Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
PLoS Pathog ; 20(5): e1012230, 2024 May.
Article in English | MEDLINE | ID: mdl-38776321

ABSTRACT

While macrophage is one of the major type I interferon (IFN-I) producers in multiple tissues during viral infections, it also serves as an important target cell for many RNA viruses. However, the regulatory mechanism for the IFN-I response of macrophages to respond to a viral challenge is not fully understood. Here we report ADAP, an immune adaptor protein, is indispensable for the induction of the IFN-I response of macrophages to RNA virus infections via an inhibition of the conjugation of ubiquitin-like ISG15 (ISGylation) to RIG-I. Loss of ADAP increases RNA virus replication in macrophages, accompanied with a decrease in LPS-induced IFN-ß and ISG15 mRNA expression and an impairment in the RNA virus-induced phosphorylation of IRF3 and TBK1. Moreover, using Adap-/- mice, we show ADAP deficiency strongly increases the susceptibility of macrophages to RNA-virus infection in vivo. Mechanically, ADAP selectively interacts and functionally cooperates with RIG-I but not MDA5 in the activation of IFN-ß transcription. Loss of ADAP results in an enhancement of ISGylation of RIG-I, whereas overexpression of ADAP exhibits the opposite effect in vitro, indicating ADAP is detrimental to the RNA virus-induced ISGylation of RIG-I. Together, our data demonstrate a novel antagonistic activity of ADAP in the cell-intrinsic control of RIG-I ISGylation, which is indispensable for initiating and sustaining the IFN-I response of macrophages to RNA virus infections and replication.


Subject(s)
Adaptor Proteins, Signal Transducing , DEAD Box Protein 58 , Interferon Type I , Macrophages , Mice, Knockout , RNA Virus Infections , Ubiquitins , Animals , Macrophages/virology , Macrophages/metabolism , Macrophages/immunology , Mice , RNA Virus Infections/immunology , RNA Virus Infections/metabolism , Ubiquitins/metabolism , Ubiquitins/genetics , DEAD Box Protein 58/metabolism , Interferon Type I/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cytokines/metabolism , Mice, Inbred C57BL , Humans , Receptors, Immunologic/metabolism , Interferon-beta/metabolism , RNA Viruses/immunology , Interferon Regulatory Factor-3/metabolism
2.
Microbiome ; 12(1): 58, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38504332

ABSTRACT

BACKGROUND: Microbiota are closely associated with human health and disease. Metaproteomics can provide a direct means to identify microbial proteins in microbiota for compositional and functional characterization. However, in-depth and accurate metaproteomics is still limited due to the extreme complexity and high diversity of microbiota samples. It is generally recommended to use metagenomic data from the same samples to construct the protein sequence database for metaproteomic data analysis. Although different metagenomics-based database construction strategies have been developed, an optimization of gene taxonomic annotation has not been reported, which, however, is extremely important for accurate metaproteomic analysis. RESULTS: Herein, we proposed an accurate taxonomic annotation pipeline for genes from metagenomic data, namely contigs directed gene annotation (ConDiGA), and used the method to build a protein sequence database for metaproteomic analysis. We compared our pipeline (ConDiGA or MD3) with two other popular annotation pipelines (MD1 and MD2). In MD1, genes were directly annotated against the whole bacterial genome database; in MD2, contigs were annotated against the whole bacterial genome database and the taxonomic information of contigs was assigned to the genes; in MD3, the most confident species from the contigs annotation results were taken as reference to annotate genes. Annotation tools, including BLAST, Kaiju, and Kraken2, were compared. Based on a synthetic microbial community of 12 species, it was found that Kaiju with the MD3 pipeline outperformed the others in the construction of protein sequence database from metagenomic data. Similar performance was also observed with a fecal sample, as well as in silico mixed datasets of the simulated microbial community and the fecal sample. CONCLUSIONS: Overall, we developed an optimized pipeline for gene taxonomic annotation to construct protein sequence databases. Our study can tackle the current taxonomic annotation reliability problem in metagenomics-derived protein sequence database and can promote the in-depth metaproteomic analysis of microbiome. The unique metagenomic and metaproteomic datasets of the 12 bacterial species are publicly available as a standard benchmarking sample for evaluating various analysis pipelines. The code of ConDiGA is open access at GitHub for the analysis of microbiota samples. Video Abstract.


Subject(s)
Microbiota , Humans , Databases, Protein , Molecular Sequence Annotation , Reproducibility of Results , Microbiota/genetics , Metagenome/genetics , Bacteria/genetics , Metagenomics/methods
3.
Cell Rep ; 43(3): 113877, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38421869

ABSTRACT

Combination therapy (lenvatinib/programmed death-1 inhibitor) is effective for treating unresectable hepatocellular carcinoma (uHCC). We reveal that responders have better overall and progression-free survival, as well as high tumor mutation burden and special somatic variants. We analyze the proteome and metabolome of 82 plasma samples from patients with hepatocellular carcinoma (HCC; n = 51) and normal controls (n = 15), revealing that individual differences outweigh treatment differences. Responders exhibit enhanced activity in the alternative/lectin complement pathway and higher levels of lysophosphatidylcholines (LysoPCs), predicting a favorable prognosis. Non-responders are enriched for immunoglobulins, predicting worse outcomes. Compared to normal controls, HCC plasma proteins show acute inflammatory response and platelet activation, while LysoPCs decrease. Combination therapy increases LysoPCs/phosphocholines in responders. Logistic regression/random forest models using metabolomic features achieve good performance in the prediction of responders. Proteomic analysis of cancer tissues unveils molecular features that are associated with side effects in responders receiving combination therapy. In conclusion, our analysis identifies plasma features associated with uHCC responders to combination therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Phenylurea Compounds , Quinolines , Humans , Carcinoma, Hepatocellular/drug therapy , Proteomics , Liver Neoplasms/drug therapy , Combined Modality Therapy
4.
Nucleic Acids Res ; 52(D1): D1407-D1417, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37739405

ABSTRACT

Advances in sequencing and imaging technologies offer a unique opportunity to unravel cell heterogeneity and develop new immunotherapy strategies for cancer research. There is an urgent need for a resource that effectively integrates a vast amount of transcriptomic profiling data to comprehensively explore cancer tissue heterogeneity and the tumor microenvironment. In this context, we developed the Single-cell and Spatially-resolved Cancer Resources (SCAR) database, a combined tumor spatial and single-cell transcriptomic platform, which is freely accessible at http://8.142.154.29/SCAR2023 or http://scaratlas.com. SCAR contains spatial transcriptomic data from 21 tumor tissues and single-cell transcriptomic data from 11 301 352 cells encompassing 395 cancer subtypes and covering a wide variety of tissues, organoids, and cell lines. This resource offers diverse functional modules to address key cancer research questions at multiple levels, including the screening of tumor cell types, metabolic features, cell communication and gene expression patterns within the tumor microenvironment. Moreover, SCAR enables the analysis of biomarker expression patterns and cell developmental trajectories. SCAR also provides a comprehensive analysis of multi-dimensional datasets based on 34 state-of-the-art omics techniques, serving as an essential tool for in-depth mining and understanding of cell heterogeneity and spatial location. The implications of this resource extend to both cancer biology research and cancer immunotherapy development.


Subject(s)
Databases, Factual , Gene Expression Profiling , Neoplasms , Humans , Cell Differentiation , Gene Expression Profiling/methods , Neoplasms/genetics , Neoplasms/pathology , Transcriptome , Tumor Microenvironment , Single-Cell Analysis
5.
Nucleic Acids Res ; 52(D1): D998-D1009, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37930842

ABSTRACT

The nervous system is one of the most complicated and enigmatic systems within the animal kingdom. Recently, the emergence and development of spatial transcriptomics (ST) and single-cell RNA sequencing (scRNA-seq) technologies have provided an unprecedented ability to systematically decipher the cellular heterogeneity and spatial locations of the nervous system from multiple unbiased aspects. However, efficiently integrating, presenting and analyzing massive multiomic data remains a huge challenge. Here, we manually collected and comprehensively analyzed high-quality scRNA-seq and ST data from the nervous system, covering 10 679 684 cells. In addition, multi-omic datasets from more than 900 species were included for extensive data mining from an evolutionary perspective. Furthermore, over 100 neurological diseases (e.g. Alzheimer's disease, Parkinson's disease, Down syndrome) were systematically analyzed for high-throughput screening of putative biomarkers. Differential expression patterns across developmental time points, cell types and ST spots were discerned and subsequently subjected to extensive interpretation. To provide researchers with efficient data exploration, we created a new database with interactive interfaces and integrated functions called the Spatiotemporal Cloud Atlas for Neural cells (SCAN), freely accessible at http://47.98.139.124:8799 or http://scanatlas.net. SCAN will benefit the neuroscience research community to better exploit the spatiotemporal atlas of the neural system and promote the development of diagnostic strategies for various neurological disorders.


Subject(s)
Databases, Genetic , Nervous System Diseases , Neurons , Single-Cell Gene Expression Analysis , Animals , Neurons/metabolism , Atlases as Topic , Nervous System Diseases/genetics
6.
Adv Sci (Weinh) ; 10(3): e2205462, 2023 01.
Article in English | MEDLINE | ID: mdl-36453571

ABSTRACT

Acetylation of extracellular proteins has been observed in many independent studies where particular attention has been given to the dynamic change of the microenvironmental protein post-translational modifications. While extracellular proteins can be acetylated within the cells prior to their micro-environmental distribution, their deacetylation in a tumor microenvironment remains elusive. Here it is described that multiple acetyl-vWA domain-carrying proteins including integrin ß3 (ITGB3) and collagen 6A (COL6A) are deacetylated by Sirtuin family member SIRT2 in extracellular space. SIRT2 is secreted by macrophages following toll-like receptor (TLR) family member TLR4 or TLR2 activation. TLR-activated SIRT2 undergoes autophagosome translocation. TNF receptor associated factor 6 (TRAF6)-mediated autophagy flux in response to TLR2/4 activation can then pump SIRT2 into the microenvironment to function as extracellular SIRT2 (eSIRT2). In the extracellular space, eSIRT2 deacetylates ITGB3 on aK416 involved in cell attachment and migration, leading to a promotion of cancer cell metastasis. In lung cancer patients, significantly increased serum eSIRT2 level correlates with dramatically decreased ITGB3-K416 acetylation in cancer cells. Thus, the extracellular space is a subcellular organelle-like arena where eSIRT2 promotes cancer cell metastasis via catalyzing extracellular protein deacetylation.


Subject(s)
Lung Neoplasms , Sirtuin 2 , Humans , Sirtuin 2/genetics , Sirtuin 2/metabolism , Toll-Like Receptor 2/metabolism , Protein Processing, Post-Translational , Acetylation , Tumor Microenvironment
7.
Nucleic Acids Res ; 51(D1): D1150-D1159, 2023 01 06.
Article in English | MEDLINE | ID: mdl-36305818

ABSTRACT

It is a challenge to efficiently integrate and present the tremendous amounts of single-cell data generated from multiple tissues of various species. Here, we create a new database named SPEED for single-cell pan-species atlas in the light of ecology and evolution for development and diseases (freely accessible at http://8.142.154.29 or http://speedatlas.net). SPEED is an online platform with 4 data modules, 7 function modules and 2 display modules. The 'Pan' module is applied for the interactive analysis of single cell sequencing datasets from 127 species, and the 'Evo', 'Devo', and 'Diz' modules provide comprehensive analysis of single-cell atlases on 18 evolution datasets, 28 development datasets, and 85 disease datasets. The 'C2C', 'G2G' and 'S2S' modules explore intercellular communications, genetic regulatory networks, and cross-species molecular evolution. The 'sSearch', 'sMarker', 'sUp', and 'sDown' modules allow users to retrieve specific data information, obtain common marker genes for cell types, freely upload, and download single-cell datasets, respectively. Two display modules ('HOME' and 'HELP') offer easier access to the SPEED database with informative statistics and detailed guidelines. All in all, SPEED is an integrated platform for single-cell RNA sequencing (scRNA-seq) and single-cell whole-genome sequencing (scWGS) datasets to assist the deep-mining and understanding of heterogeneity among cells, tissues, and species at multi-levels, angles, and orientations, as well as provide new insights into molecular mechanisms of biological development and pathogenesis.


Subject(s)
Databases, Factual , Single-Cell Analysis , Humans , Animals , Biological Evolution , Plants/genetics , Ecology
8.
Cell Mol Immunol ; 19(8): 898-912, 2022 08.
Article in English | MEDLINE | ID: mdl-35637282

ABSTRACT

Heightened platelet phagocytosis by macrophages accompanied by an increase in IFN-γ play key roles in the etiology of immune thrombocytopenia (ITP); however, it remains elusive how macrophage-mediated platelet clearance is regulated in ITP. Here, we report that adhesion and degranulation-protein adaptor protein (ADAP) restrains platelet phagocytosis by macrophages in ITP via modulation of signal transducer and activator of transcription 1 (STAT1)-FcγR signaling. We show that ITP was associated with the underexpression of ADAP in splenic macrophages. Furthermore, macrophages from Adap-/- mice exhibited elevated platelet phagocytosis and upregulated proinflammatory signaling, and thrombocytopenia in Adap-/- mice was mitigated by the depletion of macrophages. Mechanistically, ADAP interacted and competed with STAT1 binding to importin α5. ADAP deficiency potentiated STAT1 nuclear entry, leading to a selective enhancement of FcγRI/IV transcription in macrophages. Moreover, pharmacological inhibition of STAT1 or disruption of the STAT1-importin α5 interaction relieved thrombocytopenia in Adap-/- mice. Thus, our findings not only reveal a critical role for ADAP as an intracellular immune checkpoint for shaping macrophage phagocytosis in ITP but also identify the ADAP-STAT1-importin α5 module as a promising therapeutic target in the treatment of ITP.


Subject(s)
Adaptor Proteins, Signal Transducing , Macrophages , Phagocytosis , Purpura, Thrombocytopenic, Idiopathic , STAT1 Transcription Factor , Thrombocytopenia , Adaptor Proteins, Signal Transducing/metabolism , Animals , Karyopherins , Macrophages/metabolism , Mice , STAT1 Transcription Factor/metabolism , Thrombocytopenia/metabolism
9.
EMBO Mol Med ; 14(2): e14713, 2022 02 07.
Article in English | MEDLINE | ID: mdl-34978375

ABSTRACT

The prevalence of intracranial aneurysm (IA) is increasing, and the consequences of its rupture are severe. This study aimed to reveal specific, sensitive, and non-invasive biomarkers for diagnosis and classification of ruptured and unruptured IA, to benefit the development of novel treatment strategies and therapeutics altering the course of the disease. We first assembled an extensive candidate biomarker bank of IA, comprising up to 717 proteins, based on altered proteins discovered in the current tissue and serum proteomic analysis, as well as from previous studies. Mass spectrometry assays for hundreds of biomarkers were efficiently designed using our proposed deep learning-based method, termed DeepPRM. A total of 113 potential markers were further quantitated in serum cohort I (n = 212) & II (n = 32). Combined with a machine-learning-based pipeline, we built two sets of biomarker combinations (P6 & P8) to accurately distinguish IA from healthy controls (accuracy: 87.50%) or classify IA rupture patients (accuracy: 91.67%) upon evaluation in the external validation set (n = 32). This extensive circulating biomarker development study provides valuable knowledge about IA biomarkers.


Subject(s)
Aneurysm, Ruptured , Intracranial Aneurysm , Aneurysm, Ruptured/diagnosis , Aneurysm, Ruptured/metabolism , Biomarkers , Humans , Intracranial Aneurysm/diagnosis , Intracranial Aneurysm/metabolism , Proteomics , Risk Assessment
10.
Anal Chem ; 93(31): 10940-10946, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34319068

ABSTRACT

With a nearly 100% mortality rate, African swine fever (ASF) has devastated the pork industry in many countries. Without a vaccine in sight, mitigation rests on rapid diagnosis and immediately depopulating infected or exposed animals. Unfortunately, current tests require centralized laboratories with well-trained personnel, take days to report the results, and thus do not meet the need for such rapid diagnosis. In response, we developed a portable, sample-to-answer device that allows for ASF detection at the point of need in <30 min. The device employs droplet magnetofluidics to automate DNA purification from blood, tissue, or swab samples and utilizes fast thermal cycling to perform real-time quantitative polymerase chain reaction (qPCR), all within an inexpensive disposable cartridge. We evaluated its diagnostic performance at six farms and slaughter facilities. The device exhibits high diagnostic accuracy with a positive percent agreement of 92.2% and a negative percent agreement of 93.6% compared with a lab-based reference qPCR test.


Subject(s)
African Swine Fever Virus , African Swine Fever , African Swine Fever/diagnosis , African Swine Fever Virus/genetics , Animals , Real-Time Polymerase Chain Reaction , Swine
11.
Biomed Res Int ; 2021: 6098978, 2021.
Article in English | MEDLINE | ID: mdl-34195271

ABSTRACT

PURPOSE: Retrospective analysis of the effect of portable 3D gait analysis as an innovative evaluation method in the treatment with MTT on chronic ankle instability patient. METHODS: From January 1, 2019, to December 31, 2019, 56 cases of chronic ankle instability (CAI) were extracted from the medical record system of Shenzhen Longhua District Central Hospital. All the patients of 56 cases accepted the medical training therapy (MTT). As outcome parameters, the alterations of the Cumberland ankle instability tool (CAIT), foot and ankle ability measure (FAAM), were used before the treatment and after treatment; meanwhile, the portable apparatus 3D gait analysis was used to measure the gait parameters. CONCLUSION: The results showed only ankle angle parameters Y-axis, maximum dorsiflexion during support period (°) had a significant difference, and the p value is 0.039. Meanwhile, the CAIT, FAAM, and most 3D gait analysis data had no significant difference. This particular statistical difference shows that CAI can be measured scientifically and objectively, although most measurement parameters have no change. These results make further reveal that the CAI patients are suffering with dynamic abnormality of ankle motion angle; this also provides us with a measurable and systematic evaluation reference plan for CAI treatment in the future.


Subject(s)
Ankle/diagnostic imaging , Gait , Imaging, Three-Dimensional/methods , Joint Instability/diagnostic imaging , Range of Motion, Articular , Adult , Ankle Injuries , Ankle Joint/diagnostic imaging , Biomechanical Phenomena , China/epidemiology , Chronic Disease , Exercise , Female , Gait Analysis , Humans , Male , Middle Aged , Motion , Postural Balance , Retrospective Studies
12.
J Immunol ; 206(4): 814-826, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33431658

ABSTRACT

Adhesion and degranulation-promoting adapter protein (ADAP), originally identified as an essential adaptor molecule in TCR signaling and T cell adhesion, has emerged as a critical regulator in innate immune cells such as macrophages; however, its role in macrophage polarization and inflammatory responses remains unknown. In this study, we show that ADAP plays an essential role in TLR4-mediated mouse macrophage polarization via modulation of STAT3 activity. Macrophages from ADAP-deficient mice exhibit enhanced M1 polarization, expression of proinflammatory cytokines and capacity in inducing Th1 responses, but decreased levels of anti-inflammatory cytokines in response to TLR4 activation by LPS. Furthermore, overexpression of ADAP enhances, whereas loss of ADAP reduces, the LPS-mediated phosphorylation and activity of STAT3, suggesting ADAP acts as a coactivator of STAT3 activity and function. Furthermore, the coactivator function of ADAP mostly depends on the tyrosine phosphorylation at Y571 in the motif YDSL induced by LPS. Mutation of Y571 to F severely impairs the stimulating effect of ADAP on STAT3 activity and the ability of ADAP to inhibit M1-like polarization in TLR4-activated mouse macrophages. Moreover, ADAP interacts with STAT3, and loss of ADAP renders mouse macrophages less sensitive to IL-6 stimulation for STAT3 phosphorylation. Collectively, our findings revealed an additional layer of regulation of TLR4-mediated mouse macrophage plasticity whereby ADAP phosphorylation on Y571 is required to prime STAT3 for activation in TLR4-stimulated mouse macrophages.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Macrophage Activation , Macrophages/immunology , STAT3 Transcription Factor/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , Animals , Female , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , STAT3 Transcription Factor/genetics , Toll-Like Receptor 4/genetics
13.
Arch Virol ; 165(10): 2249-2258, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32696270

ABSTRACT

While infectious bursal disease virus (IBDV) mainly targets immature B cells and causes T cell infiltration in the bursa of Fabricius (BF) of chickens, the effect of IBDV infection on the properties of T cells and relevant cytokine production in avian gut-associated lymphoid tissues (GALTs) remains unknown. Here, we show that while the CD8+ T cell subset is not affected, IBDV infection decreases the percentage of CD4+ T cells in the cecal tonsil (CT), but not in esophagus tonsil, pylorus tonsil, and Meckel's diverticulum of GALTs, in contrast to BF and spleen, in which the proportion of CD4+ cells increases upon IBDV infection. Further, IBDV infection upregulates IFN-γ, IL-10, and the T cell checkpoint receptor LAG-3 mRNA expression in BF. In contrast, in CTs, IBDV infection significantly increases the production of IFN-ß and CTLA-4 mRNA, while no significant effect is seen in the case of IFN-γ, IL-10 and LAG-3. Together, our data reveal differential modulation of T cell subsets and proinflammatory cytokine production in different lymphoid tissues during the course of IBDV infection.


Subject(s)
B-Lymphocyte Subsets/immunology , Birnaviridae Infections/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Poultry Diseases/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , B-Lymphocyte Subsets/virology , Birnaviridae Infections/genetics , Birnaviridae Infections/pathology , Birnaviridae Infections/virology , Bursa of Fabricius/immunology , Bursa of Fabricius/virology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Chickens/virology , Infectious bursal disease virus/growth & development , Infectious bursal disease virus/immunology , Infectious bursal disease virus/pathogenicity , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Palatine Tonsil/immunology , Palatine Tonsil/virology , Poultry Diseases/genetics , Poultry Diseases/pathology , Poultry Diseases/virology , Lymphocyte Activation Gene 3 Protein
14.
J Immunol ; 203(11): 3023-3036, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31666306

