Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 573
Filter
1.
Nat Commun ; 15(1): 7765, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237507

ABSTRACT

Human parainfluenza virus type 3 (HPIV-3) can cause severe respiratory tract infections. There are no convenient small-animal infection models. Here, we show viral replication in the upper and lower airways of AG129 mice (double IFNα/ß and IFNγ receptor knockout mice) upon intranasal inoculation. By multiplex fluorescence RNAscope and immunohistochemistry followed by confocal microscopy, we demonstrate viral tropism to ciliated cells and club cells of the bronchiolar epithelium. HPIV-3 causes a marked lung pathology. No virus transmission of the virus was observed by cohousing HPIV-3-infected AG129 mice with other mice. Oral treatment with GS-441524, the parent nucleoside of remdesivir, reduced infectious virus titers in the lung, with a relatively normal histology. Intranasal treatment also affords an antiviral effect. Thus, AG129 mice serve as a robust preclinical model for developing therapeutic and prophylactic strategies against HPIV-3. We suggest further investigation of GS-441524 and its prodrug forms to treat HPIV-3 infection in humans.


Subject(s)
Antiviral Agents , Disease Models, Animal , Lung , Mice, Knockout , Parainfluenza Virus 3, Human , Respirovirus Infections , Animals , Lung/virology , Lung/pathology , Lung/drug effects , Mice , Parainfluenza Virus 3, Human/drug effects , Parainfluenza Virus 3, Human/physiology , Antiviral Agents/pharmacology , Respirovirus Infections/drug therapy , Respirovirus Infections/virology , Humans , Virus Replication/drug effects , Female , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/deficiency , Adenosine/analogs & derivatives , Adenosine/pharmacology , Viral Tropism , Benzamides , Phthalimides
2.
Nat Commun ; 15(1): 7165, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187481

ABSTRACT

Programmed cell death 1 (PD-1) is a premier cancer drug target for immune checkpoint blockade (ICB). Because PD-1 receptor inhibition activates tumor-specific T-cell immunity, research has predominantly focused on T-cell-PD-1 expression and its immunobiology. In contrast, cancer cell-intrinsic PD-1 functional regulation is not well understood. Here, we demonstrate induction of PD-1 in melanoma cells via type I interferon receptor (IFNAR) signaling and reversal of ICB efficacy through IFNAR pathway inhibition. Treatment of melanoma cells with IFN-α or IFN-ß triggers IFNAR-mediated Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling, increases chromatin accessibility and resultant STAT1/2 and IFN regulatory factor 9 (IRF9) binding within a PD-1 gene enhancer, and leads to PD-1 induction. IFNAR1 or JAK/STAT inhibition suppresses melanoma-PD-1 expression and disrupts ICB efficacy in preclinical models. Our results uncover type I IFN-dependent regulation of cancer cell-PD-1 and provide mechanistic insight into the potential unintended ICB-neutralizing effects of widely used IFNAR1 and JAK inhibitors.


Subject(s)
Immune Checkpoint Inhibitors , Interferon Type I , Melanoma , Programmed Cell Death 1 Receptor , Receptor, Interferon alpha-beta , Signal Transduction , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Melanoma/drug therapy , Melanoma/immunology , Melanoma/genetics , Melanoma/metabolism , Humans , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Mice , Interferon Type I/metabolism , STAT1 Transcription Factor/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Interferon-beta/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Janus Kinases/metabolism , Mice, Inbred C57BL , Interferon-alpha/pharmacology , Interferon-alpha/metabolism , Female
3.
Proc Natl Acad Sci U S A ; 121(33): e2318190121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39106307

ABSTRACT

We developed a highly sensitive assay for detecting protein-protein interaction using chimeric receptors comprising two molecules of interest in the extracellular domain and interferon alpha and beta receptor subunit 1 or 2 (IFNAR1/2) in the intracellular domain. This intracellular IFNAR1/2 reconstitution system (IFNARRS) proved markedly more sensitive than the NanoBiT system, currently considered one of the best detection systems for protein interaction. Employing chimeric receptors with extracellular domains from the IFNγ or IL-2 receptor and the intracellular domains of IFNAR1/2, the IFNARRS system effectively identifies low IFNγ or IL-2 levels. Cells stably expressing these chimeric receptors responded to IFNγ secreted by activated T cells following various stimuli, including a specific peptide-antigen. The activation signals were further enhanced by the expression of relevant genes, such as costimulators, via IFN-stimulated response elements in the promoters. Besides IFNγ or IL-2, the IFNARRS system demonstrated the capability to detect other cytokines by using the corresponding extracellular domains from these target cytokine receptors.


Subject(s)
Interferon-gamma , Interleukin-2 , Receptor, Interferon alpha-beta , T-Lymphocytes , Humans , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Interleukin-2/metabolism , Interferon-gamma/metabolism , Receptors, Interleukin-2/metabolism , Receptors, Interleukin-2/genetics , Protein Binding , Lymphocyte Activation , HEK293 Cells
4.
Cells ; 13(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38995014

ABSTRACT

PD-1 (Programmed cell death protein 1) regulates the metabolic reprogramming of myeloid-derived suppressor cells and myeloid cell differentiation, as well as the type I interferon (IFN-I) signaling pathway in myeloid cells in the tumor microenvironment. PD-1, therefore, is a key inhibitory receptor in myeloid cells. However, the regulation of PD-1 expression in myeloid cells is unknown. We report that the expression level of PDCD1, the gene that encodes the PD-1 protein, is positively correlated with the levels of IFNB1 and IFNAR1 in myeloid cells in human colorectal cancer. Treatment of mouse myeloid cell lines with recombinant IFNß protein elevated PD-1 expression in myeloid cells in vitro. Knocking out IFNAR1, the gene that encodes the IFN-I-specific receptor, diminished the inductive effect of IFNß on PD-1 expression in myeloid cells in vitro. Treatment of tumor-bearing mice with a lipid nanoparticle-encapsulated IFNß-encoding plasmid (IFNBCOL01) increased IFNß expression, resulting in elevated PD-1 expression in tumor-infiltrating myeloid cells. At the molecular level, we determined that IFNß activates STAT1 (signal transducer and activator of transcription 1) and IRFs (interferon regulatory factors) in myeloid cells. Analysis of the cd279 promoter identified IRF2-binding consensus sequence elements. ChIP (chromatin immunoprecipitation) analysis determined that the pSTAT1 directly binds to the irf2 promoter and that IRF2 directly binds to the cd279 promoter in myeloid cells in vitro and in vivo. In colon cancer patients, the expression levels of STAT1, IRF2 and PDCD1 are positively correlated in tumor-infiltrating myeloid cells. Our findings determine that IFNß activates PD-1 expression at least in part by an autocrine mechanism via the stimulation of the pSTAT1-IRF2 axis in myeloid cells.


Subject(s)
Interferon Regulatory Factor-2 , Myeloid Cells , Programmed Cell Death 1 Receptor , STAT1 Transcription Factor , Signal Transduction , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Animals , Humans , STAT1 Transcription Factor/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Mice , Interferon Regulatory Factor-2/metabolism , Interferon Regulatory Factor-2/genetics , Signal Transduction/drug effects , Interferon Type I/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Interferon-beta/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Mice, Inbred C57BL
5.
J Neuroinflammation ; 21(1): 166, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956653

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) and obstructive sleep apnea (OSA) are mutual risk factors, with both conditions inducing cognitive impairment and anxiety. However, whether OSA exacerbates cognitive impairment and anxiety in patients with T2DM remains unclear. Moreover, TREM2 upregulation has been suggested to play a protective role in attenuating microglia activation and improving synaptic function in T2DM mice. The aim of this study was to explore the regulatory mechanisms of TREM2 and the cognitive and anxiety-like behavioral changes in mice with OSA combined with T2DM. METHODS: A T2DM with OSA model was developed by treating mice with a 60% kcal high-fat diet (HFD) combined with intermittent hypoxia (IH). Spatial learning memory capacity and anxiety in mice were investigated. Neuronal damage in the brain was determined by the quantity of synapses density, the number and morphology of brain microglia, and pro-inflammatory factors. For mechanism exploration, an in vitro model of T2DM combined with OSA was generated by co-treating microglia with high glucose (HG) and IH. Regulation of TREM2 on IFNAR1-STAT1 pathway was determined by RNA sequencing and qRT-PCR. RESULTS: Our results showed that HFD mice exhibited significant cognitive dysfunction and anxiety-like behavior, accompanied by significant synaptic loss. Furthermore, significant activation of brain microglia and enhanced microglial phagocytosis of synapses were observed. Moreover, IH was found to significantly aggravate anxiety in the HFD mice. The mechanism of HG treatment may potentially involve the promotion of TREM2 upregulation, which in turn attenuates the proinflammatory microglia by inhibiting the IFNAR1-STAT1 pathway. Conversely, a significant reduction in TREM2 in IH-co-treated HFD mice and HG-treated microglia resulted in the further activation of the IFNAR1-STAT1 pathway and consequently increased proinflammatory microglial activation. CONCLUSIONS: HFD upregulated the IFNAR1-STAT1 pathway and induced proinflammatory microglia, leading to synaptic damage and causing anxiety and cognitive deficits. The upregulated TREM2 inT2DM mice brain exerted a negative regulation of the IFNAR1-STAT1 pathway. Mice with T2DM combined with OSA exacerbated anxiety via the downregulation of TREM2, causing heightened IFNAR1-STAT1 pathway activation and consequently increasing proinflammatory microglia.


Subject(s)
Anxiety , Diabetes Mellitus, Type 2 , Diet, High-Fat , Hypoxia , Membrane Glycoproteins , Mice, Inbred C57BL , Receptor, Interferon alpha-beta , Receptors, Immunologic , Signal Transduction , Animals , Mice , Diet, High-Fat/adverse effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Anxiety/etiology , Anxiety/metabolism , Signal Transduction/physiology , Signal Transduction/drug effects , Hypoxia/metabolism , Hypoxia/complications , Male , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/psychology , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Microglia/metabolism , STAT1 Transcription Factor/metabolism , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/metabolism , Sleep Apnea, Obstructive/psychology
6.
Int J Mol Sci ; 25(14)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39063175

ABSTRACT

Type I interferon (IFN-I) signaling has been shown to be upregulated in systemic sclerosis (SSc). Dysregulated B-cell functions, including antigen presentation, as well as antibody and cytokine production, all of which may be affected by IFN-I signaling, play an important role in the pathogenesis of the disease. We investigated the IFN-I signature in 71 patients with the more severe form of the disease, diffuse cutaneous SSc (dcSSc), and 33 healthy controls (HCs). Activation via Toll-like receptors (TLRs) can influence the IFN-I signaling cascade; thus, we analyzed the effects of the TLR homologue CD180 ligation on the IFN-I signature in B cells. CD180 stimulation augmented the phosphorylation of signal transducer and activator of transcription 1 (STAT1) in dcSSc B cells (p = 0.0123). The expression of IFN-I receptor (IFNAR1) in non-switched memory B cells producing natural autoantibodies was elevated in dcSSc (p = 0.0109), which was enhanced following anti-CD180 antibody treatment (p = 0.0125). Autoantibodies to IFN-Is (IFN-alpha and omega) correlated (dcSSc p = 0.0003, HC p = 0.0192) and were present at similar levels in B cells from dcSSc and HC, suggesting their regulatory role as natural autoantibodies. It can be concluded that factors other than IFN-alpha may contribute to the elevated IFN-I signature of dcSSc B cells, and one possible candidate is B-cell activation via CD180.


Subject(s)
Antigens, CD , Autoantibodies , B-Lymphocytes , Interferon Type I , Humans , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Middle Aged , Female , Male , Autoantibodies/immunology , Antigens, CD/metabolism , Adult , Interferon Type I/metabolism , STAT1 Transcription Factor/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Scleroderma, Diffuse/immunology , Scleroderma, Diffuse/metabolism , Aged , Up-Regulation , Signal Transduction
7.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-39042188

ABSTRACT

The contribution of placental immune responses to congenital Zika virus (ZIKV) syndrome remains poorly understood. Here, we leveraged a mouse model of ZIKV infection to identify mechanisms of innate immune restriction exclusively in the fetal compartment of the placenta. ZIKV principally infected mononuclear trophoblasts in the junctional zone, which was limited by mitochondrial antiviral-signaling protein (MAVS) and type I interferon (IFN) signaling mechanisms. Single nuclear RNA sequencing revealed MAVS-dependent expression of IFN-stimulated genes (ISGs) in spongiotrophoblasts but not in other placental cells that use alternate pathways to induce ISGs. ZIKV infection of Ifnar1-/- or Mavs-/- placentas was associated with greater infection of the adjacent immunocompetent decidua, and heterozygous Mavs+/- or Ifnar1+/- dams carrying immunodeficient fetuses sustained greater maternal viremia and tissue infection than dams carrying wild-type fetuses. Thus, MAVS-IFN signaling in the fetus restricts ZIKV infection in junctional zone trophoblasts, which modulates dissemination and outcome for both the fetus and the pregnant mother.


Subject(s)
Adaptor Proteins, Signal Transducing , Decidua , Fetus , Interferon Type I , Placenta , Receptor, Interferon alpha-beta , Signal Transduction , Trophoblasts , Zika Virus Infection , Zika Virus , Female , Animals , Pregnancy , Interferon Type I/metabolism , Interferon Type I/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Placenta/immunology , Placenta/virology , Placenta/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Zika Virus/physiology , Mice , Decidua/immunology , Decidua/virology , Decidua/metabolism , Fetus/immunology , Fetus/virology , Trophoblasts/immunology , Trophoblasts/virology , Trophoblasts/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Immunity, Innate , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/virology , Disease Models, Animal
8.
Int J Biol Sci ; 20(8): 3076-3093, 2024.
Article in English | MEDLINE | ID: mdl-38904031

ABSTRACT

Epizootic hemorrhagic disease (EHD), caused by Epizootic hemorrhagic disease virus (EHDV), is an emerging and severe livestock disease. Recent incursion and distribution of EHDV in Europe have outlined the emerging character of EHD. Despite its worldwide impact, numerous knowledge gaps exist. A range of inconveniences restricts utilization of natural hosts of EHDV. Here, we show that adult mice deficient in type I IFN receptor (IFNAR(-/-)) are highly susceptible to EHDV-6 and EHDV-8 infection when the virus is administered subcutaneously. Disease was characterized by ruffled hair, reluctance to move, dehydration and conjunctivitis, with viraemia detected from day 5 post-infection. A deeper characterization of EHDV-8 infection showed viral replication in the lung, liver, spleen, kidney, testis and ovaries. Importantly, increased expression levels of pro-inflammatory cytokines IL-1ß, IL-6 and CXCL2 were observed in spleen after EHDV-8 infection. Furthermore, IFNAR(-/-) adult mice immunized with a EHDV-8 inactivated vaccine elicited neutralizing antibodies specific of EHDV-8 and full protection against challenge with a lethal dose of this virus. This study also explores the possibilities of this animal model for study of BTV and EHDV coinfection. In summary, the IFNAR(-/-) mouse model faithfully recapitulates EHD and can be applied for vaccine testing, which can facilitate progress in addressing the animal health challenge posed by this virus.


Subject(s)
Disease Models, Animal , Hemorrhagic Disease Virus, Epizootic , Receptor, Interferon alpha-beta , Viral Vaccines , Animals , Mice , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Hemorrhagic Disease Virus, Epizootic/immunology , Hemorrhagic Disease Virus, Epizootic/genetics , Viral Vaccines/immunology , Reoviridae Infections/immunology , Female , Mice, Knockout , Antibodies, Neutralizing/immunology , Male
9.
J Bone Miner Res ; 39(8): 1132-1146, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-38874138

ABSTRACT

Type I interferons (IFN-I) are pleiotropic factors endowed with multiple activities that play important roles in innate and adaptive immunity. Although many studies indicate that IFN-I inducers exert favorable effects on broad-spectrum antivirus, immunomodulation, and anti-tumor activities by inducing endogenous IFN-I and IFN-stimulated genes, their function in bone homeostasis still needs further exploration. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. First, IFN-I inducers suppress the genes that control osteoclast (OC) differentiation and activity in vitro. Moreover, diABZI alleviates bone loss in Ti particle-induced osteolysis and ovariectomized -induced osteoporosis in vivo by inhibiting OC differentiation and function. In addition, the inhibitory effects of IFN-I inducers on OC differentiation are not observed in macrophages derived from Ifnar1-/-mice, which indicate that the suppressive effect of IFN-I inducers on OC is IFNAR-dependent. Mechanistically, RNAi-mediated silencing of IRF7 and IFIT3 in OC precursors impairs the suppressive effect of the IFN-I inducers on OC differentiation. Taken together, these results demonstrate that IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway.


OCs are responsible for bone resorption, and their excessive differentiation and enhanced activity will lead to bone resorption diseases such as osteoporosis and osteolysis. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. IFN-I inducers suppress OC differentiation, and particularly diABZI alleviates bone loss in osteolysis and osteoporosis mouse models. Taken together, IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway. Our in-depth and comprehensive discovery of the IFN-I inducer would provide new insight into OC biology and therapeutic targets for osteoclastic bone resorption diseases.


Subject(s)
Bone Resorption , Cell Differentiation , Interferon Regulatory Factor-7 , Osteoclasts , Poly I-C , Animals , Osteoclasts/metabolism , Osteoclasts/drug effects , Osteoclasts/pathology , Interferon Regulatory Factor-7/metabolism , Bone Resorption/pathology , Mice , Poly I-C/pharmacology , Cell Differentiation/drug effects , Female , Mice, Inbred C57BL , Mice, Knockout , Interferon Type I/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Humans , Osteolysis/pathology , Osteolysis/metabolism , Osteolysis/drug therapy
10.
Cell Mol Immunol ; 21(8): 892-904, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906982

ABSTRACT

Type I interferon (IFN-I) exhibits broad-spectrum antiviral properties and is commonly employed in clinical for the treatment of viral infections. In this study, we unveil SENP6 as a potent regulator of IFN-I antiviral activity. SENP6 does not impact the production of IFN-I induced by viruses but rather modulates IFN-I-activated signaling. Mechanistically, SENP6 constitutively interacts with USP8 and inhibits the SUMOylation of USP8, consequently restricting the interaction between USP8 and IFNAR2. The dissociation of USP8 from IFNAR2 enhances IFNAR2 ubiquitination and degradation, thus attenuating IFN-I antiviral activity. Correspondingly, the downregulation of SENP6 promotes the interaction between USP8 and IFNAR2, leading to a reduction in IFNAR2 ubiquitination and, consequently, an enhancement in IFN-I-induced signaling. This study deciphers a critical deSUMOylation-deubiquitination crosstalk that finely regulates the IFN-I response to viral infection.


Subject(s)
Endopeptidases , Interferon Type I , Receptor, Interferon alpha-beta , Signal Transduction , Sumoylation , Ubiquitin Thiolesterase , Ubiquitination , Humans , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Interferon Type I/metabolism , Ubiquitin Thiolesterase/metabolism , HEK293 Cells , Endopeptidases/metabolism , Antiviral Agents/pharmacology , Animals , Protein Binding , Virus Diseases/immunology , Endosomal Sorting Complexes Required for Transport
11.
Brain Res ; 1840: 149082, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38866307

ABSTRACT

Ubiquitin specific protease 18 (USP18) serves as a potent inhibitor of Type I interferon (IFN) signaling. Previous studies have shown that Usp18 deficient (homozygous Usp18 gene knockout) mice exhibit hydrocephalus; however, the precise molecular mechanism underlying hydrocephalus development remains elusive. In this study, we demonstrate that mice lacking both type I IFN receptor subunit 1 (Ifnar1) and Usp18 (Ifnar1/Usp18 double knockout mice) are viable and do not display a hydrocephalus phenotype. Moreover, we observed that suppression of USP18 in ependymal cells treated with IFN significantly increased cell death, including pyroptosis, and decreased proliferation. These findings suggest that heightened sensitivity to type I IFN during brain development contributes to the onset of hydrocephalus. Furthermore, they imply that inhibition of IFN signaling may hold promise as a therapeutic strategy for hydrocephalus.


Subject(s)
Hydrocephalus , Interferon Type I , Mice, Knockout , Receptor, Interferon alpha-beta , Ubiquitin Thiolesterase , Animals , Hydrocephalus/genetics , Hydrocephalus/pathology , Interferon Type I/metabolism , Mice , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Mice, Inbred C57BL , Signal Transduction/drug effects , Brain/metabolism , Brain/pathology , Brain/drug effects , Ependyma/metabolism , Cell Proliferation/drug effects , Pyroptosis/drug effects , Pyroptosis/physiology
12.
Immunity ; 57(7): 1696-1709.e10, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38878770

ABSTRACT

Aicardi-Goutières syndrome (AGS) is an autoinflammatory disease characterized by aberrant interferon (IFN)-α production. The major cause of morbidity in AGS is brain disease, yet the primary source and target of neurotoxic IFN-α remain unclear. Here, we demonstrated that the brain was the primary source of neurotoxic IFN-α in AGS and confirmed the neurotoxicity of intracerebral IFN-α using astrocyte-driven Ifna1 misexpression in mice. Using single-cell RNA sequencing, we demonstrated that intracerebral IFN-α-activated receptor (IFNAR) signaling within cerebral endothelial cells caused a distinctive cerebral small vessel disease similar to that observed in individuals with AGS. Magnetic resonance imaging (MRI) and single-molecule ELISA revealed that central and not peripheral IFN-α was the primary determinant of microvascular disease in humans. Ablation of endothelial Ifnar1 in mice rescued microvascular disease, stopped the development of diffuse brain disease, and prolonged lifespan. These results identify the cerebral microvasculature as a primary mediator of IFN-α neurotoxicity in AGS, representing an accessible target for therapeutic intervention.


Subject(s)
Brain , Interferon-alpha , Microvessels , Nervous System Malformations , Receptor, Interferon alpha-beta , Animals , Humans , Mice , Interferon-alpha/metabolism , Brain/metabolism , Brain/pathology , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Microvessels/pathology , Nervous System Malformations/genetics , Autoimmune Diseases of the Nervous System/immunology , Endothelial Cells/metabolism , Mice, Knockout , Male , Female , Signal Transduction , Mice, Inbred C57BL , Astrocytes/metabolism , Disease Models, Animal
13.
Mol Neurodegener ; 19(1): 48, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38886816

ABSTRACT

BACKGROUND: Aging significantly elevates the risk of developing neurodegenerative diseases. Neuroinflammation is a universal hallmark of neurodegeneration as well as normal brain aging. Which branches of age-related neuroinflammation, and how they precondition the brain toward pathological progression, remain ill-understood. The presence of elevated type I interferon (IFN-I) has been documented in the aged brain, but its role in promoting degenerative processes, such as the loss of neurons in vulnerable regions, has not been studied in depth. METHODS: To comprehend the scope of IFN-I activity in the aging brain, we surveyed IFN-I-responsive reporter mice at multiple ages. We also examined 5- and 24-month-old mice harboring selective ablation of Ifnar1 in microglia to observe the effects of manipulating this pathway during the aging process using bulk RNA sequencing and histological parameters. RESULTS: We detected age-dependent IFN-I signal escalation in multiple brain cell types from various regions, especially in microglia. Selective ablation of Ifnar1 from microglia in aged mice significantly reduced overall brain IFN-I signature, dampened microglial reactivity, lessened neuronal loss, restored expression of key neuronal genes and pathways, and diminished the accumulation of lipofuscin, a core hallmark of cellular aging in the brain. CONCLUSIONS: Overall, our study demonstrates pervasive IFN-I activity during normal mouse brain aging and reveals a pathogenic, pro-degenerative role played by microglial IFN-I signaling in perpetuating neuroinflammation, neuronal dysfunction, and molecular aggregation. These findings extend the understanding of a principal axis of age-related inflammation in the brain, one likely shared with multiple neurological disorders, and provide a rationale to modulate aberrant immune activation to mitigate neurodegenerative process at all stages.


Subject(s)
Aging , Brain , Interferon Type I , Microglia , Signal Transduction , Animals , Aging/metabolism , Interferon Type I/metabolism , Mice , Brain/metabolism , Brain/pathology , Signal Transduction/physiology , Microglia/metabolism , Receptor, Interferon alpha-beta/metabolism , Neurons/metabolism
14.
Nat Commun ; 15(1): 4484, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802340

ABSTRACT

Deciphering the intricate dynamic events governing type I interferon (IFN) signaling is critical to unravel key regulatory mechanisms in host antiviral defense. Here, we leverage TurboID-based proximity labeling coupled with affinity purification-mass spectrometry to comprehensively map the proximal human proteomes of all seven canonical type I IFN signaling cascade members under basal and IFN-stimulated conditions. This uncovers a network of 103 high-confidence proteins in close proximity to the core members IFNAR1, IFNAR2, JAK1, TYK2, STAT1, STAT2, and IRF9, and validates several known constitutive protein assemblies, while also revealing novel stimulus-dependent and -independent associations between key signaling molecules. Functional screening further identifies PJA2 as a negative regulator of IFN signaling via its E3 ubiquitin ligase activity. Mechanistically, PJA2 interacts with TYK2 and JAK1, promotes their non-degradative ubiquitination, and limits the activating phosphorylation of TYK2 thereby restraining downstream STAT signaling. Our high-resolution proximal protein landscapes provide global insights into the type I IFN signaling network, and serve as a valuable resource for future exploration of its functional complexities.


Subject(s)
Interferon Type I , Janus Kinase 1 , Receptor, Interferon alpha-beta , STAT2 Transcription Factor , Signal Transduction , TYK2 Kinase , Ubiquitination , Humans , HEK293 Cells , Interferon Type I/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Janus Kinase 1/metabolism , Phosphorylation , Proteome/metabolism , Receptor, Interferon alpha-beta/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , TYK2 Kinase/metabolism , Ubiquitin-Protein Ligases/metabolism
15.
Cell Death Dis ; 15(5): 369, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806478

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) is frequently overexpressed in patients with acute myeloid leukemia (AML). STAT3 exists in two distinct alternatively spliced isoforms, the full-length isoform STAT3α and the C-terminally truncated isoform STAT3ß. While STAT3α is predominantly described as an oncogenic driver, STAT3ß has been suggested to act as a tumor suppressor. To elucidate the role of STAT3ß in AML, we established a mouse model of STAT3ß-deficient, MLL-AF9-driven AML. STAT3ß deficiency significantly shortened survival of leukemic mice confirming its role as a tumor suppressor. Furthermore, RNA sequencing revealed enhanced STAT1 expression and interferon (IFN) signaling upon loss of STAT3ß. Accordingly, STAT3ß-deficient leukemia cells displayed enhanced sensitivity to blockade of IFN signaling through both an IFNAR1 blocking antibody and the JAK1/2 inhibitor Ruxolitinib. Analysis of human AML patient samples confirmed that elevated expression of IFN-inducible genes correlated with poor overall survival and low STAT3ß expression. Together, our data corroborate the tumor suppressive role of STAT3ß in a mouse model in vivo. Moreover, they provide evidence that its tumor suppressive function is linked to repression of the STAT1-mediated IFN response. These findings suggest that the STAT3ß/α mRNA ratio is a significant prognostic marker in AML and holds crucial information for targeted treatment approaches. Patients displaying a low STAT3ß/α mRNA ratio and unfavorable prognosis could benefit from therapeutic interventions directed at STAT1/IFN signaling.


Subject(s)
Leukemia, Myeloid, Acute , STAT3 Transcription Factor , Animals , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Humans , STAT3 Transcription Factor/metabolism , Mice , Signal Transduction , Interferons/metabolism , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Cell Line, Tumor , Nitriles , Pyrazoles , Pyrimidines
16.
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 , TYK2 Kinase , Adult , Aged , Female , Humans , Male , Middle Aged , COVID-19/genetics , Prognosis , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Retrospective Studies , SARS-CoV-2 , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
17.
Cell Rep Med ; 5(5): 101560, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729159

ABSTRACT

Stimulator of IFN genes (STING) is a promising target for adjuvants utilized in in situ cancer vaccination approaches. However, key barriers remain for clinical translation, including low cellular uptake and accessibility, STING variability necessitating personalized STING agonists, and interferon (IFN)-independent signals that can promote tumor growth. Here, we identify C100, a highly deacetylated chitin-derived polymer (HDCP), as an attractive alternative to conventional STING agonists. C100 promotes potent anti-tumor immune responses, outperforming less deacetylated HDCPs, with therapeutic efficacy dependent on STING and IFN alpha/beta receptor (IFNAR) signaling and CD8+ T cell mediators. Additionally, C100 injection synergizes with systemic checkpoint blockade targeting PD-1. Mechanistically, C100 triggers mitochondrial stress and DNA damage to exclusively activate the IFN arm of the cGAS-STING signaling pathway and elicit sustained IFNAR signaling. Altogether, these results reveal an effective STING- and IFNAR-dependent adjuvant for in situ cancer vaccines with a defined mechanism and distinct properties that overcome common limitations of existing STING therapeutics.


Subject(s)
Adjuvants, Immunologic , CD8-Positive T-Lymphocytes , Chitin , Membrane Proteins , Mice, Inbred C57BL , Receptor, Interferon alpha-beta , Signal Transduction , Animals , Membrane Proteins/metabolism , Membrane Proteins/immunology , Membrane Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Signal Transduction/drug effects , Humans , Cancer Vaccines/immunology , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Female , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/immunology , Neoplasms/immunology , Neoplasms/therapy
18.
Biochimie ; 223: 196-205, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38642825

ABSTRACT

Astrocytes are glial cells that play key roles in neuroinflammation, which is a common feature in diabetic encephalopathy and aging process. Metformin is an antidiabetic compound that shows neuroprotective properties, including in inflammatory models, but astroglial signaling pathways involved are still poorly known. Interferons α/ß are cytokines that participate in antiviral responses and the lack of their signaling increases susceptible to viral infections. Here, we investigated the effects of metformin on astrocytes from hypothalamus, a crucial brain region related to inflammatory processes. Astrocyte cultures were derived from interferon α/ß receptor knockout (IFNα/ßR-/-) and wild-type (WT) mice. Metformin did not change the expression of glial fibrillary acidic protein but caused an anti-inflammatory effect by decreasing pro-inflammatory cytokines (tumor necrosis factor-α and interleukin-1ß), as well as increasing gene expression of anti-inflammatory proteins interleukin-10 and Nrf2 (nuclear factor erythroid derived 2 like 2). However, nuclear factor κB p65 and cyclooxygenase 2 were downregulated in WT astrocytes and upregulated in IFNα/ßR-/- astrocytes. AMP-activated protein kinase (AMPK), a molecular target of metformin, was upregulated only in WT astrocytes, while sirtuin 1 increased in both mice models. The expression of inducible nitric oxide synthase was decreased in WT astrocytes and heme oxygenase 1 was increased in IFNα/ßR-/- astrocytes. Although loss of IFNα/ßR-mediated signaling affects some effects of metformin, our results support beneficial roles of this drug in hypothalamic astrocytes. Moreover, paradoxical response of metformin may involve AMPK. Thus, metformin can mediate glioprotection due its effects on age-related disorders in non-diabetic and diabetic encephalopathy individuals.


Subject(s)
Astrocytes , Hypothalamus , Metformin , Mice, Knockout , Animals , Metformin/pharmacology , Astrocytes/metabolism , Astrocytes/drug effects , Mice , Hypothalamus/metabolism , Hypothalamus/drug effects , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/genetics , Immunocompromised Host , AMP-Activated Protein Kinases/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Cells, Cultured , Sirtuin 1/metabolism , Sirtuin 1/genetics
19.
Int J Biol Macromol ; 268(Pt 2): 131721, 2024 May.
Article in English | MEDLINE | ID: mdl-38649079

ABSTRACT

Interferon (IFN) alpha/beta receptor 1 (IFNAR1) is indispensable for antiviral responses and the immune regulation. Dysregulation of the IFNAR1-mediaetd signaling pathways leads to deleterious autoimmune diseases such as systemic lupus erythematosus (SLE). QX006N, a humanized therapeutic monoclonal antibody, specifically targets human IFNAR1 and is in the clinical trial phase for treating SLE, but the molecular mechanism underlying the QX006N-mediated recognition of IFNAR1 remains unclear. Here, we report the high neutralization activities of QX006N against IFNAR1-mediated signal transduction. Meanwhile, we determine the structures of the fragment antigen-binding domain (Fab) of QX006N (QX006N-Fab) and QX006N-Fab in complex with the subdomains 1-3 of IFNAR1 (IFNAR1-SD123) at 2.87 Å and 2.68 Å resolutions, respectively. In the structure of the QX006N-Fab/IFNAR1-SD123 complex, QX006N-Fab only recognizes the SD3 subdomain of IFNAR1 by the hydrophobic, hydrogen-bonding and electrostatic interactions. Compared with the structure of the IFN/IFNAR1/IFNAR2 complex, the binding of QX006N-Fab to IFNAR1-SD3 blocks its association with IFN due to steric hindrance, which inhibits the IFN/IFNAR1/IFNAR2 complex formation for signal transduction. The results of this study provide the structural evidence for the specific targeting of IFNAR1 by the therapeutic antibody QX006N and pave the way for the rational design of antibody drugs to combat IFNAR1-related autoimmune diseases.


Subject(s)
Antibodies, Monoclonal, Humanized , Lupus Erythematosus, Systemic , Receptor, Interferon alpha-beta , Receptor, Interferon alpha-beta/metabolism , Receptor, Interferon alpha-beta/chemistry , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Humans , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Protein Binding , Models, Molecular , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Signal Transduction/drug effects , Structure-Activity Relationship
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL