Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 17(3): 241-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26692175

ABSTRACT

The gene encoding PTEN is one of the most frequently mutated tumor suppressor-encoding genes in human cancer. While PTEN's function in tumor suppression is well established, its relationship to anti-microbial immunity remains unknown. Here we found a pivotal role for PTEN in the induction of type I interferon, the hallmark of antiviral innate immunity, that was independent of the pathway of the kinases PI(3)K and Akt. PTEN controlled the import of IRF3, a master transcription factor responsible for IFN-ß production, into the nucleus. We further identified a PTEN-controlled negative phosphorylation site at Ser97 of IRF3 and found that release from this negative regulation via the phosphatase activity of PTEN was essential for the activation of IRF3 and its import into the nucleus. Our study identifies crosstalk between PTEN and IRF3 in tumor suppression and innate immunity.


Subject(s)
Immunity, Innate/immunology , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , PTEN Phosphohydrolase/immunology , Respirovirus Infections/immunology , Rhabdoviridae Infections/immunology , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Gene Transfer Techniques , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , MCF-7 Cells , Macrophages/immunology , Mass Spectrometry , Mice , Microscopy, Confocal , Mutagenesis, Site-Directed , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus , Vesiculovirus
2.
Proc Natl Acad Sci U S A ; 121(24): e2321991121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838012

ABSTRACT

The endoplasmic reticulum (ER) undergoes degradation by selective macroautophagy (ER-phagy) in response to starvation or the accumulation of misfolded proteins within its lumen. In yeast, actin assembly at sites of contact between the cortical ER (cER) and endocytic pits acts to displace elements of the ER from their association with the plasma membrane (PM) so they can interact with the autophagosome assembly machinery near the vacuole. A collection of proteins tether the cER to the PM. Of these, Scs2/22 and Ist2 are required for cER-phagy, most likely through their roles in lipid transport, while deletion of the tricalbins, TCB1/2/3, bypasses those requirements. An artificial ER-PM tether blocks cER-phagy in both the wild type (WT) and a strain lacking endogenous tethers, supporting the importance of cER displacement from the PM. Scs2 and Ist2 can be cross-linked to the selective cER-phagy receptor, Atg40. The COPII cargo adaptor subunit, Lst1, associates with Atg40 and is required for cER-phagy. This requirement is also bypassed by deletion of the ER-PM tethers, suggesting a role for Lst1 prior to the displacement of the cER from the PM during cER-phagy. Although pexophagy and mitophagy also require actin assembly, deletion of ER-PM tethers does not bypass those requirements. We propose that within the context of rapamycin-induced cER-phagy, Scs2/22, Ist2, and Lst1 promote the local displacement of an element of the cER from the cortex, while Tcb1/2/3 act in opposition, anchoring the cER to the plasma membrane.


Subject(s)
Autophagy , Cell Membrane , Endoplasmic Reticulum , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Endoplasmic Reticulum/metabolism , Autophagy/physiology , Cell Membrane/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics
3.
PLoS Pathog ; 17(3): e1009421, 2021 03.
Article in English | MEDLINE | ID: mdl-33690734

ABSTRACT

N6-methyladenosine (m6A) is a prevalent RNA modification that plays a key role in regulating eukaryotic cellular mRNA functions. RNA m6A modification is regulated by two groups of cellular proteins, writers and erasers that add or remove m6A, respectively. HIV-1 RNA contains m6A modifications that modulate viral infection and gene expression in CD4+ T cells. However, it remains unclear whether m6A modifications of HIV-1 RNA modulate innate immune responses in myeloid cells that are important for antiviral immunity. Here we show that m6A modification of HIV-1 RNA suppresses the expression of antiviral cytokine type-I interferon (IFN-I) in differentiated human monocytic cells and primary monocyte-derived macrophages. Transfection of differentiated monocytic U937 cells with HIV-1 RNA fragments containing a single m6A-modification significantly reduced IFN-I mRNA expression relative to their unmodified RNA counterparts. We generated HIV-1 with altered m6A levels of RNA by manipulating the expression of the m6A erasers (FTO and ALKBH5) or pharmacological inhibition of m6A addition in virus-producing cells, or by treating HIV-1 RNA with recombinant FTO in vitro. HIV-1 RNA transfection or viral infection of differentiated U937 cells and primary macrophages demonstrated that HIV-1 RNA with decreased m6A levels enhanced IFN-I expression, whereas HIV-1 RNA with increased m6A modifications had opposite effects. Our mechanistic studies indicated that m6A of HIV-1 RNA escaped retinoic acid-induced gene I (RIG-I)-mediated RNA sensing and activation of the transcription factors IRF3 and IRF7 that drive IFN-I gene expression. Together, these findings suggest that m6A modifications of HIV-1 RNA evade innate immune sensing in myeloid cells.