ABSTRACT

Although the immune adaptor SH2 domain containing leukocyte phosphoprotein of 76 kDa (SLP-76) integrates and propagates the TCR signaling, the regulation of SLP-76 during the TCR signaling is incompletely studied. In this article, we report that SLP-76 interacts with the small ubiquitin-like modifier (SUMO) E2 conjugase Ubc9 and is a substrate for Ubc9-mediated SUMOylation in human and mouse T cells. TCR stimulation promotes SLP-76-Ubc9 binding, accompanied by an increase in SLP-76 SUMOylation. Ubc9 binds to the extreme C terminus of SLP-76 spanning residues 516-533 and SUMOylates SLP-76 at two conserved residues K266 and K284. In addition, SLP-76 and Ubc9 synergizes to augment the TCR-mediated IL-2 transcription by NFAT in a manner dependent of SUMOylation of SLP-76. Moreover, although not affecting the TCR proximal signaling events, the Ubc9-mediated SUMOylation of SLP-76 is required for TCR-induced assembly of Ubc9-NFAT complex for IL-2 transcription. Together, these results suggest that Ubc9 modulates the function of SLP-76 in T cell activation both by direct interaction and by SUMOylation of SLP-76 and that the Ubc9-SLP-76 module acts as a novel regulatory complex in the control of T cell activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , NFATC Transcription Factors/metabolism , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Jurkat Cells , Mice , Sumoylation
15.
FASEB J ; 33(1): 286-300, 2019 01.
Article in English | MEDLINE | ID: mdl-29979632

ABSTRACT

Infectious bursal disease virus (IBDV) infection triggers the induction of type I IFN, which is mediated by melanoma differentiation-associated protein 5 recognition of the viral genomic double-stranded RNA (dsRNA). However, the mechanism of IBDV overcoming the type I IFN antiviral response remains poorly characterized. Here, we show that IBDV genomic dsRNA selectively binds to the host cellular RNA binding protein Staufen1 (STAU1) in vitro and in vivo. The viral dsRNA binding region was mapped to the N-terminal moiety of STAU1 (residues 1-468). Down-regulation of STAU1 impaired IBDV replication and enhanced IFN-ß transcription in response to IBDV infection, while having little effect on the viral attachment to the host cells and cellular entry. Conversely, overexpression of STAU1 but not the IBDV dsRNA-binding deficient STAU1 mutant (469-702) led to a suppression of IBDV dsRNA-induced IFN-ß promoter activity. Moreover, we found that the binding of STAU1 to IBDV dsRNA decreased the association of melanoma differentiation-associated protein 5 but not VP3 with the IBDV dsRNA in vitro. Finally, we showed that STAU1 and VP3 suppressed IFN-ß gene transcription in response to IBDV infection in an additive manner. Collectively, these findings provide a novel insight into the evasive strategies used by IBDV to escape the host IFN antiviral response.-Ye, C., Yu, Z., Xiong, Y., Wang, Y., Ruan, Y., Guo, Y., Chen, M., Luan, S., Zhang, E., Liu, H. STAU1 binds to IBDV genomic double-stranded RNA and promotes viral replication via attenuation of MDA5-dependent ß interferon induction.


Subject(s)
Birnaviridae Infections/virology , Cytoskeletal Proteins/metabolism , Infectious bursal disease virus/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Interferon-beta/metabolism , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Virus Replication , Animals , Antiviral Agents/metabolism , Birnaviridae Infections/genetics , Birnaviridae Infections/metabolism , Chickens , Cytoskeletal Proteins/genetics , Genomics , HEK293 Cells , HeLa Cells , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/genetics , RNA, Double-Stranded/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics
16.
FEMS Yeast Res ; 18(1)2018 02 01.
Article in English | MEDLINE | ID: mdl-29092019

ABSTRACT

The efficient promoter of alcohol oxidase 1 (PAOX1) in methylotrophic yeast Pichia pastoris is strictly induced by methanol but repressed by glycerol with an unclear molecular mechanism. In the present study, the gene of a previously characterized transmembrane protein glycerol transporter 1 (GT1) of P. pastoris GS115 was deleted by homologous recombination. Transcriptional profiles of the mutant (gt1Δ) and wild type (WT) were compared with different carbon sources (glycerol, methanol and glycerol-methanol mix) at various time points using high-throughput RNA-Seq techniques. We determined that the loss of glycerol transporter 1 (Gt1p) could relieve catabolite repression in the glycerol-methanol mixed medium and shared a similar transcriptional profile with the WT in methanol medium. By calculating the common differentially expressed genes in three distinct paired groups, genes involved in the stress response, nutrition deprivation and translational process were identified, explaining the potential roles of glycerol in the regulation of methanol metabolism. Based on weighted gene co-expression network analysis, the relationship between biological traits and the transcriptional profile was established. With the support of published research and our data, we propose two possible regulatory pathways that are involved in the regulation of catabolite repression (adenosine 5΄-monophosphate (AMP)-activated protein kinase /SNF1 and Mitogen-activated protein kinase/HOG), thereby providing potential targets for both research and industrial strain improvement.


Subject(s)
Fungal Proteins/genetics , Fungal Proteins/metabolism , Glycerol/metabolism , Methanol/metabolism , Pichia/genetics , Pichia/metabolism , Transcriptome , Biological Transport , Carbohydrate Metabolism , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Fungal , Transcription Factors/metabolism
17.
J Virol ; 92(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29093101

ABSTRACT

Infectious bursal disease virus (IBDV) is a bisegmented double-strand RNA (dsRNA) virus of the Birnaviridae family. While IBDV genomic dsRNA lacks a 5' cap, the means by which the uncapped IBDV genomic RNA is translated effectively is unknown. In this study, we describe a cap-independent pathway of translation initiation of IBDV uncapped RNA that relies on VP1 and VP3. We show that neither purified IBDV genomic dsRNA nor the uncapped viral plus-sense RNA transcripts were directly translated and rescued into infectious viruses in host cells. This defect in translation of the uncapped IBDV genomic dsRNA was rescued by trans-supplementation of the viral proteins VP1 and VP3 which was dependent on both the intact polymerase activity of VP1 and the dsRNA binding activity of VP3. Deletion analysis showed that both 5' and 3' untranslated regions (UTRs) of IBDV dsRNA were essential for VP1/VP3-dependent translation initiation. Significantly, VP1 and VP3 could also mediate the recovery of infectious IBDV from the authentic minus-sense strand of IBDV dsRNA. Moreover, downregulation or inhibition of the cap-binding protein eIF4E did not decrease but, rather, enhanced the VP1/VP3-mediated translation of the uncapped IBDV RNA. Collectively, our findings for the first time reveal that VP1 and VP3 compensate for the deficiency of the 5' cap and replace eIF4E to confer upon the uncapped IBDV RNA the ability to be translated and rescued into infectious viruses.IMPORTANCE A key point of control for virus replication is viral translation initiation. The current study shows that the uncapped IBDV RNA cannot be translated into viral proteins directly by host translation machinery and is thus noninfectious. Our results constitute the first direct experimental evidence that VP1 and VP3 are required and sufficient to initiate translation of uncapped IBDV genomic RNA by acting as a substitute for cap and replacing the cap-binding protein eIF4E. Significantly, VP1/VP3 mediate the recovery of infectious IBDV not only from the plus-sense strand but also from the minus-sense strand of the IBDV dsRNA. These findings provide not only new insights into the molecular mechanisms of the life cycle of IBDV but also a new tool for an alternative strategy for the recovery of IBDV from both the plus- and the minus-sense strands of the viral genomic dsRNA.


Subject(s)
Birnaviridae Infections/virology , Infectious bursal disease virus/physiology , RNA, Double-Stranded/genetics , Viral Structural Proteins/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , Animals , Cell Line , Chickens , HEK293 Cells , Humans , Mice , Protein Biosynthesis , Virus Replication
18.
J Immunol ; 199(12): 4142-4154, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29127148

ABSTRACT

Although the immune adaptor adhesion and degranulation-promoting adaptor protein (ADAP) acts as a key mediator of integrin inside-out signaling leading to T cell adhesion, the regulation of this adaptor during integrin activation and clustering remains unclear. We now identify Ubc9, the sole small ubiquitin-related modifier E2 conjugase, as an essential regulator of ADAP where it is required for TCR-induced membrane recruitment of the small GTPase Rap1 and its effector protein RapL and for activation of the small GTPase Rac1 in T cell adhesion. We show that Ubc9 interacted directly with ADAP in vitro and in vivo, and the association was increased in response to anti-CD3 stimulation. The Ubc9-binding domain on ADAP was mapped to a nuclear localization sequence (aa 674-700) within ADAP. Knockdown of Ubc9 by short hairpin RNA or expression of the Ubc9-binding-deficient ADAP mutant significantly decreased TCR-induced integrin adhesion to ICAM-1 and fibronectin, as well as LFA-1 clustering, although it had little effect on the TCR proximal signaling responses and TCR-induced IL-2 transcription. Furthermore, downregulation of Ubc9 impaired TCR-mediated Rac1 activation and attenuated the membrane targeting of Rap1 and RapL, but not Rap1-interacting adaptor molecule. Taken together, our data demonstrate for the first time, to our knowledge, that Ubc9 acts as a functional binding partner of ADAP and plays a selective role in integrin-mediated T cell adhesion via modulation of Rap1-RapL membrane recruitment and Rac1 activation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/metabolism , Monomeric GTP-Binding Proteins/metabolism , T-Lymphocytes/cytology , Ubiquitin-Conjugating Enzymes/physiology , rac1 GTP-Binding Protein/metabolism , rap1 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , COS Cells , Cell Adhesion , Chlorocebus aethiops , Enzyme Activation , Gene Knockdown Techniques , Humans , Integrins/physiology , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Models, Immunological , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , RNA, Small Interfering/pharmacology , Receptors, Antigen, T-Cell/immunology , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/genetics
19.
J Virol ; 91(3)2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27881656

ABSTRACT

While the entry of infectious bursal disease virus (IBDV) is initiated by the binding of the virus to the two major receptors integrin and HSP90, the signaling events after receptor binding and how they contribute to virus entry remain elusive. We show here that IBDV activates c-Src by inducing the phosphorylation of the Y416 residue in c-Src both in DF-1 chicken fibroblasts and in vivo in the bursa of Fabricius from specific-pathogen-free (SPF) chickens. Importantly, inactivated IBDV fails to stimulate c-Src Y416 phosphorylation, and a very virulent IBDV strain induces a much higher level of c-Src Y416 phosphorylation than does an attenuated strain. Inhibition of c-Src activation by an Src kinase inhibitor or expression of a c-Src dominant negative mutant results in a significant decrease in the internalization of IBDV but has little effect on virus adhesion. Furthermore, short hairpin RNA (shRNA) downregulation of integrin, either the α4 or ß1 subunit, but not HSP90 remarkably attenuates IBDV-induced c-Src Y416 phosphorylation, resulting in a decrease in IBDV internalization but not virus adhesion. Moreover, interestingly, inhibition of either c-Src downstream of the phosphatidylinositol 3-kinase (PI3K)/Akt-RhoA signaling cascade or actin rearrangement leads to a significant decrease in IBDV internalization irrespective of the IBDV-induced high levels of c-Src phosphorylation. Cumulatively, our results suggest a novel feed-forward model whereby IBDV activates c-Src for benefiting its cell entry via an integrin-mediated pathway by the activation of downstream PI3K/Akt-RhoA signaling and cytoskeleton actin rearrangement. IMPORTANCE: While IBDV-caused immunosuppression is highly related to viral invasion, the molecular basis of the cellular entry of IBDV remains elusive. In this study, we demonstrate that IBDV activates c-Src by inducing the phosphorylation of the Y416 residue in c-Src to promote virus internalization but not virus adhesion. The ability to induce the level of c-Src Y416 phosphorylation correlates with the pathogenicity of an IBDV strain. IBDV-induced c-Src Y416 activation is α4ß1 integrin but not HSP90 dependent and involves the activation of the downstream PI3K/Akt-RhoA GTPase-actin rearrangement cascade. Thus, our findings provide new insights into the IBDV infection process and the potential for c-Src as a candidate target for the development of IBDV therapeutic drugs.


Subject(s)
Birnaviridae Infections/metabolism , Birnaviridae Infections/virology , Infectious bursal disease virus/physiology , Integrin alpha4beta1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Virus Internalization , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism , Actins/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Chick Embryo , Chickens , Fibroblasts , HSP90 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Humans , Phosphorylation , Signal Transduction , Virus Attachment
20.
Mol Cell ; 59(5): 840-9, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26321253

ABSTRACT

While immune cell adaptors regulate proximal T cell signaling, direct regulation of the nuclear pore complex (NPC) has not been reported. NPC has cytoplasmic filaments composed of RanGAP1 and RanBP2 with the potential to interact with cytoplasmic mediators. Here, we show that the immune cell adaptor SLP-76 binds directly to SUMO-RanGAP1 of cytoplasmic fibrils of the NPC, and that this interaction is needed for optimal NFATc1 and NF-κB p65 nuclear entry in T cells. Transmission electron microscopy showed anti-SLP-76 cytoplasmic labeling of the majority of NPCs in anti-CD3 activated T cells. Further, SUMO-RanGAP1 bound to the N-terminal lysine 56 of SLP-76 where the interaction was needed for optimal RanGAP1-NPC localization and GAP exchange activity. While the SLP-76-RanGAP1 (K56E) mutant had no effect on proximal signaling, it impaired NF-ATc1 and p65/RelA nuclear entry and in vivo responses to OVA peptide. Overall, we have identified SLP-76 as a direct regulator of nuclear pore function in T cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , GTPase-Activating Proteins/metabolism , Phosphoproteins/immunology , Phosphoproteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line , Humans , Jurkat Cells , Mice , Microscopy, Electron, Transmission , NFATC Transcription Factors/metabolism , Nuclear Pore/metabolism , Protein Binding , T-Lymphocytes/ultrastructure , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...