for Ad uptake and subsequent cytokine overproduction by LPS. Interestingly, not all IFN-αß-pretreated macrophage types exposed to LPS exhibit an enhanced TNF-α and IL-6 response. Pretreated alveolar macrophages and alveolar macrophage-like murine cell lines (MPI cells) show enhanced responses, while bone marrow-derived and peritoneal macrophages show a weaker response. This correlates with the respective absence or presence of the anti-inflammatory IL-10 response in these different macrophage types. In contrast, Ad or IFN-ß pretreatment enhances the subsequent induction of IFN-αß in all macrophage types. IRF3 is dispensable for the LPS-induced IFN-αß overproduction in infected MPI cells and partly dispensable in infected mice, while IRF7 is required. The expression of the LPS co-receptor CD14 is important but not absolutely required for the elicitation of a TNF-α over-response to LPS in Ad-infected mice. CONCLUSION: Viral infections or application of virus-based vaccines induces type I interferon and can tip the balance of the innate immune system in the direction of hyperreactivity to a subsequent exposure to TLR ligands. The adenoviral model presented here is one example of how multiple factors, both environmental and genetic, affect the physiological responses to pathogens. Being able to measure the current reactivity state of the immune system would have important benefits for infection-specific therapies and for the prevention of vaccination-elicited adverse effects.


Subject(s)
Adenoviridae , Cytokines , Interferon Regulatory Factor-3 , Lipopolysaccharides , Macrophages , Mice, Knockout , Animals , Mice , Lipopolysaccharides/immunology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Macrophages/immunology , Cytokines/metabolism , Mice, Inbred C57BL , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Genetic Vectors , Adenoviridae Infections/immunology , Interferon Type I/metabolism , Lipopolysaccharide Receptors/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Cells, Cultured , Dendritic Cells/immunology , Interferon-beta/metabolism
3.
Am J Physiol Lung Cell Mol Physiol ; 326(5): L551-L561, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38375579

ABSTRACT

Excessive or persistent inflammation may have detrimental effects on lung structure and function. Currently, our understanding of conserved host mechanisms that control the inflammatory response remains incompletely understood. In this study, we investigated the role of type I interferon signaling in the inflammatory response against diverse clinically relevant stimuli. Using mice deficient in type I interferon signaling (IFNAR1-/-), we demonstrate that the absence of interferon signaling resulted in a robust and persistent inflammatory response against Pseudomonas aeruginosa, lipopolysaccharide, and chemotherapeutic agent bleomycin. The elevated inflammatory response in IFNAR1-/- mice was manifested as elevated myeloid cells, such as macrophages and neutrophils, in the bronchoalveolar lavage. The inflammatory cell response in the IFNAR1-/- mice persisted to 14 days and there is impaired recovery and fibrotic remodeling of the lung in IFNAR1-/- mice after bleomycin injury. In the Pseudomonas infection model, the elevated inflammatory cell response led to improved bacterial clearance in IFNAR1-/- mice, although there was similar lung injury and survival. We performed RNA sequencing of lung tissue in wild-type and IFNAR1-/- mice after LPS and bleomycin injury. Our unbiased analysis identified differentially expressed genes between IFNAR1-/- and wild-type mice, including previously unknown regulation of nucleotide-binding oligomerization domain (NOD)-like receptor signaling, retinoic acid-inducible gene-I (RIG-I) signaling, and necroptosis pathway by type I interferon signaling in both models. These data provide novel insights into the conserved anti-inflammatory mechanisms of the type I interferon signaling.NEW & NOTEWORTHY Type I interferons are known for their antiviral activities. In this study, we demonstrate a conserved anti-inflammatory role of type I interferon signaling against diverse stimuli in the lung. We show that exacerbated inflammatory response in the absence of type I interferon signaling has both acute and chronic consequences in the lung including structural changes.


Subject(s)
Interferon Type I , Lung , Mice, Inbred C57BL , Mice, Knockout , Receptor, Interferon alpha-beta , Signal Transduction , Animals , Interferon Type I/metabolism , Lung/metabolism , Lung/immunology , Lung/pathology , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Mice , Bleomycin , Pseudomonas aeruginosa , Lipopolysaccharides/pharmacology , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas Infections/pathology , Pseudomonas Infections/microbiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/immunology , Male
4.
J Antibiot (Tokyo) ; 77(2): 102-110, 2024 02.
Article in English | MEDLINE | ID: mdl-38102186

ABSTRACT

We aimed to investigate the effects of tumor necrosis factor (TNF)-α on the expression of interferon α/ß receptor subunit 1 (IFNAR1) and cervical squamous cancer (CSCC) resistance to Cisplatin, as well as the underlying mechanisms. Kaplan-Meier analysis was used to plot the overall survival curves. SiHa cells were treated with 20 ng/ml TNF-α to determine cell proliferation in human CSCC cells and the expression of IFNAR1. The effects of TNF-α on the downstream signaling pathway, including casein kinase 1α (CK1α), were investigated using the caspase protease inhibitor FK009, the c-Jun kinase inhibitor SP600125, and the nuclear factor kappa-B inhibitor ammonium pyrrolidinedithiocarbamate (PDTC). TNF-α induced down-regulation of IFNAR1 in human CSCC cells and promoted proliferation of SiHa cells. SiHa cells were transfected with the catalytic inactive mutant CK1α K49A, and the ability of TNF-α to induce down-regulation of IFNAR1 expression was found to be significantly diminished in this context. FK009 and PDTC had no obvious effect on the expression of CK1α, however, SP600125 significantly reduced the expression of CK1α in the presence of TNF-α. SiHa cells treated with TNF-α showed reduced sensitivity to Cisplatin and exhibited higher cell viability, while the sensitivity of SiHa cells to Cisplatin was restored after treatment with CK1α inhibitor D4476. Additionally, we constructed a TNF-α overexpressing SiHa cell line and a transplanted tumor model. The results were similar to those of in vitro efficacy. We demonstrate that TNF-α-induced down-regulation of type I interferon receptor contributes to acquired resistance of cervical squamous cancer to Cisplatin.


Subject(s)
Anthracenes , Carcinoma, Squamous Cell , Proline/analogs & derivatives , Thiocarbamates , Uterine Cervical Neoplasms , Female , Humans , Cisplatin/pharmacology , Cisplatin/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Down-Regulation , Uterine Cervical Neoplasms/drug therapy , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Apoptosis
5.
Immunity ; 57(1): 68-85.e11, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38141610

ABSTRACT

Tissue factor (TF), which is a member of the cytokine receptor family, promotes coagulation and coagulation-dependent inflammation. TF also exerts protective effects through unknown mechanisms. Here, we showed that TF bound to interferon-α receptor 1 (IFNAR1) and antagonized its signaling, preventing spontaneous sterile inflammation and maintaining immune homeostasis. Structural modeling and direct binding studies revealed binding of the TF C-terminal fibronectin III domain to IFNAR1, which restricted the expression of interferon-stimulated genes (ISGs). Podocyte-specific loss of TF in mice (PodΔF3) resulted in sterile renal inflammation, characterized by JAK/STAT signaling, proinflammatory cytokine expression, disrupted immune homeostasis, and glomerulopathy. Inhibiting IFNAR1 signaling or loss of Ifnar1 expression in podocytes attenuated these effects in PodΔF3 mice. As a heteromer, TF and IFNAR1 were both inactive, while dissociation of the TF-IFNAR1 heteromer promoted TF activity and IFNAR1 signaling. These data suggest that the TF-IFNAR1 heteromer is a molecular switch that controls thrombo-inflammation.


Subject(s)
Signal Transduction , Thromboplastin , Animals , Mice , Inflammation , Interferon-alpha , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Thromboplastin/genetics
6.
Nat Commun ; 14(1): 2007, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37037810

ABSTRACT

Viral tropism within the brain and the role(s) of vertebrate immune response to neurotropic flaviviruses infection is largely understudied. We combine multimodal imaging (cm-nm scale) with single nuclei RNA-sequencing to study Langat virus in wildtype and interferon alpha/beta receptor knockout (Ifnar-/-) mice to visualize viral pathogenesis and define molecular mechanisms. Whole brain viral infection is imaged by Optical Projection Tomography coregistered to ex vivo MRI. Infection is limited to grey matter of sensory systems in wildtype mice, but extends into white matter, meninges and choroid plexus in Ifnar-/- mice. Cells in wildtype display strong type I and II IFN responses, likely due to Ifnb expressing astrocytes, infiltration of macrophages and Ifng-expressing CD8+ NK cells, whereas in Ifnar-/-, the absence of this response contributes to a shift in cellular tropism towards non-activated resident microglia. Multimodal imaging-transcriptomics exemplifies a powerful way to characterize mechanisms of viral pathogenesis and tropism.


Subject(s)
Encephalitis Viruses, Tick-Borne , Interferon Type I , Ticks , Mice , Animals , Interferon Type I/metabolism , Neurons/metabolism , Mice, Knockout , Brain/diagnostic imaging , Brain/metabolism , Encephalitis Viruses, Tick-Borne/genetics , Encephalitis Viruses, Tick-Borne/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Tropism , Ticks/metabolism , Mice, Inbred C57BL
7.
Nat Cell Biol ; 25(3): 425-438, 2023 03.
Article in English | MEDLINE | ID: mdl-36797476

ABSTRACT

Activation of the JAK-STAT pathway by type I interferons (IFNs) requires clathrin-dependent endocytosis of the IFN-α and -ß receptor (IFNAR), indicating a role for endosomal sorting in this process. The molecular machinery that brings the selective activation of IFN-α/ß-induced JAK-STAT signalling on endosomes remains unknown. Here we show that the constitutive association of STAM with IFNAR1 and TYK2 kinase at the plasma membrane prevents TYK2 activation by type I IFNs. IFN-α-stimulated IFNAR endocytosis delivers the STAM-IFNAR complex to early endosomes where it interacts with Hrs, thereby relieving TYK2 inhibition by STAM and triggering signalling of IFNAR at the endosome. In contrast, when stimulated by IFN-ß, IFNAR signalling occurs independently of Hrs as IFNAR is sorted to a distinct endosomal subdomain. Our results identify the molecular machinery that controls the spatiotemporal activation of IFNAR by IFN-α and establish the central role of endosomal sorting in the differential regulation of JAK-STAT signalling by IFN-α and IFN-ß.


Subject(s)
Interferon Type I , Janus Kinases , Janus Kinases/metabolism , Signal Transduction/physiology , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Interferon-alpha/pharmacology , Interferon-alpha/metabolism , Endosomes/metabolism
8.
J Interferon Cytokine Res ; 42(11): 580-589, 2022 11.
Article in English | MEDLINE | ID: mdl-36346319

ABSTRACT

Type I interferons (IFN-Is) play central roles in regulating immune responses. The role of IFNAR2 in IFN-I signaling is an open question since a previous report showed that IFNß was still functional in the absence of IFNAR2 in mice. In this study, we report that IFN-I signaling in human monocyte-derived THP1 cells absolutely depends on IFNAR2, as determined by using a knockout mutant made by CRISPR/Cas9. Additionally, we demonstrated that a 7-bp deletion mutant (Δ7) of IFNAR2 remains responsive to IFNß stimulation and upregulates a subset of interferon-stimulated genes (s-ISGs). The s-ISGs largely overlap with tonic ISGs, which depend on the basal expression level of IFN-I. We also showed that IFN signaling in Δ7 still depends on IFNAR2. Then, we found that the 7-bp deletion in the genome results in the loss of the entire third exon (42 bp) from the mRNA and in the expression of a functionally impaired IFNAR2. These findings clarified the requirement of IFNAR2 for human IFN-I signaling and highlighted that caution should be used with CRISPR/Cas9 technology to prevent misleading interpretations caused by residual protein expression due to exon skipping or other mechanisms.


Subject(s)
Interferon Type I , Receptor, Interferon alpha-beta , Humans , Antiviral Agents/pharmacology , CRISPR-Cas Systems , Exons/genetics , Interferon Type I/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism
9.
Immunity ; 55(11): 2074-2084.e5, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36243008

ABSTRACT

Down syndrome (DS) is typically caused by triplication of chromosome 21. Phenotypically, DS presents with developmental, neurocognitive, and immune features. Epidemiologically, individuals with DS have less frequent viral infection, but when present, these infections lead to more severe disease. The potent antiviral cytokine type I Interferon (IFN-I) receptor subunits IFNAR1 and IFNAR2 are located on chromosome 21. While increased IFNAR1/2 expression initially caused hypersensitivity to IFN-I, it triggered excessive negative feedback. This led to a hypo-response to subsequent IFN-I stimuli and an ensuing viral susceptibility in DS compared to control cells. Upregulation of IFNAR2 expression phenocopied the DS IFN-I dynamics independent of trisomy 21. CD14+ monocytes from individuals with DS exhibited markers of prior IFN-I exposure and had muted responsiveness to ex vivo IFN-I stimulation. Our findings unveil oscillations of hyper- and hypo-response to IFN-I in DS, predisposing individuals to both lower incidence of viral disease and increased infection-related morbidity and mortality.


Subject(s)
Down Syndrome , Interferon Type I , Humans , Interferon Type I/metabolism , Down Syndrome/genetics , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Antiviral Agents , Disease Susceptibility , Receptors, Interferon/metabolism
10.
Sci Rep ; 12(1): 16488, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36182964

ABSTRACT

Blood-brain barrier (BBB) dysfunction is emerging as a key pathogenic factor in the progression of Alzheimer's disease (AD), where increased microvascular endothelial permeability has been proposed to play an important role. However, the molecular mechanisms leading to increased brain microvascular permeability in AD are not fully understood. We studied brain endothelial permeability in female APPswe/PS1∆E9 (APP/PS1) mice which constitute a transgenic mouse model of amyloid-beta (Aß) amyloidosis and found that permeability increases with aging in the areas showing the greatest amyloid plaque deposition. We performed an unbiased bulk RNA-sequencing analysis of brain endothelial cells (BECs) in female APP/PS1 transgenic mice. We observed that upregulation of interferon signaling gene expression pathways in BECs was among the most prominent transcriptomic signatures in the brain endothelium. Immunofluorescence analysis of isolated BECs from female APP/PS1 mice demonstrated higher levels of the Type I interferon-stimulated gene IFIT2. Immunoblotting of APP/PS1 BECs showed downregulation of the adherens junction protein VE-cadherin. Stimulation of human brain endothelial cells with interferon-ß decreased the levels of the adherens junction protein VE-cadherin as well as tight junction proteins Occludin and Claudin-5 and increased barrier leakiness. Depletion of the Type I interferon receptor in human brain endothelial cells prevented interferon-ß-induced VE-cadherin downregulation and restored endothelial barrier integrity. Our study suggests that Type I interferon signaling contributes to brain endothelial dysfunction in AD.


Subject(s)
Alzheimer Disease , Interferon Type I , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Claudin-5/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium/metabolism , Female , Humans , Interferon Type I/metabolism , Interferon-beta/metabolism , Mice , Mice, Transgenic , Occludin/metabolism , Plaque, Amyloid/pathology , RNA/metabolism , Receptor, Interferon alpha-beta/metabolism , Tight Junction Proteins/metabolism
11.
Cytokine ; 159: 156008, 2022 11.
Article in English | MEDLINE | ID: mdl-36063748

ABSTRACT

IFN-α receptor (IFNAR) is critical for maintaining the crosstalk between cancer cells and lymphocytes. We investigated IFNAR1 expression in peripheral blood CD4+ and CD8+ T cells and explored their relationships with plasma cytokines, chemosensitivity and infiltrated T cells in the tumor microenvironment (TME) of colorectal cancer (CRC). The levels of IFNAR1, IFN-γ, and PD1 in peripheral T cells were tested using flow cytometry. Immunohistochemical staining of IFNAR1 in CRC tissues was performed. A cytometric bead array was used to determine the plasma concentrations of cytokines. In CRC patients, IFNAR1 levels were significantly increased in peripheral blood T cells, and plasma IL-6 levels were also significantly increased. Pearson correlation analysis revealed that IFNAR1 expression in CD8+ T cells was negatively associated with plasma IL-2, IFN-γ, and TNFα. IFNAR1 expression in CD4+ T cells was positively associated with TME infiltrated levels of CD8+ T cells. The levels of CD8+ T cells with IFNAR1 and plasma IFN-γ were associated with chemosensitivity. Collectively, IFNAR1 levels in CD4+ and CD8+ T cells were significantly upregulated in CRC patients and positively associated with T-cell infiltration. IFNAR1 may be a chemotherapy biomarker for predicting response.


Subject(s)
Colorectal Neoplasms , Lymphocytes, Tumor-Infiltrating , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Colorectal Neoplasms/metabolism , Cytokines/metabolism , Humans , Interleukin-2/metabolism , Interleukin-6/metabolism , Receptor, Interferon alpha-beta/metabolism , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
12.
Front Immunol ; 13: 961094, 2022.
Article in English | MEDLINE | ID: mdl-36119026

ABSTRACT

Ov-ASP-1 (rASP-1), a parasite-derived protein secreted by the helminth Onchocerca volvulus, is an adjuvant which enhances the potency of the influenza trivalent vaccine (IIV3), even when used with 40-fold less IIV3. This study is aimed to provide a deeper insight into the molecular networks that underline the adjuvanticity of rASP-1. Here we show that rASP-1 stimulates mouse CD11c+ bone marrow-derived dendritic (BMDCs) to secrete elevated levels of IL-12p40, TNF-α, IP-10 and IFN-ß in a TRIF-dependent but MyD88-independent manner. rASP-1-activated BMDCs promoted the differentiation of naïve CD4+ T cells into Th1 cells (IFN-γ+) that was TRIF- and type I interferon receptor (IFNAR)-dependent, and into Tfh-like cells (IL21+) and Tfh1 (IFN-γ+ IL21+) that were TRIF-, MyD88- and IFNAR-dependent. rASP-1-activated BMDCs promoted the differentiation of naïve CD4+ T cells into Th17 (IL-17+) cells only when the MyD88 pathway was inhibited. Importantly, rASP-1-activated human blood cDCs expressed upregulated genes that are associated with DC maturation, type I IFN and type II IFN signaling, as well as TLR4-TRIF dependent signaling. These activated cDCs promoted the differentiation of naïve human CD4+ T cells into Th1, Tfh-like and Th17 cells. Our data thus confirms that the rASP-1 is a potent innate adjuvant that polarizes the adaptive T cell responses to Th1/Tfh1 in both mouse and human DCs. Notably, the rASP-1-adjuvanted IIV3 vaccine elicited protection of mice from a lethal H1N1 infection that is also dependent on the TLR4-TRIF axis and IFNAR signaling pathway, as well as on its ability to induce anti-IIV3 antibody production.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Adaptor Proteins, Vesicular Transport/metabolism , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic , Animals , Chemokine CXCL10/metabolism , Humans , Interleukin-12 Subunit p40 , Interleukin-17/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
J Reprod Immunol ; 153: 103677, 2022 09.
Article in English | MEDLINE | ID: mdl-35907379

ABSTRACT

Interferon-tau (IFNτ), as an antiluteolytic factor secreted by trophoderm during the pregnancy of ruminants, actually functions by activating the IFNτ receptor 1 (IFNAR1) and IFNτ receptor 2 (IFNAR2). However, it has not been clearly understood how IFNτ-IFNAR cascade regulation processes between the embryo and uterine epithelial cells in ruminants. In this study, we found the expression and location of IFNτ in the bovine blastocysts from different production sources. IFNτ, IFNAR1 and IFNAR2 were all located in the trophoblast cells of the blastocyst. However, the fluorescence intensity of IFNAR1 was consistent with that of IFNτ. Antagonizing the expressions of IFNAR1 and IFNAR2 in embryos and co-culture with endometrial epithelium cells (EECs) reduced the expressions of Integrin αv ß3, WNT7A, and ISG15 in EECs. Knocking out IFNAR1 and IFNAR2 reduce the expressions of Integrin αv ß3 and WNT7A in EECs, the deletion of IFNAR2 gene has a greater impact than that of IFNAR1 gene. IFNAR1-/IFNAR2+ and IFNAR1+/IFNAR2- EECs were co-cultured with IVF embryos, the expression of Integrin αv ß3 was inhibited, and the inhibition of IFNAR1+/IFNAR2- was much stronger, and the expression of WNT7A was not inhibited. The expressions of Integrin αv ß3 and WNT7A did not change significantly after IFNAR1-/IFNAR2+ and IFNAR1+/IFNAR2- co-culture with PA embryos. All of these results strongly suggest that specific activation of embryonic IFNAR1 and endometrial IFNAR2 induced by embryonic IFNτ directs normal uterine preparation for bovine early implantation.


Subject(s)
Interferon Type I/metabolism , Pregnancy Proteins/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Blastocyst/metabolism , Cattle , Embryo Implantation , Endometrium/metabolism , Female , Humans , Pregnancy , Trophoblasts/metabolism
14.
J Immunol ; 209(4): 806-819, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35906001

ABSTRACT

In mammals, type I IFNs, which commonly contain one or two disulfide bonds, activate the JAK-STAT signaling pathway through binding to the common cell surface receptor formed by IFN-α/ß receptor (IFNAR)1 and IFNAR2 subunits. Although type I IFNs are also known to be essential for antiviral defense in teleost fish, very little is known about mechanisms underlying the recognition of fish type I IFNs by associated receptors. In this study, we demonstrate that a type I IFN of large yellow croaker Larimichthys crocea (LcIFNi), belonging to a new subgroup of fish type I IFNs, triggers antiviral response via the conserved JAK-STAT pathway through stable binding with a heterodimeric receptor comprising subunits LcCRFB5 and LcCRFB2. LcIFNi binds to LcCRFB5 with a much higher affinity than to LcCRFB2. Furthermore, we determined the crystal structure of LcIFNi at a 1.39 Å resolution. The high-resolution structure is, to our knowledge, the first reported structure of a type I IFN with three disulfide bonds, all of which were found to be indispensable for folding and stability of LcIFNi. Using structural analysis, mutagenesis, and biochemical assays, we identified key LcIFNi residues involved in receptor interaction and proposed a structural model of LcIFNi bound to the LcCRFB2-LcCRFB5 receptor. The results show that LcIFNi-LcCRFB2 exhibits a similar binding pattern to human IFN-ω-IFNAR2, whereas the binding pattern of LcIFNi-LcCRFB5 is quite different from that of IFN-ω-IFNAR1. Altogether, our findings reveal the structural basis for receptor interaction and signaling of a type I IFN with three disulfide bonds and provide new insights into the mechanisms underlying type I IFN recognition in teleosts.


Subject(s)
Perciformes , Signal Transduction , Animals , Antiviral Agents , Disulfides/metabolism , Fishes/metabolism , Humans , Janus Kinases/metabolism , Mammals/metabolism , Receptor, Interferon alpha-beta/metabolism , STAT Transcription Factors/metabolism
15.
Placenta ; 126: 150-159, 2022 08.
Article in English | MEDLINE | ID: mdl-35816776

ABSTRACT

INTRODUCTION: Throughout human pregnancy there is a delicate balance between the maintenance of a proliferative, trophoblast stem cell pool (TSC) and the differentiation from TSC to placental cell sub-lineages like the syncytiotrophoblast (STB). The STB is comprised of multinucleated cells that come into direct contact with maternal blood and provides the first line of defense to protect the fetus from maternal infections. The differentiation of TSC towards STB is primarily driven by human endogenous retroviruses (HERV), specifically Syncytin-1 (ERVW-1) and Syncytin-2 (ERVFRD-1). Beyond cell fusion, there is also evidence to suggest they can regulate cell proliferation and an antiviral response in other cell types. Therefore, we hypothesized that HERV can regulate cell proliferation as well as an antiviral response in TSCs. METHOD: shRNA was used to knockdown ERVW-1 in TSCs and revealed reduction in cell proliferation, differentiation, and cell fusion. RT-qPCR and flow cytometry was used to measure other HERV and the presence of Type I and Type II interferon receptors. RESULTS: ERVW-1 knockdown (KD) TSCs had a significantly longer cell doubling time and reduced expression of the proliferation marker Ki67. ERVW-1 KD cells also demonstrated a marked deficiency in the ability to differentiate. Interestingly, ERVFRD-1 was upregulated in both ERVW-1 KD TSC and STB cells compared to controls. Finally, we found that the Type I interferon receptors, IFNAR1 and IFNAR2 were significantly increased in ERVW-1 KD STB cells. DISCUSSION: These findings uncover critical HERV functions in the trophoblasts and a novel role for ERVW-1 during early human placental development.


Subject(s)
Endogenous Retroviruses , Trophoblasts , Antiviral Agents , Cell Proliferation , Endogenous Retroviruses/genetics , Female , Gene Products, env , Humans , Placenta/metabolism , Pregnancy , Pregnancy Proteins , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Trophoblasts/metabolism
16.
Int Endod J ; 55(10): 1042-1052, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35869806

ABSTRACT

AIM: The aim of this study was to evaluate the IFI16 and IFN-α/ß receptors expression during the genesis and development of experimental apical periodontitis (AP) in mice teeth. METHODOLOGY: Apical periodontitis was induced in the lower first molars of 40 C57BL/6 mice. They were divided according to the experimental periods 2, 7, 14, 21 and 42 days (n = 8 per group). Five animals were used as a control group (without AP). Specimens were submitted to histological processing for description of the inflammatory process, immunostaining for the presence/absence and localization of IFI16 and IFN-α/ß receptors (qualitative and semi-quantitative analysis) and tartrate-resistant acid phosphatase (TRAP) histoenzimology. RESULTS: The results showed a gradual development of AP over the experimental times. The expression of IFI16 was noticeably more exacerbated in the experimental early period (day 2) whilst the lowest expression was observed in the control group (p = .02). For IFN-α/ß receptors, a higher intensity staining was observed 42 days after AP induction, that was statistically different from the control group (p = .02). In addition, the number of TRAP-positive cells was higher on the later periods (days 21 and 42; p < .001). CONCLUSION: IFI16 protein expression was highest during the early periods after AP induction in mice teeth, whilst IFN-α/ß receptor expression was highest after AP became established.


Subject(s)
Interferon-gamma , Nuclear Proteins/metabolism , Periapical Periodontitis , Phosphoproteins/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Mice , Mice, Inbred C57BL , Molar/pathology , Osteoclasts/metabolism , Periapical Periodontitis/pathology
17.
Front Cell Infect Microbiol ; 12: 862582, 2022.
Article in English | MEDLINE | ID: mdl-35586249

ABSTRACT

Irg1 is an enzyme that generates itaconate, a metabolite that plays a key role in the regulation of inflammatory responses. Previous studies have implicated Irg1 as an important mediator in preventing excessive inflammation and tissue damage in Mycobacterium tuberculosis (Mtb) infection. Here, we investigated the pattern recognition receptors and signaling pathways by which Mtb triggers Irg1 gene expression by comparing the responses of control and genetically deficient BMDMs. Using this approach, we demonstrated partial roles for TLR-2 (but not TLR-4 or -9), MyD88 and NFκB signaling in Irg1 induction by Mtb bacilli. In addition, drug inhibition studies revealed major requirements for phagocytosis and endosomal acidification in Irg1 expression triggered by Mtb but not LPS or PAM3CSK4. Importantly, the Mtb-induced Irg1 response was highly dependent on the presence of the bacterial ESX-1 secretion system, as well as host STING and Type I IFN receptor (IFNAR) signaling with Type II IFN (IFN-γ) signaling playing only a minimal role. Based on these findings we hypothesize that Mtb induces Irg1 expression in macrophages via the combination of two independent triggers both dependent on bacterial phagocytosis: 1) a major signal stimulated by phagocytized Mtb products released by an ESX-1-dependent mechanism into the cytosol where they activate the STING pathway leading to Type I-IFN production, and 2) a secondary TLR-2, MyD88 and NFκB dependent signal that enhances Irg1 production independently of Type I IFN induction.


Subject(s)
Hydro-Lyases , Macrophages , Membrane Proteins , Mycobacterium tuberculosis , Receptor, Interferon alpha-beta , Toll-Like Receptor 2 , Adaptor Proteins, Signal Transducing/metabolism , Animals , Enzyme Induction , Hydro-Lyases/biosynthesis , Hydro-Lyases/immunology , Macrophages/immunology , Macrophages/microbiology , Membrane Proteins/metabolism , Mice , Mycobacterium tuberculosis/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phagocytosis , Receptor, Interferon alpha-beta/metabolism , Toll-Like Receptor 2/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology
18.
Front Immunol ; 13: 814491, 2022.
Article in English | MEDLINE | ID: mdl-35464475

ABSTRACT

Rotavirus (RV) infection induces strong adaptive immunity. While protection from reinfection requires humoral immunity, initial clearance of infection depends on cytotoxic CD8 T cells. Type I classical dendritic cells (cDC1) excel at CD8 T cell induction through cross-presentation and are essential for optimal cytotoxicity towards RV. Upon sensing of infection-induced innate immune signals through pattern recognition receptors (PRRs), cumulating in autocrine type I interferon (IFN) signaling, cDC1 mature and migrate to the draining lymph nodes (LNs), where they prime adaptive immune cells. To analyze which PRR pathways lead to robust cytotoxicity in the context of RV infection, we measured RV-specific CD8 T cell priming in mice deficient for Toll-like receptor 3 (TLR3), recognizing double-stranded RNA, or for MyD88, the adapter for all other TLRs and IL-1 family cytokines. Individual TLR3- and MyD88-mediated signaling was not required for the priming of CD8 T cell responses to RV and neither deficiency impacted on RV clearance. Surprisingly, the accumulation of RV-specific CD8 T cells was also not altered in the absence of type I IFN signaling, while their ability to produce IFNγ and granzyme were blunted. Together, this suggests a substantial level of redundancy in the sensing of RV infection and the translation of signals into protective CD8 T cell immunity.


Subject(s)
Rotavirus Infections , Rotavirus , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD8-Positive T-Lymphocytes , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Toll-Like Receptor 3/metabolism
19.
Cytokine ; 153: 155849, 2022 05.
Article in English | MEDLINE | ID: mdl-35339044

ABSTRACT

As a member of JAK family of non-receptor tyrosine kinases, TYK2 has a crucial role in regulation of immune responses. This protein has a crucial role in constant expression of IFNAR1 on surface of cells and initiation of type I IFN signaling. In the current study, we measured expression of IFNAR1 and TYK2 levels in venous blood samples of COVID-19 patients and matched controls. TYK2 was significantly down-regulated in male patients compared with male controls (RME = 0.34, P value = 0.03). Though, levels of TYK2 were not different between female cases and female controls, or between ICU-admitted and non-ICU-admitted cases. Expression of IFNAR1 was not different either between COVID-19 cases and controls or between patients required ICU admission and non-ICU-admitted cases. However, none of these transcripts can properly diffrentiate COVID-19 cases from controls or separate patients based on disease severity. The current study proposes down-regulation of TYK2 as a molecular mechanism for incapacity of SARS-CoV-2 in induction of a competent IFN response.


Subject(s)
COVID-19 , Female , Humans , Male , Proteins/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , SARS-CoV-2 , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
20.
Cancer Sci ; 113(4): 1352-1361, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35133062

ABSTRACT

Radiotherapy (RT) combined with immune checkpoint inhibitors has recently produced outstanding results and is expected to be adaptable for various cancers. However, the precise molecular mechanism by which immune reactions are induced by fractionated RT is still controversial. We aimed to investigate the mechanism of the immune response regarding multifractionated, long-term radiation, which is most often combined with immunotherapy. Two human esophageal cancer cell lines, KYSE-450 and OE-21, were irradiated by fractionated irradiation (FIR) daily at a dose of 3 Gy in 5 d/wk for 2 weeks. Western blot analysis and RNA sequencing identified type I interferon (IFN) and the stimulator of IFN genes (STING) pathway as candidates that regulate immune response by FIR. We inhibited STING, IFNAR1, STAT1, and IFN regulatory factor 1 (IRF1) and investigated the effects on the immune response in cancer cells and the invasion of surrounding immune cells. We herein revealed type I IFN-dependent immune reactions and the positive feedback of STING, IRF1, and phosphorylated STAT1 induced by FIR. Knocking out STING, IFNAR1, STAT1, and IRF1 resulted in a poorer immunological response than that in WT cells. The STING-KO KYSE-450 cell line showed significantly less invasion of PBMCs than the WT cell line under FIR. In the analysis of STING-KO cells and migrated PBMCs, we confirmed the occurrence of STING-dependent immune activation under FIR. In conclusion, we identified that the STING-IFNAR1-STAT1-IRF1 axis regulates immune reactions in cancer cells triggered by FIR and that the STING pathway also contributes to immune cell invasion of cancer cells.


Subject(s)
Esophageal Neoplasms , Immunity , Interferon Regulatory Factor-1 , STAT1 Transcription Factor , Cell Line/radiation effects , Esophageal Neoplasms/genetics , Humans , Immunity/radiation effects , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/radiation effects , Interferon Type I , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/radiation effects , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/radiation effects , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...