Subject(s)
HIV Infections/immunology , HIV-1/metabolism , Interferon Type I/biosynthesis , Myeloid Cells/virology , RNA Processing, Post-Transcriptional/immunology , RNA, Viral/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Gene Expression Regulation/immunology , HIV-1/immunology , Humans , Immunity, Innate/immunology , Macrophages/metabolism , Macrophages/virology , Monocytes/metabolism , Monocytes/virology , Myeloid Cells/immunology , Myeloid Cells/metabolism , RNA, Viral/immunology
4.
Proc Natl Acad Sci U S A ; 117(31): 18530-18539, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32690699

ABSTRACT

Endoplasmic reticulum (ER) macroautophagy (hereafter called ER-phagy) uses autophagy receptors to selectively degrade ER domains in response to starvation or the accumulation of aggregation-prone proteins. Autophagy receptors package the ER into autophagosomes by binding to the ubiquitin-like yeast protein Atg8 (LC3 in mammals), which is needed for autophagosome formation. In budding yeast, cortical and cytoplasmic ER-phagy requires the autophagy receptor Atg40. While different ER autophagy receptors have been identified, little is known about other components of the ER-phagy machinery. In an effort to identify these components, we screened the genome-wide library of viable yeast deletion mutants for defects in the degradation of cortical ER following treatment with rapamycin, a drug that mimics starvation. Among the mutants we identified was vps13Δ. While yeast has one gene that encodes the phospholipid transporter VPS13, humans have four vacuolar protein-sorting (VPS) protein 13 isoforms. Mutations in all four human isoforms have been linked to different neurological disorders, including Parkinson's disease. Our findings have shown that Vps13 acts after Atg40 engages the autophagy machinery. Vps13 resides at contact sites between the ER and several organelles, including late endosomes. In the absence of Vps13, the cortical ER marker Rtn1 accumulated at late endosomes, and a dramatic decrease in ER packaging into autophagosomes was observed. Together, these studies suggest a role for Vps13 in the sequestration of the ER into autophagosomes at late endosomes. These observations may have important implications for understanding Parkinson's and other neurological diseases.


Subject(s)
Autophagosomes/metabolism , Endoplasmic Reticulum/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Autophagy , Cell Line , Endoplasmic Reticulum/genetics , Endosomes/genetics , Endosomes/metabolism , Humans , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 115(27): E6237-E6244, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29915089

ABSTRACT

The endoplasmic reticulum (ER) forms a contiguous network of tubules and sheets that is predominantly associated with the cell cortex in yeast. Upon treatment with rapamycin, the ER undergoes degradation by selective autophagy. This process, termed ER-phagy, requires Atg40, a selective autophagy receptor that localizes to the cortical ER. Here we report that ER-phagy also requires Lnp1, an ER membrane protein that normally resides at the three-way junctions of the ER network, where it serves to stabilize the network as it is continually remodeled. Rapamycin treatment increases the expression of Atg40, driving ER domains marked by Atg40 puncta to associate with Atg11, a scaffold protein needed to form autophagosomes. Although Atg40 largely localizes to the cortical ER, the autophagy machinery resides in the cell interior. The localization of Atg40 to sites of autophagosome formation is blocked in an lnp1Δ mutant or upon treatment of wild-type cells with the actin-depolymerizing drug Latrunculin A. This prevents the association of Atg40 with Atg11 and the packaging of the ER into autophagosomes. We propose that Lnp1 is needed to stabilize the actin-dependent remodeling of the ER that is essential for ER-phagy.


