Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Proc Natl Acad Sci U S A ; 120(14): e2218823120, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36996106

ABSTRACT

Myelin is a multilayered membrane that tightly wraps neuronal axons, enabling efficient, high-speed signal propagation. The axon and myelin sheath form tight contacts, mediated by specific plasma membrane proteins and lipids, and disruption of these contacts causes devastating demyelinating diseases. Using two cell-based models of demyelinating sphingolipidoses, we demonstrate that altered lipid metabolism changes the abundance of specific plasma membrane proteins. These altered membrane proteins have known roles in cell adhesion and signaling, with several implicated in neurological diseases. The cell surface abundance of the adhesion molecule neurofascin (NFASC), a protein critical for the maintenance of myelin-axon contacts, changes following disruption to sphingolipid metabolism. This provides a direct molecular link between altered lipid abundance and myelin stability. We show that the NFASC isoform NF155, but not NF186, interacts directly and specifically with the sphingolipid sulfatide via multiple binding sites and that this interaction requires the full-length extracellular domain of NF155. We demonstrate that NF155 adopts an S-shaped conformation and preferentially binds sulfatide-containing membranes in cis, with important implications for protein arrangement in the tight axon-myelin space. Our work links glycosphingolipid imbalances to disturbance of membrane protein abundance and demonstrates how this may be driven by direct protein-lipid interactions, providing a mechanistic framework to understand the pathogenesis of galactosphingolipidoses.


Subject(s)
Demyelinating Diseases , Sulfoglycosphingolipids , Humans , Glycosphingolipids/metabolism , Carrier Proteins/metabolism , Nerve Growth Factors/metabolism , Myelin Sheath/metabolism , Cell Adhesion Molecules/metabolism , Demyelinating Diseases/pathology
2.
Wellcome Open Res ; 7: 224, 2022.
Article in English | MEDLINE | ID: mdl-36483314

ABSTRACT

Background: Quantitative proteomics is able to provide a comprehensive, unbiased description of changes to cells caused by viral infection, but interpretation may be complicated by differential changes in infected and uninfected 'bystander' cells, or the use of non-physiological cellular models. Methods: In this paper, we use fluorescence-activated cell sorting (FACS) and quantitative proteomics to analyse cell-autonomous changes caused by authentic SARS-CoV-2 infection of respiratory epithelial cells, the main target of viral infection in vivo. First, we determine the relative abundance of proteins in primary human airway epithelial cells differentiated at the air-liquid interface (basal, secretory and ciliated cells). Next, we specifically characterise changes caused by SARS-CoV-2 infection of ciliated cells. Finally, we compare temporal proteomic changes in infected and uninfected 'bystander' Calu-3 lung epithelial cells and compare infection with B.29 and B.1.1.7 (Alpha) variants. Results: Amongst 5,709 quantified proteins in primary human airway ciliated cells, the abundance of 226 changed significantly in the presence of SARS-CoV-2 infection (q <0.05 and >1.5-fold). Notably, viral replication proceeded without inducing a type-I interferon response. Amongst 6,996 quantified proteins in Calu-3 cells, the abundance of 645 proteins changed significantly in the presence of SARS-CoV-2 infection (q < 0.05 and > 1.5-fold). In contrast to the primary cell model, a clear type I interferon (IFN) response was observed. Nonetheless, induction of IFN-inducible proteins was markedly attenuated in infected cells, compared with uninfected 'bystander' cells. Infection with B.29 and B.1.1.7 (Alpha) variants gave similar results. Conclusions: Taken together, our data provide a detailed proteomic map of changes in SARS-CoV-2-infected respiratory epithelial cells in two widely used, physiologically relevant models of infection. As well as identifying dysregulated cellular proteins and processes, the effectiveness of strategies employed by SARS-CoV-2 to avoid the type I IFN response is illustrated in both models.

3.
Elife ; 112022 11 24.
Article in English | MEDLINE | ID: mdl-36421765

ABSTRACT

EROS (essential for reactive oxygen species) protein is indispensable for expression of gp91phox, the catalytic core of the phagocyte NADPH oxidase. EROS deficiency in humans is a novel cause of the severe immunodeficiency, chronic granulomatous disease, but its mechanism of action was unknown until now. We elucidate the role of EROS, showing it acts at the earliest stages of gp91phox maturation. It binds the immature 58 kDa gp91phox directly, preventing gp91phox degradation and allowing glycosylation via the oligosaccharyltransferase machinery and the incorporation of the heme prosthetic groups essential for catalysis. EROS also regulates the purine receptors P2X7 and P2X1 through direct interactions, and P2X7 is almost absent in EROS-deficient mouse and human primary cells. Accordingly, lack of murine EROS results in markedly abnormal P2X7 signalling, inflammasome activation, and T cell responses. The loss of both ROS and P2X7 signalling leads to resistance to influenza infection in mice. Our work identifies EROS as a highly selective chaperone for key proteins in innate and adaptive immunity and a rheostat for immunity to infection. It has profound implications for our understanding of immune physiology, ROS dysregulation, and possibly gene therapy.


Subject(s)
Granulomatous Disease, Chronic , NADPH Oxidases , Humans , Animals , Mice , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Phagocytes/metabolism , Signal Transduction/physiology
4.
EMBO J ; 41(19): e110777, 2022 10 04.
Article in English | MEDLINE | ID: mdl-35993436

ABSTRACT

The regulation of membrane lipid composition is critical for cellular homeostasis. Cells are particularly sensitive to phospholipid saturation, with increased saturation causing membrane rigidification and lipotoxicity. How mammalian cells sense membrane lipid composition and reverse fatty acid (FA)-induced membrane rigidification is poorly understood. Here we systematically identify proteins that differ between mammalian cells fed saturated versus unsaturated FAs. The most differentially expressed proteins were two ER-resident polytopic membrane proteins: the E3 ubiquitin ligase RNF145 and the lipid hydrolase ADIPOR2. In unsaturated lipid membranes, RNF145 is stable, promoting its lipid-sensitive interaction, ubiquitination and degradation of ADIPOR2. When membranes become enriched in saturated FAs, RNF145 is rapidly auto-ubiquitinated and degraded, stabilising ADIPOR2, whose hydrolase activity restores lipid homeostasis and prevents lipotoxicity. We therefore identify RNF145 as a FA-responsive ubiquitin ligase which, together with ADIPOR2, defines an autoregulatory pathway that controls cellular membrane lipid homeostasis and prevents acute lipotoxic stress.


Subject(s)
Hydrolases , Membrane Fluidity , Animals , Fatty Acids/metabolism , Hydrolases/metabolism , Mammals , Membrane Proteins/metabolism , Phospholipids , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
5.
Elife ; 112022 05 19.
Article in English | MEDLINE | ID: mdl-35587364

ABSTRACT

The outcome of infection is dependent on the ability of viruses to manipulate the infected cell to evade immunity, and the ability of the immune response to overcome this evasion. Understanding this process is key to understanding pathogenesis, genetic risk factors, and both natural and vaccine-induced immunity. SARS-CoV-2 antagonises the innate interferon response, but whether it manipulates innate cellular immunity is unclear. An unbiased proteomic analysis determined how cell surface protein expression is altered on SARS-CoV-2-infected lung epithelial cells, showing downregulation of activating NK ligands B7-H6, MICA, ULBP2, and Nectin1, with minimal effects on MHC-I. This occurred at the level of protein synthesis, could be mediated by Nsp1 and Nsp14, and correlated with a reduction in NK cell activation. This identifies a novel mechanism by which SARS-CoV-2 host-shutoff antagonises innate immunity. Later in the disease process, strong antibody-dependent NK cell activation (ADNKA) developed. These responses were sustained for at least 6 months in most patients, and led to high levels of pro-inflammatory cytokine production. Depletion of spike-specific antibodies confirmed their dominant role in neutralisation, but these antibodies played only a minor role in ADNKA compared to antibodies to other proteins, including ORF3a, Membrane, and Nucleocapsid. In contrast, ADNKA induced following vaccination was focussed solely on spike, was weaker than ADNKA following natural infection, and was not boosted by the second dose. These insights have important implications for understanding disease progression, vaccine efficacy, and vaccine design.


Subject(s)
COVID-19 , SARS-CoV-2 , Antibodies , Antibodies, Viral , Humans , Killer Cells, Natural , Proteomics
6.
Blood ; 139(16): 2471-2482, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35134130

ABSTRACT

The accessibility of cell surface proteins makes them tractable for targeting by cancer immunotherapy, but identifying suitable targets remains challenging. Here we describe plasma membrane profiling of primary human myeloma cells to identify an unprecedented number of cell surface proteins of a primary cancer. We used a novel approach to prioritize immunotherapy targets and identified a cell surface protein not previously implicated in myeloma, semaphorin-4A (SEMA4A). Using knock-down by short-hairpin RNA and CRISPR/nuclease-dead Cas9 (dCas9), we show that expression of SEMA4A is essential for normal myeloma cell growth in vitro, indicating that myeloma cells cannot downregulate the protein to avoid detection. We further show that SEMA4A would not be identified as a myeloma therapeutic target by standard CRISPR/Cas9 knockout screens because of exon skipping. Finally, we potently and selectively targeted SEMA4A with a novel antibody-drug conjugate in vitro and in vivo.


Subject(s)
Multiple Myeloma , Semaphorins , Cell Membrane/metabolism , Humans , Immunologic Factors , Immunotherapy , Membrane Proteins , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Proteomics , Semaphorins/genetics , Semaphorins/metabolism
7.
Front Pediatr ; 9: 794544, 2021.
Article in English | MEDLINE | ID: mdl-34966707

ABSTRACT

The rise in rates of opioid abuse in recent years in the United States has led to a dramatic increase in the incidence of neonatal abstinence syndrome (NAS). Despite improved understanding of NAS and its acute symptoms, there remains a paucity of information regarding the long-term effects of prenatal exposure to drugs of abuse on neurological development. The primary goal of this study was to investigate the effects of prenatal drug exposure on synaptic connectivity within brain regions associated with the mesolimbic dopamine pathway, the primary reward pathway associated with drug abuse and addiction, in a mouse model. Our secondary goal was to examine the role of the Ca+2 channel subunit α2δ-1, known to be involved in key developmental synaptogenic pathways, in mediating these effects. Pregnant mouse dams were treated orally with either the opioid drug buprenorphine (commonly used in medication-assisted treatment for substance use patients), gabapentin (neuropathic pain drug that binds to α2δ-1 and has been increasingly co-abused with opioids), a combination of both drugs, or vehicle daily from gestational day 6 until postnatal day 11. Confocal fluorescence immunohistochemistry (IHC) imaging of the brains of the resulting wild-type (WT) pups at postnatal day 21 revealed a number of significant alterations in excitatory and inhibitory synaptic populations within the anterior cingulate cortex (ACC), nucleus accumbens (NAC), and medial prefrontal cortex (PFC), particularly in the buprenorphine or combinatorial buprenorphine/gabapentin groups. Furthermore, we observed several drug- and region-specific differences in synaptic connectivity between WT and α2δ-1 haploinsufficient mice, indicating that critical α2δ-1-associated synaptogenic pathways are disrupted with early life drug exposure.

8.
eNeuro ; 8(4)2021.
Article in English | MEDLINE | ID: mdl-34266964

ABSTRACT

The regulation of synaptic connectivity in the brain is vital to proper functioning and development of the CNS. Formation of neural networks in the CNS has been shown to be heavily influenced by astrocytes, which secrete factors, including thrombospondin (TSP) family proteins, that promote synaptogenesis. However, whether this process is different between males and females has not been thoroughly investigated. In this study, we found that cortical neurons purified from newborn male rats showed a significantly more robust synaptogenic response compared with female-derived cells when exposed to factors secreted from astrocytes. This difference was driven largely by the neuronal response to TSP2, which increased synapses in male neurons while showing no effect on female neurons. Blockade of endogenous 17ß-estradiol (E2) production with letrozole normalized the TSP response between male and female cells, indicating a level of regulation by estrogen signaling. Our results suggest that male and female neurons show a divergent response to TSP synaptogenic signaling, contributing to sex differences in astrocyte-mediated synaptic connectivity.


Subject(s)
Astrocytes , Sex Factors , Thrombospondins , Animals , Female , Male , Neurogenesis , Neurons , Rats , Synapses
9.
PLoS Pathog ; 17(7): e1009771, 2021 07.
Article in English | MEDLINE | ID: mdl-34314469

ABSTRACT

The Salmonella enterica effector SteD depletes mature MHC class II (mMHCII) molecules from the surface of infected antigen-presenting cells through ubiquitination of the cytoplasmic tail of the mMHCII ß chain. This requires the Nedd4 family HECT E3 ubiquitin ligase Wwp2 and a tumor-suppressing transmembrane protein adaptor Tmem127. Here, through a proteomic screen of dendritic cells, we found that SteD targets the plasma membrane protein CD97 for degradation by a similar mechanism. SteD enhanced ubiquitination of CD97 on K555 and mutation of this residue eliminated the effect of SteD on CD97 surface levels. We showed that CD97 localises to and stabilises the immunological synapse between dendritic cells and T cells. Removal of CD97 by SteD inhibited dendritic cell-T cell interactions and reduced T cell activation, independently of its effect on MHCII. Therefore, SteD suppresses T cell immunity by two distinct processes.


Subject(s)
Bacterial Proteins/metabolism , Dendritic Cells/immunology , Immunological Synapses/immunology , Receptors, G-Protein-Coupled/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Salmonella Infections/metabolism , Salmonella enterica
10.
PLoS Biol ; 19(4): e3001166, 2021 04.
Article in English | MEDLINE | ID: mdl-33826607

ABSTRACT

Neural stem cell (NSC) transplantation induces recovery in animal models of central nervous system (CNS) diseases. Although the replacement of lost endogenous cells was originally proposed as the primary healing mechanism of NSC grafts, it is now clear that transplanted NSCs operate via multiple mechanisms, including the horizontal exchange of therapeutic cargoes to host cells via extracellular vesicles (EVs). EVs are membrane particles trafficking nucleic acids, proteins, metabolites and metabolic enzymes, lipids, and entire organelles. However, the function and the contribution of these cargoes to the broad therapeutic effects of NSCs are yet to be fully understood. Mitochondrial dysfunction is an established feature of several inflammatory and degenerative CNS disorders, most of which are potentially treatable with exogenous stem cell therapeutics. Herein, we investigated the hypothesis that NSCs release and traffic functional mitochondria via EVs to restore mitochondrial function in target cells. Untargeted proteomics revealed a significant enrichment of mitochondrial proteins spontaneously released by NSCs in EVs. Morphological and functional analyses confirmed the presence of ultrastructurally intact mitochondria within EVs with conserved membrane potential and respiration. We found that the transfer of these mitochondria from EVs to mtDNA-deficient L929 Rho0 cells rescued mitochondrial function and increased Rho0 cell survival. Furthermore, the incorporation of mitochondria from EVs into inflammatory mononuclear phagocytes restored normal mitochondrial dynamics and cellular metabolism and reduced the expression of pro-inflammatory markers in target cells. When transplanted in an animal model of multiple sclerosis, exogenous NSCs actively transferred mitochondria to mononuclear phagocytes and induced a significant amelioration of clinical deficits. Our data provide the first evidence that NSCs deliver functional mitochondria to target cells via EVs, paving the way for the development of novel (a)cellular approaches aimed at restoring mitochondrial dysfunction not only in multiple sclerosis, but also in degenerative neurological diseases.


Subject(s)
Extracellular Vesicles/metabolism , Mitochondria/metabolism , Neural Stem Cells/metabolism , Animals , Biological Transport , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/ultrastructure
11.
Cell Host Microbe ; 29(5): 792-805.e6, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33811831

ABSTRACT

Silencing of nuclear DNA is an essential feature of innate immune responses to invading pathogens. Early in infection, unintegrated lentiviral cDNA accumulates in the nucleus yet remains poorly expressed. In HIV-1-like lentiviruses, the Vpr accessory protein enhances unintegrated viral DNA expression, suggesting Vpr antagonizes cellular restriction. We previously showed how Vpr remodels the host proteome, identifying multiple cellular targets. We now screen these using a targeted CRISPR-Cas9 library and identify SMC5-SMC6 complex localization factor 2 (SLF2) as the Vpr target responsible for silencing unintegrated HIV-1. SLF2 recruits the SMC5/6 complex to unintegrated lentiviruses, and depletion of SLF2, or the SMC5/6 complex, increases viral expression. ATAC-seq demonstrates that Vpr-mediated SLF2 depletion increases chromatin accessibility of unintegrated virus, suggesting that the SMC5/6 complex compacts viral chromatin to silence gene expression. This work implicates the SMC5/6 complex in nuclear immunosurveillance of extrachromosomal DNA and defines its targeting by Vpr as an evolutionarily conserved antagonism.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , HIV Infections/metabolism , HIV-1/physiology , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , HIV Infections/genetics , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Virus Integration , Virus Replication , vpr Gene Products, Human Immunodeficiency Virus/genetics
12.
Front Immunol ; 12: 600056, 2021.
Article in English | MEDLINE | ID: mdl-33628210

ABSTRACT

The cellular response to interferon (IFN) is essential for antiviral immunity, IFN-based therapy and IFN-related disease. The plasma membrane (PM) provides a critical interface between the cell and its environment, and is the initial portal of entry for viruses. Nonetheless, the effect of IFN on PM proteins is surprisingly poorly understood, and has not been systematically investigated in primary immune cells. Here, we use multiplexed proteomics to quantify IFNα2a-stimulated PM protein changes in primary human CD14+ monocytes and CD4+ T cells from five donors, quantifying 606 and 482 PM proteins respectively. Comparison of cell surface proteomes revealed a remarkable invariance between donors in the overall composition of the cell surface from each cell type, but a marked donor-to-donor variability in the effects of IFNα2a. Furthermore, whereas only 2.7% of quantified proteins were consistently upregulated by IFNα2a at the surface of CD4+ T cells, 6.8% of proteins were consistently upregulated in primary monocytes, suggesting that the magnitude of the IFNα2a response varies according to cell type. Among these differentially regulated proteins, we found the viral target Endothelin-converting enzyme 1 (ECE1) to be an IFNα2a-stimulated protein exclusively upregulated at the surface of CD4+ T cells. We therefore provide a comprehensive map of the cell surface of IFNα2a-stimulated primary human immune cells, including previously uncharacterized interferon stimulated genes (ISGs) and candidate antiviral factors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endothelin-Converting Enzymes/immunology , Interferon-alpha/pharmacology , Monocytes/immunology , CD4-Positive T-Lymphocytes/cytology , Humans , Monocytes/cytology , Proteomics
13.
J Virol ; 95(3)2021 01 13.
Article in English | MEDLINE | ID: mdl-33148793

ABSTRACT

Herpes simplex virus 1 (HSV-1) induces a profound host shutoff during lytic infection. The virion host shutoff (vhs) protein plays a key role in this process by efficiently cleaving host and viral mRNAs. Furthermore, the onset of viral DNA replication is accompanied by a rapid decline in host transcriptional activity. To dissect relative contributions of both mechanisms and elucidate gene-specific host transcriptional responses throughout the first 8 h of lytic HSV-1 infection, we used transcriptome sequencing of total, newly transcribed (4sU-labeled) and chromatin-associated RNA in wild-type (WT) and Δvhs mutant infection of primary human fibroblasts. Following virus entry, vhs activity rapidly plateaued at an elimination rate of around 30% of cellular mRNAs per hour until 8 h postinfection (p.i.). In parallel, host transcriptional activity dropped to 10 to 20%. While the combined effects of both phenomena dominated infection-induced changes in total RNA, extensive gene-specific transcriptional regulation was observable in chromatin-associated RNA and was surprisingly concordant between WT and Δvhs infections. Both induced strong transcriptional upregulation of a small subset of genes that were poorly expressed prior to infection but already primed by H3K4me3 histone marks at their promoters. Most interestingly, analysis of chromatin-associated RNA revealed vhs-nuclease-activity-dependent transcriptional downregulation of at least 150 cellular genes, in particular of many integrin adhesome and extracellular matrix components. This was accompanied by a vhs-dependent reduction in protein levels by 8 h p.i. for many of these genes. In summary, our study provides a comprehensive picture of the molecular mechanisms that govern cellular RNA metabolism during the first 8 h of lytic HSV-1 infection.IMPORTANCE The HSV-1 virion host shutoff (vhs) protein efficiently cleaves both host and viral mRNAs in a translation-dependent manner. In this study, we model and quantify changes in vhs activity, as well as virus-induced global loss of host transcriptional activity, during productive HSV-1 infection. In general, HSV-1-induced alterations in total RNA levels were dominated by these two global effects. In contrast, chromatin-associated RNA depicted gene-specific transcriptional changes. This revealed highly concordant transcriptional changes in WT and Δvhs infections, confirmed DUX4 as a key transcriptional regulator in HSV-1 infection, and identified vhs-dependent transcriptional downregulation of the integrin adhesome and extracellular matrix components. The latter explained seemingly gene-specific effects previously attributed to vhs-mediated mRNA degradation and resulted in a concordant loss in protein levels by 8 h p.i. for many of the respective genes.


Subject(s)
Gene Expression Regulation, Viral , Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , RNA, Viral/metabolism , Ribonucleases/metabolism , Viral Proteins/metabolism , Virus Replication , Fibroblasts/metabolism , Fibroblasts/virology , Herpes Simplex/genetics , Herpes Simplex/pathology , Herpes Simplex/virology , Humans , Protein Biosynthesis , Proteome , RNA, Viral/genetics , Ribonucleases/genetics , Transcriptome , Viral Proteins/genetics
14.
Nat Commun ; 11(1): 4940, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009411

ABSTRACT

The HUSH complex represses retroviruses, transposons and genes to maintain the integrity of vertebrate genomes. HUSH regulates deposition of the epigenetic mark H3K9me3, but how its three core subunits - TASOR, MPP8 and Periphilin - contribute to assembly and targeting of the complex remains unknown. Here, we define the biochemical basis of HUSH assembly and find that its modular architecture resembles the yeast RNA-induced transcriptional silencing complex. TASOR, the central HUSH subunit, associates with RNA processing components. TASOR is required for H3K9me3 deposition over LINE-1 repeats and repetitive exons in transcribed genes. In the context of previous studies, this suggests that an RNA intermediate is important for HUSH activity. We dissect the TASOR and MPP8 domains necessary for transgene repression. Structure-function analyses reveal TASOR bears a catalytically-inactive PARP domain necessary for targeted H3K9me3 deposition. We conclude that TASOR is a multifunctional pseudo-PARP that directs HUSH assembly and epigenetic regulation of repetitive genomic targets.


Subject(s)
DNA Transposable Elements/genetics , Epigenesis, Genetic , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Sequence , Antigens, Neoplasm/metabolism , Binding Sites , Exons/genetics , Genome , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Lysine/metabolism , Magnetic Resonance Spectroscopy , Methylation , NAD/metabolism , Nuclear Proteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Domains , RNA/metabolism , RNA Processing, Post-Transcriptional , Transcription, Genetic
15.
Cell Rep ; 33(2): 108249, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053346

ABSTRACT

Kaposi's sarcoma herpesvirus (KSHV) is an oncogenic human virus and the leading cause of mortality in HIV infection. KSHV reactivation from latent- to lytic-stage infection initiates a cascade of viral gene expression. Here we show how these changes remodel the host cell proteome to enable viral replication. By undertaking a systematic and unbiased analysis of changes to the endothelial cell proteome following KSHV reactivation, we quantify >7,000 cellular proteins and 71 viral proteins and provide a temporal profile of protein changes during the course of lytic KSHV infection. Lytic KSHV induces >2-fold downregulation of 291 cellular proteins, including PKR, the key cellular sensor of double-stranded RNA. Despite the multiple episomes per cell, CRISPR-Cas9 efficiently targets KSHV genomes. A complementary KSHV genome-wide CRISPR genetic screen identifies K5 as the viral gene responsible for the downregulation of two KSHV targets, Nectin-2 and CD155, ligands of the NK cell DNAM-1 receptor.


Subject(s)
Endothelial Cells/immunology , Endothelial Cells/virology , Herpesvirus 8, Human/physiology , Immunomodulation , Proteomics , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/virology , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Line , DNA-Directed DNA Polymerase/metabolism , Down-Regulation , Gene Library , Gene Ontology , Genes, Viral , Genetic Testing , Herpesvirus 8, Human/genetics , Humans , Kinetics , Ligands , Mutation/genetics , Proteome/metabolism , Up-Regulation , Viral Proteins/metabolism , Virus Activation , eIF-2 Kinase/metabolism
16.
Tree Physiol ; 40(10): 1466-1473, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32510135

ABSTRACT

Accurate measurement of total fine root decomposition (the amount of dead fine roots decomposed per unit soil volume) is essential for constructing a soil carbon budget. However, the ingrowth/soil core-based models are dependent on the assumptions that fine roots in litterbags/intact cores have the same relative decomposition rate as those in intact soils and that fine root growth and death rates remain constant over time, while minirhizotrons cannot quantify the total fine root decomposition. To improve the accuracy of estimates for total fine root decomposition, we propose a new method (balanced hybrid) with two models that integrate measurements of soil coring and minirhizotrons into a mass balance model. Model input parameters were fine root biomass, necromass and turnover rate for Model 1, and fine root biomass, necromass and death rate for Model 2. We tested the balanced hybrid method in a loblolly pine plantation forest in coastal North Carolina, USA. The total decomposition rate of absorptive fine roots (ARs) (a combination of first- and second-order fine roots) using Models 1 and 2 was 107 ± 13 g m-2 year-1 and 129 ± 12 g m-2 year-1, respectively. Monthly total AR decomposition was highest from August to November, which corresponded with the highest monthly total ARs mortality. The ARs imaged by minirhizotrons well represent those growing in intact soils, evident by a significant and positive relationship between the standing biomass and the standing length. The total decomposition estimate in both models was sensitive to changes in fine root biomass, turnover rate and death rate but not to change in necromass. Compared with Model 2, Model 1 can avoid the technical difficulty of deciding dead time of individual fine roots but requires greater time and effort to accurately measure fine root biomass dynamics. The balanced hybrid method is an improved technique for measuring total fine root decomposition in plantation forests in which the estimates are based on empirical data from soil coring and minirhizotrons, moving beyond assumptions of traditional approaches.


Subject(s)
Soil , Trees , Biomass , Forests , Plant Roots
17.
Elife ; 92020 04 15.
Article in English | MEDLINE | ID: mdl-32292164

ABSTRACT

The seminal description of the cellular restriction factor APOBEC3G and its antagonism by HIV-1 Vif has underpinned two decades of research on the host-virus interaction. We recently reported that HIV-1 Vif is also able to degrade the PPP2R5 family of regulatory subunits of key cellular phosphatase PP2A (PPP2R5A-E; Greenwood et al., 2016; Naamati et al., 2019). We now identify amino acid polymorphisms at positions 31 and 128 of HIV-1 Vif which selectively regulate the degradation of PPP2R5 family proteins. These residues covary across HIV-1 viruses in vivo, favouring depletion of PPP2R5A-E. Through analysis of point mutants and naturally occurring Vif variants, we further show that degradation of PPP2R5 family subunits is both necessary and sufficient for Vif-dependent G2/M cell cycle arrest. Antagonism of PP2A by HIV-1 Vif is therefore independent of APOBEC3 family proteins, and regulates cell cycle progression in HIV-infected cells.


Subject(s)
Cell Cycle Checkpoints , HIV-1/genetics , Protein Phosphatase 2/metabolism , vif Gene Products, Human Immunodeficiency Virus/physiology , APOBEC Deaminases/metabolism , Flow Cytometry , HIV-1/physiology , Host Microbial Interactions , Humans , Point Mutation/genetics , vif Gene Products, Human Immunodeficiency Virus/genetics
18.
Sci Transl Med ; 12(526)2020 01 15.
Article in English | MEDLINE | ID: mdl-31941829

ABSTRACT

Overcoming drug resistance remains a key challenge to cure patients with acute and chronic B cell malignancies. Here, we describe a stromal cell-autonomous signaling pathway, which contributes to drug resistance of malignant B cells. We show that protein kinase C (PKC)-ß-dependent signals from bone marrow-derived stromal cells markedly decrease the efficacy of cytotoxic therapies. Conversely, small-molecule PKC-ß inhibitors antagonize prosurvival signals from stromal cells and sensitize tumor cells to targeted and nontargeted chemotherapy, resulting in enhanced cytotoxicity and prolonged survival in vivo. Mechanistically, stromal PKC-ß controls the expression of adhesion and matrix proteins, required for activation of phosphoinositide 3-kinases (PI3Ks) and the extracellular signal-regulated kinase (ERK)-mediated stabilization of B cell lymphoma-extra large (BCL-XL) in tumor cells. Central to the stroma-mediated drug resistance is the PKC-ß-dependent activation of transcription factor EB, regulating lysosome biogenesis and plasma membrane integrity. Stroma-directed therapies, enabled by direct inhibition of PKC-ß, enhance the effectiveness of many antileukemic therapies.


Subject(s)
Protein Kinase C beta/metabolism , Apoptosis/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Phosphorylation/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured
19.
Cell Rep ; 27(5): 1579-1596.e7, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31042482

ABSTRACT

HIV-1 encodes four "accessory proteins" (Vif, Vpr, Vpu, and Nef), dispensable for viral replication in vitro but essential for viral pathogenesis in vivo. Well characterized cellular targets have been associated with Vif, Vpu, and Nef, which counteract host restriction and promote viral replication. Conversely, although several substrates of Vpr have been described, their biological significance remains unclear. Here, we use complementary unbiased mass spectrometry-based approaches to demonstrate that Vpr is both necessary and sufficient for the DCAF1/DDB1/CUL4 E3 ubiquitin ligase-mediated degradation of at least 38 cellular proteins, causing systems-level changes to the cellular proteome. We therefore propose that promiscuous targeting of multiple host factors underpins complex Vpr-dependent cellular phenotypes and validate this in the case of G2/M cell cycle arrest. Our model explains how Vpr modulates so many cell biological processes and why the functional consequences of previously described Vpr targets, identified and studied in isolation, have proved elusive.


Subject(s)
HIV Infections/metabolism , HIV-1/metabolism , Proteome/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line, Tumor , Conserved Sequence , G2 Phase Cell Cycle Checkpoints , HEK293 Cells , HIV Infections/virology , HIV-1/pathogenicity , Humans , Proteolysis , Proteome/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , vpr Gene Products, Human Immunodeficiency Virus/genetics
20.
Elife ; 82019 03 12.
Article in English | MEDLINE | ID: mdl-30857592

ABSTRACT

Viruses manipulate host cells to enhance their replication, and the identification of cellular factors targeted by viruses has led to key insights into both viral pathogenesis and cell biology. In this study, we develop an HIV reporter virus (HIV-AFMACS) displaying a streptavidin-binding affinity tag at the surface of infected cells, allowing facile one-step selection with streptavidin-conjugated magnetic beads. We use this system to obtain pure populations of HIV-infected primary human CD4+ T cells for detailed proteomic analysis, and quantitate approximately 9000 proteins across multiple donors on a dynamic background of T cell activation. Amongst 650 HIV-dependent changes (q < 0.05), we describe novel Vif-dependent targets FMR1 and DPH7, and 192 proteins not identified and/or regulated in T cell lines, such as ARID5A and PTPN22. We therefore provide a high-coverage functional proteomic atlas of HIV infection, and a mechanistic account of host factors subverted by the virus in its natural target cell.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/virology , Gene Expression Regulation , HIV Infections/metabolism , Proteome , Cell Separation , Cluster Analysis , DNA-Binding Proteins/chemistry , Fragile X Mental Retardation Protein/chemistry , Green Fluorescent Proteins/chemistry , HIV-1/physiology , Humans , Hydrogen-Ion Concentration , Lentivirus , Lymphocyte Activation , Magnetics , Methyltransferases/chemistry , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 22/chemistry , Proteomics , Streptavidin/chemistry , Virus Replication , vif Gene Products, Human Immunodeficiency Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...