Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.502
Filter
1.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35110413

ABSTRACT

Changes in population density lead to phenotypic differentiation of solitary and gregarious locusts, which display different resistance to fungal pathogens; however, how to regulate their cellular immune strategies remains unknown. Here, our stochastic simulation of pathogen proliferation suggested that humoral defense always enhanced resistance to fungal pathogens, while phagocytosis sometimes reduced defense against pathogens. Further experimental data proved that gregarious locusts had significantly decreased phagocytosis of hemocytes compared to solitary locusts. Additionally, transcriptional analysis showed that gregarious locusts promoted immune effector expression (gnbp1 and dfp) and reduced phagocytic gene expression (eater) and the cytokine tumor necrosis factor (TNF). Interestingly, higher expression of the cytokine TNF in solitary locusts simultaneously promoted eater expression and inhibited gnbp1 and dfp expression. Moreover, inhibition of TNF increased the survival of solitary locusts, and injection of TNF decreased the survival of gregarious locusts after fungal infection. Therefore, our results indicate that the alerted expression of TNF regulated the immune strategy of locusts to adapt to environmental changes.


Subject(s)
Grasshoppers/immunology , Grasshoppers/microbiology , Immunity, Cellular/immunology , Metarhizium/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Gene Expression/immunology , Phagocytosis/immunology , Population Density , Transcription, Genetic/immunology
2.
FASEB J ; 36(2): e22007, 2022 02.
Article in English | MEDLINE | ID: mdl-35051300

ABSTRACT

The aim of this study was to further elucidate the molecular mechanisms that mediate pathologic foreign body response (FBR) to biomedical implants. The longevity of biomedical implants is limited by the FBR, which leads to implant failure and patient morbidity. Since the specific molecular mechanisms underlying fibrotic responses to biomedical implants have yet to be fully described, there are currently no targeted approaches to reduce pathologic FBR. We utilized proteomics analysis of human FBR samples to identify potential molecular targets for therapeutic inhibition of FBR. We then employed a murine model of FBR to further evaluate the role of this potential target. We performed histological and immunohistochemical analysis on the murine FBR capsule tissue, as well as single-cell RNA sequencing (scRNA-seq) on cells isolated from the capsules. We identified IQ motif containing GTPase activating protein 1 (IQGAP1) as the most promising of several targets, serving as a central molecular mediator in human and murine FBR compared to control subcutaneous tissue. IQGAP1-deficient mice displayed a significantly reduced FBR compared to wild-type mice as evidenced by lower levels of collagen deposition and maturity. Our scRNA-seq analysis revealed that decreasing IQGAP1 resulted in diminished transcription of mechanotransduction, inflammation, and fibrosis-related genes, which was confirmed on the protein level with immunofluorescent staining. The deficiency of IQGAP1 significantly attenuates FBR by deactivating downstream mechanotransduction signaling, inflammation, and fibrotic pathways. IQGAP1 may be a promising target for rational therapeutic design to mitigate pathologic FBR around biomedical implants.


Subject(s)
Biocompatible Materials/adverse effects , Foreign Bodies/immunology , Prostheses and Implants/adverse effects , Signal Transduction/immunology , ras GTPase-Activating Proteins/immunology , Animals , Collagen/immunology , Fibrosis/immunology , Humans , Inflammation/immunology , Male , Mechanotransduction, Cellular/immunology , Mice , Mice, Inbred C57BL , Transcription, Genetic/immunology
3.
Nature ; 601(7891): 110-117, 2022 01.
Article in English | MEDLINE | ID: mdl-34758478

ABSTRACT

Individuals with potential exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) do not necessarily develop PCR or antibody positivity, suggesting that some individuals may clear subclinical infection before seroconversion. T cells can contribute to the rapid clearance of SARS-CoV-2 and other coronavirus infections1-3. Here we hypothesize that pre-existing memory T cell responses, with cross-protective potential against SARS-CoV-2 (refs. 4-11), would expand in vivo to support rapid viral control, aborting infection. We measured SARS-CoV-2-reactive T cells, including those against the early transcribed replication-transcription complex (RTC)12,13, in intensively monitored healthcare workers (HCWs) who tested repeatedly negative according to PCR, antibody binding and neutralization assays (seronegative HCWs (SN-HCWs)). SN-HCWs had stronger, more multispecific memory T cells compared with a cohort of unexposed individuals from before the pandemic (prepandemic cohort), and these cells were more frequently directed against the RTC than the structural-protein-dominated responses observed after detectable infection (matched concurrent cohort). SN-HCWs with the strongest RTC-specific T cells had an increase in IFI27, a robust early innate signature of SARS-CoV-2 (ref. 14), suggesting abortive infection. RNA polymerase within RTC was the largest region of high sequence conservation across human seasonal coronaviruses (HCoV) and SARS-CoV-2 clades. RNA polymerase was preferentially targeted (among the regions tested) by T cells from prepandemic cohorts and SN-HCWs. RTC-epitope-specific T cells that cross-recognized HCoV variants were identified in SN-HCWs. Enriched pre-existing RNA-polymerase-specific T cells expanded in vivo to preferentially accumulate in the memory response after putative abortive compared to overt SARS-CoV-2 infection. Our data highlight RTC-specific T cells as targets for vaccines against endemic and emerging Coronaviridae.


Subject(s)
Asymptomatic Infections , COVID-19/immunology , COVID-19/virology , DNA-Directed RNA Polymerases/immunology , Memory T Cells/immunology , SARS-CoV-2/immunology , Seroconversion , Cell Proliferation , Cohort Studies , DNA-Directed RNA Polymerases/metabolism , Evolution, Molecular , Female , Health Personnel , Humans , Male , Membrane Proteins/immunology , Memory T Cells/cytology , Multienzyme Complexes/immunology , SARS-CoV-2/enzymology , SARS-CoV-2/growth & development , Transcription, Genetic/immunology
4.
J Allergy Clin Immunol ; 149(3): 923-933.e6, 2022 03.
Article in English | MEDLINE | ID: mdl-34902435

ABSTRACT

BACKGROUND: Treatments for coronavirus disease 2019, which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), are urgently needed but remain limited. SARS-CoV-2 infects cells through interactions of its spike (S) protein with angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) on host cells. Multiple cells and organs are targeted, particularly airway epithelial cells. OM-85, a standardized lysate of human airway bacteria with strong immunomodulating properties and an impeccable safety profile, is widely used to prevent recurrent respiratory infections. We found that airway OM-85 administration inhibits Ace2 and Tmprss2 transcription in the mouse lung, suggesting that OM-85 might hinder SARS-CoV-2/host cell interactions. OBJECTIVES: We sought to investigate whether and how OM-85 treatment protects nonhuman primate and human epithelial cells against SARS-CoV-2. METHODS: ACE2 and TMPRSS2 mRNA and protein expression, cell binding of SARS-CoV-2 S1 protein, cell entry of SARS-CoV-2 S protein-pseudotyped lentiviral particles, and SARS-CoV-2 cell infection were measured in kidney, lung, and intestinal epithelial cell lines, primary human bronchial epithelial cells, and ACE2-transfected HEK293T cells treated with OM-85 in vitro. RESULTS: OM-85 significantly downregulated ACE2 and TMPRSS2 transcription and surface ACE2 protein expression in epithelial cell lines and primary bronchial epithelial cells. OM-85 also strongly inhibited SARS-CoV-2 S1 protein binding to, SARS-CoV-2 S protein-pseudotyped lentivirus entry into, and SARS-CoV-2 infection of epithelial cells. These effects of OM-85 appeared to depend on SARS-CoV-2 receptor downregulation. CONCLUSIONS: OM-85 inhibits SARS-CoV-2 epithelial cell infection in vitro by downregulating SARS-CoV-2 receptor expression. Further studies are warranted to assess whether OM-85 may prevent and/or reduce the severity of coronavirus disease 2019.


Subject(s)
Adjuvants, Immunologic/administration & dosage , COVID-19/prevention & control , Cell Extracts/administration & dosage , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/immunology , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/antagonists & inhibitors , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , COVID-19/immunology , COVID-19/virology , Caco-2 Cells , Cell Extracts/immunology , Cells, Cultured , Chlorocebus aethiops , Down-Regulation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , HEK293 Cells , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , In Vitro Techniques , Lung/drug effects , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Serine Endopeptidases/drug effects , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Vero Cells
5.
Mol Immunol ; 141: 79-86, 2022 01.
Article in English | MEDLINE | ID: mdl-34837777

ABSTRACT

Our previous study had shown that member 13 (Hspa13) of heat shock protein family A (Hsp70) promotes plasma cell (PC) production and antibody secretion. To further explore Hspa13 expression and function, we combined single-cell RNA-sequencing and antigen receptor lineage (BCR) analysis to characterize sheep red cell‒primed splenocytes. The single-cell transcriptional profiles revealed that Hspa13 is specifically and highly expressed in PCs. These results suggest that Hspa13 is a novel PC-specific marker. In terms of its function, we found that the CD19cre-mediated conditional knock-out (cKO) of Hspa13 reduced the expression of Ebi3 and IL-10 in PCs. Ebi3 and IL-10 are important factors in IL-4‒secreting type 2 helper T cell (Th2) activation and differentiation. As expected, we found that the Hspa13 cKO reduced IL‒4-expressing follicular helper T (Tfh2) cells. Finally, the single-cell antigen receptor analysis demonstrated that the Hspa13 cKO reduced the Aicda-mediated antibody class-switching recombination (CSR) and somatic hypermutation (SHM) in germinal centers (GCs) B cells. Altogether, the single-cell atlas of splenocytes revealed a critical indirect role for the novel PC-specific marker Hspa13 in CSR and SHM in GC B cells by promoting Ebi3 and IL-10 expression in PCs to induce IL-4-expressing Tfh2 cells. Further exploration of Hspa13 expression and function will provide valuable clues for how to use Hspa13 in the treatment of autoimmune diseases.


Subject(s)
Antibodies/immunology , Germinal Center/immunology , HSP70 Heat-Shock Proteins/immunology , Recombination, Genetic/immunology , Somatic Hypermutation, Immunoglobulin/immunology , Animals , Antigens, CD19/immunology , Biomarkers/blood , Cell Differentiation/immunology , Gene Rearrangement/immunology , Mice , Mice, Knockout , Sheep , Th2 Cells/immunology , Transcription, Genetic/immunology
6.
Immunol Lett ; 240: 123-136, 2021 12.
Article in English | MEDLINE | ID: mdl-34715236

ABSTRACT

Intracellular adhesion molecule 1 (ICAM-1) is one of the most extensively studied inducible cell adhesion molecules which is responsible for several immune functions like T cell activation, extravasation, inflammation, etc. The molecule is constitutively expressed over the cell surface and is regulated up / down in response to inflammatory mediators like cellular stress, proinflammatory cytokines, viral infection. These stimuli modulate the expression of ICAM-1 primarily through regulating the ICAM-1 gene transcription. On account of the presence of various binding sites for NF-κB, AP-1, SP-1, and many other transcription factors, the architecture of the ICAM-1 promoter become complex. Transcription factors in union with other transcription factors, coactivators, and suppressors promote their assembly in a stereospecific manner on ICAM-1 promoter which mediates ICAM-1 regulation in response to different stimuli. Along with transcriptional regulation, epigenetic modifications also play a pivotal role in controlling ICAM-1 expression on different cell types. In this review, we summarize the regulation of ICAM-1 expression both at the transcriptional as well as post-transcriptional level with an emphasis on transcription factors and signaling pathways involved.


Subject(s)
Gene Expression Regulation/immunology , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Activation , Signal Transduction/immunology , T-Lymphocytes/immunology , Transcription, Genetic/immunology , Humans , Response Elements/immunology , Transcription Factors/immunology
7.
J Biol Chem ; 297(4): 101209, 2021 10.
Article in English | MEDLINE | ID: mdl-34562454

ABSTRACT

DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is known primarily for its function in DNA double-stranded break repair and nonhomologous end joining (NHEJ). However, DNA-PKcs also has a critical yet undefined role in immunity impacting both myeloid and lymphoid cell lineages spurring interest in targeting DNA-PKcs for therapeutic strategies in immune-related diseases. To gain insight into the function of DNA-PKcs within immune cells, we performed a quantitative phosphoproteomic screen in T cells to identify phosphorylation targets of DNA-PKcs. Our results indicate that DNA-PKcs phosphorylates the transcription factor Egr1 (early growth response protein 1) at serine 301. Expression of Egr1 is induced early upon T cell activation and dictates T cell response by modulating expression of cytokines and key costimulatory molecules such as IL (interleukin) 2, IL6, IFNγ, and NFκB. Inhibition of DNA-PKcs by treatment with a DNA-PKcs specific inhibitor NU7441 or shRNA knockdown increased proteasomal degradation of Egr1. Mutation of serine 301 to alanine via CRISPR-Cas9 reduced EGR1 protein expression and decreased Egr1-dependent transcription of IL2 in activated T cells. Our findings identify DNA-PKcs as a critical intermediary link between T cell activation and T cell fate and a novel phosphosite involved in regulating Egr1 activity.


Subject(s)
DNA-Activated Protein Kinase/immunology , DNA-Binding Proteins/immunology , Early Growth Response Protein 1/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Cytokines/genetics , Cytokines/immunology , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/genetics , Early Growth Response Protein 1/genetics , Humans , Jurkat Cells , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Mutation, Missense , Protein Stability , Transcription, Genetic/immunology
8.
Cell Prolif ; 54(10): e13116, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34469018

ABSTRACT

OBJECTIVES: The discrepancy between supply and demand of organ has led to an increased utilization of steatotic liver for liver transplantation (LT). Hepatic steatosis, however, is a major risk factor for graft failure due to increased susceptibility to ischaemia-reperfusion (I/R) injury during transplantation. MATERIALS AND METHODS: To assess the plasticity and phenotype of immune cells within the microenvironment of steatotic liver graft at single-cell level, single-cell RNA-sequencing (scRNA-Seq) was carried out on 23 675 cells from transplanted rat livers. Bioinformatic analyses and multiplex immunohistochemistry were performed to assess the functional properties, transcriptional regulation, phenotypic switching and cell-cell interactions of different cell subtypes. RESULTS: We have identified 11 different cell types in transplanted livers and found that the highly complex ecosystem was shaped by myeloid-derived cell subsets that transit between different states and interact mutually. Notably, a pro-inflammatory phenotype of Kupffer cells (KCs) with high expression of colony-stimulating factor 3 (CSF3) that was enriched in transplanted steatotic livers was potentially participated in fatty graft injury. We have also detected a subset of dendritic cells (DCs) with highly expressing XCR1 that was correlated with CD8+ T cells, mediating the severer steatotic liver damage by I/R injury. CONCLUSIONS: The findings of our study provide new insight into the mechanisms by which steatosis exacerbates liver damage from I/R injury. Interventions based on these observations create opportunities in attenuating fatty liver graft injury and expanding the donor pool.


Subject(s)
Fatty Liver/immunology , Liver/immunology , Reperfusion Injury/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Disease Models, Animal , Kupffer Cells/immunology , Liver Transplantation/methods , Phenotype , Rats , Rats, Sprague-Dawley , Single-Cell Analysis/methods , Transcription, Genetic/immunology
9.
J Immunol ; 207(2): 613-625, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34272232

ABSTRACT

Alphaherpesviruses are large dsDNA viruses with an ability to establish persistent infection in hosts, which rely partly on their ability to evade host innate immune responses, notably the type I IFN response. However, the relevant molecular mechanisms are not well understood. In this study, we report the UL42 proteins of alphaherpesvirus pseudorabies virus (PRV) and HSV type 1 (HSV1) as a potent antagonist of the IFN-I-induced JAK-STAT signaling pathway. We found that ectopic expression of UL42 in porcine macrophage CRL and human HeLa cells significantly suppresses IFN-α-mediated activation of the IFN-stimulated response element (ISRE), leading to a decreased transcription and expression of IFN-stimulated genes (ISGs). Mechanistically, UL42 directly interacts with ISRE and interferes with ISG factor 3 (ISGF3) from binding to ISRE for efficient gene transcription, and four conserved DNA-binding sites of UL42 are required for this interaction. The substitution of these DNA-binding sites with alanines results in reduced ISRE-binding ability of UL42 and impairs for PRV to evade the IFN response. Knockdown of UL42 in PRV remarkably attenuates the antagonism of virus to IFN in porcine kidney PK15 cells. Our results indicate that the UL42 protein of alphaherpesviruses possesses the ability to suppress IFN-I signaling by preventing the association of ISGF3 and ISRE, thereby contributing to immune evasion. This finding reveals UL42 as a potential antiviral target.


Subject(s)
DNA-Directed DNA Polymerase/immunology , Exodeoxyribonucleases/immunology , Herpesvirus 1, Suid/immunology , Interferon Type I/immunology , Interferon-Stimulated Gene Factor 3, gamma Subunit/immunology , Viral Proteins/immunology , Animals , Cell Line , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/immunology , Humans , Immune Evasion/immunology , Immunity, Innate/immunology , Pseudorabies/immunology , Response Elements/immunology , Signal Transduction/immunology , Swine , Transcription, Genetic/immunology
10.
Emerg Microbes Infect ; 10(1): 1320-1330, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34112056

ABSTRACT

Ebola virus (EBOV) is a negative single-stranded RNA virus within the Filoviridae family and the causative agent of Ebola virus disease (EVD). Nonhuman primates (NHPs), including cynomolgus and rhesus macaques, are considered the gold standard animal model to interrogate mechanisms of EBOV pathogenesis. However, despite significant genetic similarity (>90%), NHP species display different clinical presentation following EBOV infection, notably a ∼1-2 days delay in disease progression. Consequently, evaluation of therapeutics is generally conducted in rhesus macaques, whereas cynomolgus macaques are utilized to determine efficacy of preventative treatments, notably vaccines. This observation is in line with reported differences in disease severity and host responses between these two NHP following infection with simian varicella virus, influenza A and SARS-CoV-2. However, the molecular underpinnings of these differential outcomes following viral infections remain poorly defined. In this study, we compared published transcriptional profiles obtained from cynomolgus and rhesus macaques infected with the EBOV-Makona Guinea C07 using bivariate and regression analyses to elucidate differences in host responses. We report the presence of a shared core of differentially expressed genes (DEGs) reflecting EVD pathology, including aberrant inflammation, lymphopenia, and coagulopathy. However, the magnitudes of change differed between the two macaque species. These findings suggest that the differential clinical presentation of EVD in these two species is mediated by altered transcriptional responses.


Subject(s)
Gene Expression Regulation/immunology , Hemorrhagic Fever, Ebola/veterinary , Macaca fascicularis , Macaca mulatta , Monkey Diseases/immunology , Transcription, Genetic/immunology , Animals , COVID-19 , Ebolavirus , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/mortality , Humans , Immunity , Monkey Diseases/genetics , Monkey Diseases/mortality , RNA, Viral/metabolism , SARS-CoV-2 , Species Specificity
11.
Sci Rep ; 11(1): 11498, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34075120

ABSTRACT

MicroRNA miR-155 is an important regulatory molecule in the immune system and is highly expressed and functional in Th17 cells, a subset of CD4+ T helper cells which are key players in autoimmune diseases. Small molecules that can modulate miR-155 may potentially provide new therapeutic avenues to inhibit Th17 cell-mediated autoimmune diseases. Here, we present a novel high-throughput screening assay using primary T cells from genetically engineered Mir155 reporter mice, and its use to screen libraries of small molecules to identify novel modulators of Th17 cell function. We have discovered a chemical series of (E)-1-(phenylsulfonyl)-2-styryl-1H-benzo[d] imidazoles as novel down-regulators of Mir155 reporter and cytokine expression in Th17 cells. In addition, we found that FDA approved antiparasitic agents belonging to the 'azole' family also down-regulate Mir155 reporter and cytokine expression in Th17 cells, and thus could potentially be repurposed to treat Th17-driven immunopathologies.


Subject(s)
Down-Regulation/drug effects , Genes, Reporter , Imidazoles/pharmacology , MicroRNAs/biosynthesis , Th17 Cells/metabolism , Transcription, Genetic/drug effects , Animals , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Down-Regulation/genetics , Down-Regulation/immunology , Imidazoles/chemistry , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/immunology , Th17 Cells/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology
12.
Front Immunol ; 12: 639489, 2021.
Article in English | MEDLINE | ID: mdl-33968031

ABSTRACT

Rainbow trout (Oncorhynchus mykiss) is one of the most common aquaculture fish species worldwide. Vibriosis disease outbreaks cause significant setbacks to aquaculture. The stress and immune responses are bidirectionally modulated in response to the health challenges. Therefore, an investigation into the regulatory mechanisms of the stress and immune responses in trout is invaluable for identifying potential vibriosis treatments. We investigated the transcriptional profiles of genes associated with stress and trout immune functions after Vibrio anguillarum infection. We compared the control trout (CT, 0.9% saline injection), asymptomatic trout (AT, surviving trout with minor or no symptoms after bacteria injection), and symptomatic trout (ST, moribund trout with severe symptoms after bacteria injection). Our results showed activated immunomodulatory genes in the cytokine network and downregulated glucocorticoid and mineralocorticoid receptors in both AT and ST, indicating activation of the proinflammatory cytokine cascade as a common response in AT and ST. Moreover, the AT specifically activated the complement- and TNF-associated immune defenses in response to V. anguillarum infection. However, the complement and coagulation cascades, as well as steroid hormone homeostasis in ST, were disturbed by V. anguillarum. Our studies provide new insights toward understanding regulatory mechanisms in stress and immune functions in response to diseases.


Subject(s)
Immunity/genetics , Immunity/immunology , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Vibrio/immunology , Animals , Complement System Proteins/genetics , Complement System Proteins/immunology , Cytokines/genetics , Cytokines/immunology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/microbiology , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Oncorhynchus mykiss/microbiology , Vibrio Infections/genetics , Vibrio Infections/immunology , Vibrio Infections/microbiology
13.
Front Immunol ; 12: 639634, 2021.
Article in English | MEDLINE | ID: mdl-34025649

ABSTRACT

The prognosis of acute myeloid leukemia (AML) is closely related to immune response changes. Further exploration of the pathobiology of AML focusing on immune-related genes would contribute to the development of more advanced evaluation and treatment strategies. In this study, we established a novel immune-17 signature based on transcriptome data from The Cancer Genome Atlas (TCGA) and The Genotype-Tissue Expression (GTEx) databases. We found that immune biology processes and transcriptional dysregulations are critical factors in the development of AML through enrichment analyses. We also formulated a prognostic model to predict the overall survival of AML patients by using LASSO (Least Absolute Shrinkage and Selection Operator) regression analysis. Furthermore, we incorporated the immune-17 signature to improve the prognostic accuracy of the ELN2017 risk stratification system. We concluded that the immune-17 signature represents a novel useful model for evaluating AML survival outcomes and may be implemented to optimize treatment selection in the next future.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Prognosis , Transcription, Genetic/genetics , Transcription, Genetic/immunology , Transcriptome/genetics , Transcriptome/immunology
14.
J Exp Med ; 218(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34014253

ABSTRACT

Beyond hemostasis, platelets actively participate in immune cell recruitment and host defense, yet their potential in the resolution of inflammatory processes remains unknown. Here, we demonstrate that platelets are recruited into the lung together with neutrophils during the onset of inflammation and alongside regulatory T (T reg) cells during the resolution phase. This partnering dichotomy is regulated by differential adhesion molecule expression during resolution. Mechanistically, intravascular platelets form aggregates with T reg cells, a prerequisite for their recruitment into the lung. This interaction relies on platelet activation by sCD40L and platelet P-selectin binding to PSGL-1 on T reg cells. Physical platelet-T reg cell interactions are necessary to modulate the transcriptome and instruct T reg cells to release the anti-inflammatory mediators IL-10 and TGFß. Notably, the presence of platelet-T reg cell aggregates in the lung was also required for macrophage transcriptional reprogramming, polarization toward an anti-inflammatory phenotype, and effective resolution of pulmonary inflammation. Thus, platelets partner with successive immune cell subsets to orchestrate both the initiation and resolution of inflammation.


Subject(s)
Blood Platelets/immunology , Lung/immunology , Macrophages/immunology , Pneumonia/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Adhesion/immunology , Hemostasis/immunology , Male , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Transcription, Genetic/immunology
15.
J Biol Chem ; 296: 100687, 2021.
Article in English | MEDLINE | ID: mdl-33891947

ABSTRACT

Glucocorticoids are potent anti-inflammatory drugs that are used to treat an extraordinary range of human disease, including COVID-19, underscoring the ongoing importance of understanding their molecular mechanisms. Early studies of GR signaling led to broad acceptance of models in which glucocorticoid receptor (GR) monomers tether repressively to inflammatory transcription factors, thus abrogating inflammatory gene expression. However, newer data challenge this core concept and present an exciting opportunity to reframe our understanding of GR signaling. Here, we present an alternate, two-part model for transcriptional repression by glucocorticoids. First, widespread GR-mediated induction of transcription results in rapid, primary repression of inflammatory gene transcription and associated enhancers through competition-based mechanisms. Second, a subset of GR-induced genes, including targets that are regulated in coordination with inflammatory transcription factors such as NF-κB, exerts secondary repressive effects on inflammatory gene expression. Within this framework, emerging data indicate that the gene set regulated through the cooperative convergence of GR and NF-κB signaling is central to the broad clinical effectiveness of glucocorticoids in terminating inflammation and promoting tissue repair.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , COVID-19 Drug Treatment , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , NF-kappa B/genetics , Receptors, Glucocorticoid/genetics , Animals , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Gene Expression Regulation , Genomics/methods , Humans , Inflammation/prevention & control , Models, Genetic , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/immunology , SARS-CoV-2/growth & development , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Signal Transduction , Transcription, Genetic/drug effects , Transcription, Genetic/immunology
16.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800829

ABSTRACT

Tumor-associated macrophages (TAMs) are the essential components of the tumor microenvironment. TAMs originate from blood monocytes and undergo pro- or anti-inflammatory polarization during their life span within the tumor. The balance between macrophage functional populations and the efficacy of their antitumor activities rely on the transcription factors such as STAT1, NF-κB, IRF, and others. These molecular tools are of primary importance, as they contribute to the tumor adaptations and resistance to radio- and chemotherapy and can become important biomarkers for theranostics. Herein, we describe the major transcriptional mechanisms specific for TAM, as well as how radio- and chemotherapy can impact gene transcription and functionality of macrophages, and what are the consequences of the TAM-tumor cooperation.


Subject(s)
Antineoplastic Agents/adverse effects , Gene Expression Regulation, Neoplastic , Immunotherapy/adverse effects , Radiotherapy/adverse effects , Transcription, Genetic , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/radiation effects , Antineoplastic Agents/pharmacology , Cytokines/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Inflammation , Interferon Regulatory Factors/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/radiotherapy , STAT Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/immunology , Transcription, Genetic/radiation effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Tumor Suppressor Protein p53/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
17.
Int J Mol Sci ; 22(5)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803441

ABSTRACT

Interferon regulatory factor-4 (IRF4) and IRF8 regulate differentiation, growth and functions of lymphoid and myeloid cells. Targeted deletion of irf8 in T cells (CD4-IRF8KO) has been shown to exacerbate colitis and experimental autoimmune uveitis (EAU), a mouse model of human uveitis. We therefore generated mice lacking irf4 in T cells (CD4-IRF4KO) and investigated whether expression of IRF4 by T cells is also required for regulating T cells that suppress autoimmune diseases. Surprisingly, we found that CD4-IRF4KO mice are resistant to EAU. Suppression of EAU derived in part from inhibiting pathogenic responses of Th17 cells while inducing expansion of regulatory lymphocytes that secrete IL-10 and/or IL-35 in the eye and peripheral lymphoid tissues. Furthermore, CD4-IRF4KO T cells exhibit alterations in cell metabolism and are defective in the expression of two Ikaros zinc-finger (IKZF) transcription factors (Ikaros, Aiolos) that are required for lymphocyte differentiation, metabolism and cell-fate decisions. Thus, synergistic effects of IRF4 and IkZFs might induce metabolic reprogramming of differentiating lymphocytes and thereby dynamically regulate relative abundance of T and B lymphocyte subsets that mediate immunopathogenic mechanisms during uveitis. Moreover, the diametrically opposite effects of IRF4 and IRF8 during EAU suggests that intrinsic function of IRF4 in T cells might be activating proinflammatory responses while IRF8 promotes expansion of immune-suppressive mechanisms.


Subject(s)
Autoimmune Diseases , CD4-Positive T-Lymphocytes , Cell Differentiation , Gene Deletion , Interferon Regulatory Factors/deficiency , Transcription, Genetic/immunology , Uveitis , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Mice , Mice, Knockout , Uveitis/genetics , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology
18.
Eur J Immunol ; 51(7): 1824-1838, 2021 07.
Article in English | MEDLINE | ID: mdl-33818775

ABSTRACT

Multiple factors linked to host genetics/inherent biology play a role in interindividual variability in immune response outcomes after rubella vaccination. In order to identify these factors, we conducted a study of rubella-specific humoral immunity before (Baseline) and after (Day 28) a third dose of MMR-II vaccine in a cohort of 109 women of childbearing age. We performed mRNA-Seq profiling of PBMCs after rubella virus in vitro stimulation to delineate genes associated with post-vaccination rubella humoral immunity and to define genes mediating the association between prior immune response status (high or low antibody) and subsequent immune response outcome. Our study identified novel genes that mediated the association between prior immune response and neutralizing antibody titer after a third MMR vaccine dose. These genes included the following: CDC34; CSNK1D; APOBEC3F; RAD18; AAAS; SLC37A1; FAS; and JAK2. The encoded proteins are involved in innate antiviral response, IFN/cytokine signaling, B cell repertoire generation, the clonal selection of B lymphocytes in germinal centers, and somatic hypermutation/antibody affinity maturation to promote optimal antigen-specific B cell immune function. These data advance our understanding of how subjects' prior immune status and/or genetic propensity to respond to rubella/MMR vaccination ultimately affects innate immunity and humoral immune outcomes after vaccination.


Subject(s)
Immunity, Humoral/immunology , Measles-Mumps-Rubella Vaccine/immunology , Rubella virus/immunology , Transcription, Genetic/immunology , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Cohort Studies , Female , Humans , Immunity, Innate/immunology , Leukocytes, Mononuclear/immunology , Middle Aged , Rubella/immunology , Vaccination/methods , Young Adult
19.
Int J Mol Sci ; 22(4)2021 Feb 09.
Article in English | MEDLINE | ID: mdl-33572313

ABSTRACT

Long noncoding RNAs (lncRNAs) represent key regulators of gene transcription during the inflammatory response. Recent findings showed lncRNAs to be dysregulated in human diseases, such as inflammatory bowel disease, diabetes, allergies, asthma, and cancer. These noncoding RNAs are crucial for immune mechanism, as they are involved in differentiation, cell migration and in the production of inflammatory mediators through regulating protein-protein interactions or their ability to assemble with RNA and DNA. The last interaction can occur in cis or trans and is responsible for all the possible lncRNAs biological effects. Our proposal is to provide an overview on lncRNAs roles and functions related to immunity and immune mediated diseases, since these elucidations could be beneficial to untangle the complex bond between them.


Subject(s)
Adaptive Immunity/genetics , Autoimmune Diseases/genetics , Gene Expression Regulation/immunology , Immunity, Innate/genetics , RNA, Long Noncoding/metabolism , Animals , Autoimmune Diseases/immunology , Cell Movement/genetics , Cell Movement/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Models, Animal , Oligonucleotides/metabolism , Protein Interaction Maps/genetics , Protein Interaction Maps/immunology , RNA, Long Noncoding/genetics , Transcription, Genetic/immunology
20.
Cell Rep ; 34(6): 108742, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33567280

ABSTRACT

Glucocorticoids (GCs) are effective anti-inflammatory drugs; yet, their mechanisms of action are poorly understood. GCs bind to the glucocorticoid receptor (GR), a ligand-gated transcription factor controlling gene expression in numerous cell types. Here, we characterize GR's protein interactome and find the SETD1A (SET domain containing 1A)/COMPASS (complex of proteins associated with Set1) histone H3 lysine 4 (H3K4) methyltransferase complex highly enriched in activated mouse macrophages. We show that SETD1A/COMPASS is recruited by GR to specific cis-regulatory elements, coinciding with H3K4 methylation dynamics at subsets of sites, upon treatment with lipopolysaccharide (LPS) and GCs. By chromatin immunoprecipitation sequencing (ChIP-seq) and RNA-seq, we identify subsets of GR target loci that display SETD1A occupancy, H3K4 mono-, di-, or tri-methylation patterns, and transcriptional changes. However, our data on methylation status and COMPASS recruitment suggest that SETD1A has additional transcriptional functions. Setd1a loss-of-function studies reveal that SETD1A/COMPASS is required for GR-controlled transcription of subsets of macrophage target genes. We demonstrate that the SETD1A/COMPASS complex cooperates with GR to mediate anti-inflammatory effects.


Subject(s)
Enhancer Elements, Genetic/immunology , Macrophages/immunology , Multiprotein Complexes , RNA-Seq , Receptors, Glucocorticoid , Transcription, Genetic/immunology , Animals , Inflammation/genetics , Inflammation/immunology , Mice , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...