Subject(s)
Autophagosomes/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Endoplasmic Reticulum/genetics , Membrane Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Thiazolidines/pharmacology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 115(16): E3798-E3807, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610295

ABSTRACT

Sterile alpha motif and HD-domain-containing protein 1 (SAMHD1) blocks replication of retroviruses and certain DNA viruses by reducing the intracellular dNTP pool. SAMHD1 has been suggested to down-regulate IFN and inflammatory responses to viral infections, although the functions and mechanisms of SAMHD1 in modulating innate immunity remain unclear. Here, we show that SAMHD1 suppresses the innate immune responses to viral infections and inflammatory stimuli by inhibiting nuclear factor-κB (NF-κB) activation and type I interferon (IFN-I) induction. Compared with control cells, infection of SAMHD1-silenced human monocytic cells or primary macrophages with Sendai virus (SeV) or HIV-1, or treatment with inflammatory stimuli, induces significantly higher levels of NF-κB activation and IFN-I induction. Exogenous SAMHD1 expression in cells or SAMHD1 reconstitution in knockout cells suppresses NF-κB activation and IFN-I induction by SeV infection or inflammatory stimuli. Mechanistically, SAMHD1 inhibits NF-κB activation by interacting with NF-κB1/2 and reducing phosphorylation of the NF-κB inhibitory protein IκBα. SAMHD1 also interacts with the inhibitor-κB kinase ε (IKKε) and IFN regulatory factor 7 (IRF7), leading to the suppression of the IFN-I induction pathway by reducing IKKε-mediated IRF7 phosphorylation. Interactions of endogenous SAMHD1 with NF-κB and IFN-I pathway proteins were validated in human monocytic cells and primary macrophages. Comparing splenocytes from SAMHD1 knockout and heterozygous mice, we further confirmed SAMHD1-mediated suppression of NF-κB activation, suggesting an evolutionarily conserved property of SAMHD1. Our findings reveal functions of SAMHD1 in down-regulating innate immune responses to viral infections and inflammatory stimuli, highlighting the importance of SAMHD1 in modulating antiviral immunity.


Subject(s)
Immunity, Innate , Inflammation/immunology , Interferon-alpha/biosynthesis , NF-kappa B/metabolism , SAM Domain and HD Domain-Containing Protein 1/physiology , Virus Diseases/immunology , Animals , Cells, Cultured , Down-Regulation , Gene Expression Regulation/drug effects , Gene Silencing , HEK293 Cells , HIV/physiology , Humans , I-kappa B Kinase/antagonists & inhibitors , Interferon Regulatory Factor-7/antagonists & inhibitors , Interferon-alpha/genetics , Macrophages/immunology , Macrophages/virology , Male , Mice , NF-KappaB Inhibitor alpha/metabolism , Phosphorylation , Protein Processing, Post-Translational , Recombinant Proteins/immunology , Sendai virus/physiology , Signal Transduction/immunology , THP-1 Cells
7.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30651369

ABSTRACT

HIV-1 enters cells through binding between viral envelope glycoprotein (Env) and cellular receptors to initiate virus and cell fusion. HIV-1 Env precursor (gp160) is cleaved into two units noncovalently bound to form a trimer on virions, including a surface unit (gp120) and a transmembrane unit (gp41) responsible for virus binding and membrane fusion, respectively. The polar region (PR) at the N terminus of gp41 comprises 17 residues, including 7 polar amino acids. Previous studies suggested that the PR contributes to HIV-1 membrane fusion and infectivity; however, the precise role of the PR in Env-mediated viral entry and the underlying mechanisms remain unknown. Here, we show that the PR is critical for HIV-1 fusion and infectivity by stabilizing Env trimers. Through analyzing the PR sequences of 57,645 HIV-1 isolates, we performed targeted mutagenesis and functional studies of three highly conserved polar residues in the PR (S532P, T534A, and T536A) which have not been characterized previously. We found that single or combined mutations of these three residues abolished or significantly decreased HIV-1 infectivity without affecting viral production. These PR mutations abolished or significantly reduced HIV-1 fusion with target cells and also Env-mediated cell-cell fusion. Three PR mutations containing S532P substantially reduced gp120 and gp41 association, Env trimer stability, and increased gp120 shedding. Furthermore, S532A mutation significantly reduced HIV-1 infectivity and fusogenicity but not Env expression and cleavage. Our findings suggest that the PR of gp41, particularly the key residue S532, is structurally essential for maintaining HIV-1 Env trimer, viral fusogenicity, and infectivity.IMPORTANCE Although extensive studies of the transmembrane unit (gp41) of HIV-1 Env have led to a fusion inhibitor clinically used to block viral entry, the functions of different domains of gp41 in HIV-1 fusion and infectivity are not fully elucidated. The polar region (PR) of gp41 has been proposed to participate in HIV-1 membrane fusion in biochemical analyses, but its role in viral entry and infectivity remain unclear. In our effort to characterize three nucleotide mutations of an HIV-1 RNA element that partially overlaps the PR coding sequence, we identified a novel function of the PR that determines viral fusion and infectivity. We further demonstrated the structural and functional impact of six PR mutations on HIV-1 Env stability, viral fusion, and infectivity. Our findings reveal the previously unappreciated function of the PR and the underlying mechanisms, highlighting the important role of the PR in regulating HIV-1 fusion and infectivity.


Subject(s)
HIV Envelope Protein gp120/metabolism , HIV Envelope Protein gp41/metabolism , HIV Infections/virology , HIV-1/metabolism , HIV-1/physiology , Cell Line , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans , Virion/metabolism , Virion/physiology , Virus Internalization , env Gene Products, Human Immunodeficiency Virus/metabolism
8.
Genes Immun ; 20(4): 327-337, 2019 04.
Article in English | MEDLINE | ID: mdl-29961753

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) causes persistent infection in human and induces miR-146a expression in infected cells. miR-146a represses the innate immune response by inhibiting the expression of TRAF6 and IRAK1 genes, thus negatively controls the NF-κB-related cytokines and interferon stimulated genes. Here we reported that lentiviral CRISPR/Cas9 system was highly efficient in introducing mutations in the precursor miR-146a genomic sequences, resulting in a loss of miR-146a expression and function. miR-146a ablation led to increasing cytokines production in LPS-stimulated A549 cells. Moreover, miR-146a knockout in HIV-1 infected MT2 cells markedly increased the expression of cytokines and HIV-1 restriction factors and reversed T cell exhaustion markers expression, thus influencing HIV-1 replication. Our study indicates that lentiviral CRISPR/Cas9-mediated gene editing is an effective approach to abrogate miR-146a expression, which consequently inhibits HIV-1 replication as well as proviral reactivation by enhancing the expression of cytokines and HIV-1 restriction factors.


Subject(s)
Gene Deletion , HIV-1/physiology , MicroRNAs/genetics , Virus Replication , CRISPR-Cas Systems , Cell Line, Tumor , Cytokines/metabolism , HEK293 Cells , Host-Pathogen Interactions , Humans
9.
Retrovirology ; 16(1): 15, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31186067

ABSTRACT

BACKGROUND: The chemokine receptor CCR5, which belongs to the superfamily of G protein-coupled receptors, is the major co-receptor for HIV-1 entry. Individuals with a homozygous CCR5Δ32 mutation have a long lasting and increased resistance to HIV-1 infection. Therefore, CCR5 represents an optimal target for HIV-1/AIDS gene therapy. The CRISPR/Cas9 system has been developed as one of the most efficacious gene editing tools in mammalian cells and the small-sized version from Staphylococcus aureus (SaCas9) has an advantage of easier delivery compared to the most commonly used version from Streptococcus pyogenes Cas9 (SpCas9). RESULTS: Here, we demonstrated that CCR5 could be specifically and efficiently edited by CRISPR/SaCas9 together with two sgRNAs, which were identified through a screening of 13 sgRNAs. Disruption of CCR5 expression by lentiviral vector-mediated CRISPR/SaCas9 led to increased resistance against HIV-1 infection in human primary CD4+ T cells. Moreover, humanized mice engrafted with CCR5-disrupted CD4+ T cells showed selective survival and enrichment when challenged with CCR5 (R5)-tropic HIV-1 in comparison to mock-treated CD4+ T cells. We also observed CCR5 could be targeted by CRISPR/SaCas9 in human CD34+ hematopoietic stem/progenitor cells without obvious differentiation deficiencies. CONCLUSIONS: This work provides an alternative approach to disrupt human CCR5 by CRISPR/SaCas9 for a potential gene therapy strategy against HIV-1/AIDS.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Gene Editing , Hematopoietic Stem Cells/cytology , Receptors, CCR5/genetics , Animals , CRISPR-Associated Protein 9 , Cells, Cultured , HIV Infections/virology , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , RNA, Guide, Kinetoplastida , Staphylococcus aureus/enzymology
10.
Retrovirology ; 16(1): 20, 2019 Jul 23.
Article in English | MEDLINE | ID: mdl-31337409

ABSTRACT

Following publication of their article [1], the authors realized that they inadvertently omitted the contribution of Dr. Li Wu (Ohio State University) who commented on the manuscript at the early stage of the manuscript preparation and provided one plasmid related to this work.

11.
J Cell Biochem ; 119(4): 3030-3043, 2018 04.
Article in English | MEDLINE | ID: mdl-29058807

ABSTRACT

ABIN1, an important immune regulator, has been shown to be involved in various cellular functions, such as immunity, development, tissue homeostasis, and tumor progression. It inhibits TNF- and TLR-induced NF-κB signaling activation and the consequent gene expression. Despite its functional significance, the mechanism of ABIN1 in the regulation of various cellular functions remains unclear. In this study, we identified HDAC1, a key regulator of eukaryotic gene expression and many important cellular events, including cell proliferation, differentiation, cancer and immunity, as an interacting partner of ABIN1. The results showed that ABIN1 acted as a modulator to down-regulate HDAC1 ubiquitination via three different linkages, thereby stabilizing HDAC1 by inhibiting its lysosomal and proteasomal degradation. Interestingly, the inhibitory function of ABIN1 required direct binding with HDAC1. Moreover, the level of p53, which was a tumor suppressor and a well-studied substrate of HDAC1, was under the regulation of ABIN1 via the modulation of HDAC1 levels, suggesting that ABIN1 was physiologically significant in tumor progression. This study has revealed a new function of ABIN1 in mediating HDAC1 modification and stability.


Subject(s)
DNA-Binding Proteins/metabolism , Histone Deacetylase 1/metabolism , Muramidase/metabolism , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Tumor Suppressor Protein p53/metabolism , A549 Cells , Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , HeLa Cells , Hep G2 Cells , Histone Deacetylase 1/chemistry , Humans , K562 Cells , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Protein Stability , Ubiquitination
12.
Proc Natl Acad Sci U S A ; 112(2): 418-23, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25548161

ABSTRACT

The endoplasmic reticulum (ER) consists of a polygonal network of sheets and tubules interconnected by three-way junctions. This network undergoes continual remodeling through competing processes: the branching and fusion of tubules forms new three-way junctions and new polygons, and junction sliding and ring closure leads to polygon loss. However, little is known about the machinery required to generate and maintain junctions. We previously reported that yeast Lnp1 localizes to ER junctions, and that loss of Lnp1 leads to a collapsed, densely reticulated ER network. In mammalian cells, only approximately half the junctions contain Lnp1. Here we use live cell imaging to show that mammalian Lnp1 (mLnp1) affects ER junction mobility and hence network dynamics. Three-way junctions with mLnp1 are less mobile than junctions without mLnp1. Newly formed junctions that acquire mLnp1 remain stable within the ER network, whereas nascent junctions that fail to acquire mLnp1 undergo rapid ring closure. These findings imply that mLnp1 plays a key role in stabilizing nascent three-way ER junctions.


Subject(s)
Endoplasmic Reticulum/metabolism , Homeodomain Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteolipids/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Cell Analysis
13.
Retrovirology ; 14(1): 12, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28193275

ABSTRACT

BACKGROUND: A20-binding inhibitor of NF-κB activation (ABIN1), an important immune regulator, was previously shown to be involved in HIV-1 replication. However, the reported studies done with overexpressed ABIN1 provided controversial results. RESULTS: Here we identified ABIN1 as a suppressor of HIV-1 transcription since transient knockdown of ABIN1 led to increased HIV-1 replication both in transformed Jurkat T cell line and in primary human CD4+ T lymphocytes. Depletion of ABIN1 specifically enhanced the HIV-1 transcription from the integrated genome during viral life cycle, but not the earlier steps such as reverse transcription or integration. Immunoprecipitation assays revealed that ABIN1 specifically inhibits the proto-oncogene HDM2 catalyzed K63-linked polyubiquitination of Tat at Lys71, which is critical for the transactivation activity of Tat. The ubiquitin chain binding activity of ABIN1 carried by UBAN domain turned out to be essential for the inhibitory role of ABIN1. The results of immunofluorescence localization experiments suggested that ABIN1 may obstruct Tat ubiquitination by redistributing some of HDM2 from the nucleus to the cytoplasm. CONCLUSIONS: Our findings have revealed ABIN1 as intrinsic suppressor of HIV-1 mRNA transcription by regulating the ubiquitination of Tat.


Subject(s)
DNA-Binding Proteins/metabolism , HIV-1/immunology , HIV-1/physiology , Host-Pathogen Interactions , Transcription, Genetic , tat Gene Products, Human Immunodeficiency Virus/antagonists & inhibitors , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Knockdown Techniques , HIV-1/genetics , Humans , Proto-Oncogene Mas , Ubiquitination
14.
Retrovirology ; 14(1): 51, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141633

ABSTRACT

BACKGROUND: The CRISPR/Cas9 system has been widely used for genome editing in mammalian cells. CXCR4 is a co-receptor for human immunodeficiency virus type 1 (HIV-1) entry, and loss of CXCR4 function can protect cells from CXCR4 (X4)-tropic HIV-1 infection, making CXCR4 an important target for HIV-1 gene therapy. However, the large size of the CRISPR/SpCas9 system presents an obstacle to its efficient delivery into primary CD4+ T cells. Recently, a small Staphylococcus aureus Cas9 (SaCas9) has been developed as a genome editing tool can address this question. Therefore, it provides a promising strategy for HIV-1 gene therapy if it is used to target CXCR4. RESULTS: Here, we employed a short version of Cas9 from Staphylococcus aureus (SaCas9) for targeting CXCR4. We demonstrated that transduction of lenti-virus expressing SaCas9 and selected single-guided RNAs of CXCR4 in human CD4+ T cell lines efficiently induced the editing of the CXCR4 gene, making these cell lines resistant to X4-tropic HIV-1 infection. Moreover, we efficiently transduced primary human CD4+ T cells using adeno-associated virus-delivered CRISPR/SaCas9 and disrupted CXCR4 expression. We also showed that CXCR4-edited primary CD4+ T cells proliferated normally and were resistant to HIV-1 infection. CONCLUSIONS: Our study provides a basis for possible application of CXCR4-targeted genome editing by CRISPR/SaCas9 in HIV-1 gene therapy.


Subject(s)
CD4-Positive T-Lymphocytes/virology , CRISPR-Cas Systems/genetics , Disease Resistance/genetics , Gene Editing/methods , HIV Infections/genetics , Receptors, CXCR4/genetics , Staphylococcus aureus/enzymology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Endonucleases/metabolism , Gene Expression Regulation , Gene Knockout Techniques , HEK293 Cells , HIV Infections/metabolism , HIV Infections/virology , HIV-1 , Host-Pathogen Interactions/genetics , Humans , Jurkat Cells , Receptors, CXCR4/metabolism
15.
Biochem Biophys Res Commun ; 470(3): 697-703, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26774344

ABSTRACT

The nonstructural protein 5B (NS5B) of hepatitis C virus (HCV) is an RNA-dependent RNA polymerase (RdRp) and responsible for replicating the whole HCV genome with help of viral and cellular proteins. However, how cellular factors influence NS5B and, in turn, regulating HCV replication are still poorly defined. The well known tumor suppressor Fbw7, a component of E3 ubiquitin ligase SCF(Fbw7), targets oncoproteins or cellular regulatory proteins for ubiquitin-mediated degradation through a highly conserved binding site called a Cdc4 phosphodegron (CPD). But little is known about whether Fbw7 plays a role in regulation of viral proteins. In this study, we revealed that the conserved CPD is shared by NS5B of almost all genotype of HCV and our data demonstrated that NS5B is a bona fide substrate of Fbw7. Forced expression of Fbw7 promoted the ubiquination of NS5B and negatively regulated its turnover in the proteasome-dependent manner. We further revealed the interaction between NS5B and Fbw7, which resulted in the relocation of Fbw7 from nucleus to cytoplasm. During HCV replication, ectopic expression of Fbw7 could strongly down-regulate NS5B level and consequently inhibited the virus replication. When endogenous Fbw7 was knocked down, both NS5B protein abundance and HCV replication were remarkably up-regulated. The results provide more insights into the interplay of HCV and cellular factors and shed light on molecular mechanisms of HCV replication and pathogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Hepacivirus/physiology , Hepatocytes/virology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Cell Line , F-Box-WD Repeat-Containing Protein 7 , Hepatocytes/metabolism , Humans
16.
J Biol Chem ; 289(6): 3114-25, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24318983

ABSTRACT

Cardiolipin (CL) that is synthesized de novo is deacylated to monolysocardiolipin (MLCL), which is reacylated by tafazzin. Remodeled CL contains mostly unsaturated fatty acids. In eukaryotes, loss of tafazzin leads to growth and respiration defects, and in humans, this results in the life-threatening disorder Barth syndrome. Tafazzin deficiency causes a decrease in the CL/MLCL ratio and decreased unsaturated CL species. Which of these biochemical outcomes contributes to the physiological defects is not known. Yeast cells have a single CL-specific phospholipase, Cld1, that can be exploited to distinguish between these outcomes. The cld1Δ mutant has decreased unsaturated CL, but the CL/MLCL ratio is similar to that of wild type cells. We show that cld1Δ rescues growth, life span, and respiratory defects of the taz1Δ mutant. This suggests that defective growth and respiration in tafazzin-deficient cells are caused by the decreased CL/MLCL ratio and not by a deficiency in unsaturated CL. CLD1 expression is increased during respiratory growth and regulated by the heme activator protein transcriptional activation complex. Overexpression of CLD1 leads to decreased mitochondrial respiration and growth and instability of mitochondrial DNA. However, ATP concentrations are maintained by increasing glycolysis. We conclude that transcriptional regulation of Cld1-mediated deacylation of CL influences energy metabolism by modulating the relative contribution of glycolysis and respiration.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Barth Syndrome , Energy Metabolism/physiology , Oxygen Consumption/physiology , Phospholipases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Cardiolipins/genetics , Cardiolipins/metabolism , DNA, Fungal/genetics , DNA, Fungal/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Gene Deletion , Humans , Mitochondria/genetics , Mitochondria/metabolism , Phospholipases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
17.
J Gen Virol ; 96(8): 2252-2261, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25904148

ABSTRACT

Hepatitis B virus (HBV) remains a global health threat as chronic HBV infection may lead to liver cirrhosis or cancer. Current antiviral therapies with nucleoside analogues can inhibit the replication of HBV, but do not disrupt the already existing HBV covalently closed circular DNA. The newly developed CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated 9) system is a powerful tool to target cellular genome DNA for gene editing. In order to investigate the possibility of using the CRISPR/Cas9 system to disrupt the HBV DNA templates, we designed eight guide RNAs (gRNAs) that targeted the conserved regions of different HBV genotypes, which could significantly inhibit HBV replication both in vitro and in vivo. Moreover, the HBV-specific gRNA/Cas9 system could inhibit the replication of HBV of different genotypes in cells, and the viral DNA was significantly reduced by a single gRNA/Cas9 system and cleared by a combination of different gRNA/Cas9 systems.


Subject(s)
CRISPR-Cas Systems , Gene Targeting , Genome, Viral , Hepatitis B virus/genetics , Hepatitis B/virology , Animals , Base Sequence , Clustered Regularly Interspaced Short Palindromic Repeats , Conserved Sequence , Gene Targeting/methods , Hepatitis B virus/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Virus Replication
18.
Biochem Biophys Res Commun ; 459(2): 270-276, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25732088

ABSTRACT

Eukaryotic cellular and most viral RNAs carry a 5'-terminal cap structure, a 5'-5' triphosphate linkage between the 5' end of the RNA and a guanosine nucleotide (cap-0). SARS coronavirus (SARS-CoV) nonstructural protein nsp16 functions as a methyltransferase, to methylate mRNA cap-0 structure at the ribose 2'-O position of the first nucleotide to form cap-1 structures. However, whether there is interplay between nsp16 and host proteins was not yet clear. In this report, we identified several potential cellular nsp16-interacting proteins from a human thymus cDNA library by yeast two-hybrid screening. VHL, one of these proteins, was proven to interact with nsp16 both in vitro and in vivo. Further studies showed that VHL can inhibit SARS-CoV replication by regulating nsp16 ubiquitination and promoting its degradation. Our results have revealed the role of cellular VHL in the regulation of SARS-CoV replication.


Subject(s)
Methyltransferases/metabolism , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Chlorocebus aethiops , Exoribonucleases/genetics , Exoribonucleases/metabolism , Host-Pathogen Interactions/physiology , Humans , Methyltransferases/chemistry , Methyltransferases/genetics , Protein Stability , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Two-Hybrid System Techniques , Ubiquitination , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics
19.
Biol Cell ; 105(5): 208-218, 2013 May.
Article in English | MEDLINE | ID: mdl-23387829

ABSTRACT

BACKGROUND INFORMATION: The translationally controlled tumour protein (TCTP) plays an important role in maintaining cell proliferation and its high expression is associated with many tumours. The tumour suppressor von Hippel-Lindau protein (VHL) has been shown to function as an E3 ubiquitin ligase. Although great progress has been made, biological roles of these factors and relevant molecular mechanisms remain largely unknown. RESULTS: In this study, we have shown that TCTP specifically binds to VHL through its ß domain and competes with hypoxia-inducible factor-1α (HIF1α). TCTP over-expression decreased the protein level of VHL and the inhibition of TCTP expression by miRNA resulted in an increase of the VHL protein level. Moreover, TCTP over-expression promoted the K48-linked ubiquitination of VHL, thus degradation through the ubiquitin-proteasome pathway. In addition, we showed that TCTP increased the protein level of HIF1α, which promoted both vascular endothelial growth factor-hypoxic response element-promoter-driven luciferase reporter and endogenous VEGF expression. CONCLUSIONS: These data have demonstrated that TCTP binds to the ß domain of VHL through competition with HIF1α, which promotes VHL degradation by the ubiquitin-proteasome system and HIF1α stability.


Subject(s)
Biomarkers, Tumor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Binding, Competitive , COS Cells , Chlorocebus aethiops , Gene Expression , HEK293 Cells , Humans , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Stability , Proteolysis , Transcriptional Activation , Tumor Protein, Translationally-Controlled 1 , Ubiquitination , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/chemistry
20.
Biochem J ; 456(1): 55-66, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23961993

ABSTRACT

Loss and/or inactivation of the VHL (von Hippel-Lindau) tumour suppressor causes various tumours. Using a yeast two-hybrid system, we have identified the AR (androgen receptor) co-activator UXT (ubiquitously expressed transcript), as a VHL-interacting protein. GST pull-down and co-immunoprecipitation assays show that UXT interacts with VHL. In addition, UXT recruits VHL to the nucleus. VHL associates with the DBD (DNA-binding domain) and hinge domains of the AR and induces AR ubiquitination. Moreover, VHL interaction with the AR activates AR transactivation upon DHT (dihydrotestosterone) treatment. VHL knockdown inhibits AR ubiquitination and decreases transcriptional activation of the AR. Our data suggest that the VHL-UXT interaction and VHL-induced ubiquitination of AR regulate transcriptional activation of the AR.


Subject(s)
Neoplasm Proteins/metabolism , Receptors, Androgen/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Active Transport, Cell Nucleus , Animals , COS Cells , Cell Cycle Proteins , Cell Nucleus/metabolism , Chlorocebus aethiops , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Mice , Molecular Chaperones , Receptors, Androgen/metabolism , Transcriptional Activation , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL