Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Proteomics ; 24(9): e2300312, 2024 May.
Article in English | MEDLINE | ID: mdl-38446070

ABSTRACT

The ectoparasitic mite Varroa destructor transmits and triggers viral infections that have deleterious effects on honey bee colonies worldwide. We performed a manipulative experiment in which worker bees collected at emergence were exposed to Varroa for 72 h, and their proteomes were compared with those of untreated control bees. Label-free quantitative proteomics identified 77 differentially expressed A. mellifera proteins (DEPs). In addition, viral proteins were identified by orthogonal analysis, and most importantly, Deformed wing virus (DWV) was found at high levels/intensity in Varroa-exposed bees. Pathway enrichment analysis suggested that the main pathways affected included peroxisomal metabolism, cyto-/exoskeleton reorganization, and cuticular proteins. Detailed examination of individual DEPs revealed that additional changes in DEPs were associated with peroxisomal function. In addition, the proteome data support the importance of TGF-ß signaling in Varroa-DWV interaction and the involvement of the mTORC1 and Hippo pathways. These results suggest that the effect of DWV on bees associated with Varroa feeding results in aberrant autophagy. In particular, autophagy is selectively modulated by peroxisomes, to which the observed proteome changes strongly corresponded. This study complements previous research with different study designs and suggests the importance of the peroxisome, which plays a key role in viral infections.


Subject(s)
Peroxisomes , RNA Viruses , Varroidae , Animals , Bees/virology , Bees/parasitology , Varroidae/virology , Peroxisomes/metabolism , Peroxisomes/virology , RNA Viruses/physiology , Proteomics/methods , Proteome/metabolism , Proteome/analysis , Insect Proteins/metabolism , Signal Transduction , Host-Parasite Interactions
2.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638559

ABSTRACT

Although peroxisomes play an essential role in viral pathogenesis, and viruses are known to change peroxisome morphology, the role of genotype in the peroxisomal response to viruses remains poorly understood. Here, we analyzed the impact of wheat streak mosaic virus (WSMV) on the peroxisome proliferation in the context of pathogen response, redox homeostasis, and yield in two wheat cultivars, Patras and Pamir, in the field trials. We observed greater virus content and yield losses in Pamir than in Patras. Leaf chlorophyll and protein content measured at the beginning of flowering were also more sensitive to WSMV infection in Pamir. Patras responded to the WSMV infection by transcriptional up-regulation of the peroxisome fission genes PEROXIN 11C (PEX11C), DYNAMIN RELATED PROTEIN 5B (DRP5B), and FISSION1A (FIS1A), greater peroxisome abundance, and activation of pathogenesis-related proteins chitinase, and ß-1,3-glucanase. Oppositely, in Pamir, WMSV infection suppressed transcription of peroxisome biogenesis genes and activity of chitinase and ß-1,3-glucanase, and did not affect peroxisome abundance. Activity of ROS scavenging enzymes was higher in Patras than in Pamir. Thus, the impact of WMSV on peroxisome proliferation is genotype-specific and peroxisome abundance can be used as a proxy for the magnitude of plant immune response.


Subject(s)
Disease Resistance/immunology , Peroxisomes/metabolism , Plant Diseases/immunology , Plant Diseases/microbiology , Potyviridae , Triticum/immunology , Triticum/virology , Chitinases/metabolism , Chlorophyll/metabolism , Glucan 1,3-beta-Glucosidase/metabolism , Oxidation-Reduction , Peroxidases/metabolism , Peroxisomes/genetics , Peroxisomes/virology , Phenotype , Plant Leaves/immunology , Plant Leaves/virology , Reactive Oxygen Species/metabolism
3.
Mol Biol Cell ; 32(14): 1273-1282, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34010015

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus that has triggered global health and economic crises. Here we report the effects of SARS-CoV-2 infection on peroxisomes of human cell lines Huh-7 and SK-N-SH. Peroxisomes undergo dramatic changes in morphology in SARS-CoV-2-infected cells. Rearrangement of peroxisomal membranes is followed by redistribution of peroxisomal matrix proteins to the cytosol, resulting in a dramatic decrease in the number of mature peroxisomes. The SARS-CoV-2 ORF14 protein was shown to interact physically with human PEX14, a peroxisomal membrane protein required for matrix protein import and peroxisome biogenesis. Given the important roles of peroxisomes in innate immunity, SARS-CoV-2 may directly target peroxisomes, resulting in loss of peroxisome structural integrity, matrix protein content and ability to function in antiviral signaling.


Subject(s)
Peroxisomes/virology , Animals , Cell Line , Cell Membrane/pathology , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Membrane Proteins/metabolism , Peroxisomes/metabolism , Peroxisomes/pathology , Phosphoproteins/metabolism , Repressor Proteins/metabolism , SARS-CoV-2/metabolism , Vero Cells
4.
Int J Mol Sci ; 20(15)2019 Aug 03.
Article in English | MEDLINE | ID: mdl-31382586

ABSTRACT

Peroxisomes are ubiquitous organelles with well-defined functions in lipid and reactive oxygen species metabolism, having a significant impact on a large number of important diseases. Growing evidence points to them, in concert with mitochondria, as important players within the antiviral response. In this review we summarize and discuss the recent findings concerning the relevance of peroxisomes within innate immunity. We not only emphasize their importance as platforms for cellular antiviral signaling but also review the current information concerning their role in the control of bacterial infections. We furthermore review the recent data that pinpoints peroxisomes as regulators of inflammatory processes.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Peroxisomes/immunology , Adaptor Proteins, Signal Transducing/immunology , Antiviral Agents/therapeutic use , Bacterial Infections/microbiology , Bacterial Infections/virology , Humans , Peroxisomes/microbiology , Peroxisomes/virology , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/immunology
5.
Cells ; 8(7)2019 07 15.
Article in English | MEDLINE | ID: mdl-31311201

ABSTRACT

Zika virus (ZIKV) has emerged as an important human pathogen that can cause congenital defects in the fetus and neurological conditions in adults. The interferon (IFN) system has proven crucial in restricting ZIKV replication and pathogenesis. The canonical IFN response is triggered by the detection of viral RNA through RIG-I like receptors followed by activation of the adaptor protein MAVS on mitochondrial membranes. Recent studies have shown that a second organelle, peroxisomes, also function as a signaling platforms for the IFN response. Here, we investigated how ZIKV infection affects peroxisome biogenesis and antiviral signaling. We show that ZIKV infection depletes peroxisomes in human fetal astrocytes, a brain cell type that can support persistent infection. The peroxisome biogenesis factor PEX11B was shown to inhibit ZIKV replication, likely by increasing peroxisome numbers and enhancing downstream IFN-dependent antiviral signaling. Given that peroxisomes play critical roles in brain development and nerve function, our studies provide important insights into the roles of peroxisomes in regulating ZIKV infection and potentially neuropathogenesis.


Subject(s)
Host-Pathogen Interactions , Peroxisomes/virology , Zika Virus/pathogenicity , Animals , Astrocytes/immunology , Astrocytes/virology , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , Humans , Immunity, Innate , Interferons/metabolism , Membrane Proteins/metabolism , Signal Transduction , Vero Cells , Virus Replication , Zika Virus/physiology
6.
Cell Host Microbe ; 24(4): 526-541.e7, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30269970

ABSTRACT

Viral proteins have evolved to target cellular organelles and usurp their functions for virus replication. Despite the knowledge of these critical functions for several organelles, little is known about peroxisomes during infection. Peroxisomes are primarily metabolic organelles with important functions in lipid metabolism. Here, we discovered that the enveloped viruses human cytomegalovirus (HCMV) and herpes simplex virus type 1 (HSV-1) induce the biogenesis of and unique morphological changes to peroxisomes to support their replication. Targeted proteomic quantification revealed a global virus-induced upregulation of peroxisomal proteins. Mathematical modeling and microscopy structural analysis show that infection triggers peroxisome growth and fission, leading to increased peroxisome numbers and irregular disc-like structures. HCMV-induced peroxisome biogenesis increased the phospholipid plasmalogen, thereby enhancing virus production. Peroxisome regulation and dependence were not observed for the non-enveloped adenovirus. Our findings uncover a role of peroxisomes in viral pathogenesis, with likely implications for multiple enveloped viruses.


Subject(s)
Cytomegalovirus/physiology , Herpesvirus 1, Human/physiology , Organelle Biogenesis , Peroxisomes/virology , Virus Replication/physiology , Adenoviridae/metabolism , Adenoviridae/pathogenicity , Adenoviridae Infections/virology , Cell Line , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/virology , Fibroblasts/virology , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Humans , Peroxisomes/metabolism , Primary Cell Culture , Proteomics
7.
PLoS Pathog ; 14(5): e1007058, 2018 05.
Article in English | MEDLINE | ID: mdl-29746593

ABSTRACT

Human herpesvirus 8 (HHV-8) is causally related to human malignancies. HHV-8 latent viral FLICE-inhibitory protein (vFLIP) is a viral oncoprotein that is linked to pathogenesis, but how its expression is regulated is largely unknown. In an attempt to understand the role of the mitochondrial antiviral signaling (MAVS) adaptor in HHV-8 infection, we discovered that vFLIP expression was post-translationally up-regulated by the MAVS signaling complex on peroxisomes. Furthermore, we demonstrated that vFLIP could be targeted to the peroxisomes, where it was oncogenically active, in a PEX19-dependent manner. Targeted disruption of vFLIP and MAVS interaction resulted in a decrease in vFLIP expression and selectively promoted death of latently HHV-8-infected cells, providing therapeutic potential for treating HHV-8 diseases. Collectively, our experimental results suggest novel involvement of peroxisomes and MAVS in the stabilization of vFLIP and thereby in the establishment or maintenance of HHV-8 latency and associated pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Herpesvirus 8, Human/physiology , Peroxisomes/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Viral Proteins/metabolism , Virus Latency , Adaptor Proteins, Signal Transducing/genetics , Autophagy , Cell Line , Gene Knockout Techniques , HEK293 Cells , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Immunoblotting , Lymphoma, B-Cell/pathology , Peroxisomes/metabolism , Peroxisomes/virology , Real-Time Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Viral Proteins/genetics , Virus Latency/physiology
8.
DNA Cell Biol ; 37(5): 417-421, 2018 May.
Article in English | MEDLINE | ID: mdl-29443540

ABSTRACT

Peroxisomes are membrane-bound organelles that are best known for their roles in lipid metabolism. Mounting evidence indicates that they are also important nodes for antiviral signaling. While research over the past few decades has revealed effective viral strategies to block antiviral signalling pathways from the plasma membrane, mitochondria and/or the nucleus, until recently, very little was known about how viruses interfere with peroxisome-based antiviral signaling. In this essay, we review how viruses use a variety of strategies to interfere with peroxisome biogenesis, a phenomenon that has implications for evasion of the host immune system as well as pathogenesis.


Subject(s)
Host-Pathogen Interactions , Immune Evasion/physiology , Peroxisomes/physiology , Peroxisomes/virology , Virus Diseases , Virus Replication/physiology , Animals , HIV Infections/virology , HIV-1/physiology , Humans , Signal Transduction/physiology , Virus Diseases/virology
9.
PLoS Pathog ; 13(6): e1006360, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28594894

ABSTRACT

HIV-associated neurocognitive disorders (HAND) represent a spectrum neurological syndrome that affects up to 25% of patients with HIV/AIDS. Multiple pathogenic mechanisms contribute to the development of HAND symptoms including chronic neuroinflammation and neurodegeneration. Among the factors linked to development of HAND is altered expression of host cell microRNAs (miRNAs) in brain. Here, we examined brain miRNA profiles among HIV/AIDS patients with and without HAND. Our analyses revealed differential expression of 17 miRNAs in brain tissue from HAND patients. A subset of the upregulated miRNAs (miR-500a-5p, miR-34c-3p, miR-93-3p and miR-381-3p), are predicted to target peroxisome biogenesis factors (PEX2, PEX7, PEX11B and PEX13). Expression of these miRNAs in transfected cells significantly decreased levels of peroxisomal proteins and concomitantly decreased peroxisome numbers or affected their morphology. The levels of miR-500a-5p, miR-34c-3p, miR-93-3p and miR-381-3p were not only elevated in the brains of HAND patients, but were also upregulated during HIV infection of primary macrophages. Moreover, concomitant loss of peroxisomal proteins was observed in HIV-infected macrophages as well as in brain tissue from HIV-infected patients. HIV-induced loss of peroxisomes was abrogated by blocking the functions of the upregulated miRNAs. Overall, these findings point to previously unrecognized miRNA expression patterns in the brains of HIV patients. Targeting peroxisomes by up-regulating miRNAs that repress peroxisome biogenesis factors may represent a novel mechanism by which HIV-1 subverts innate immune responses and/or causes neurocognitive dysfunction.


Subject(s)
HIV Infections/genetics , HIV-1/physiology , MicroRNAs/metabolism , Neurocognitive Disorders/virology , Peroxisomes/metabolism , Brain/metabolism , Brain/virology , HIV Infections/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , Humans , MicroRNAs/genetics , Neurocognitive Disorders/genetics , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/pathology , Neuropathology , Peroxisomes/genetics , Peroxisomes/virology , Up-Regulation
10.
Nat Plants ; 3: 17094, 2017 Jun 19.
Article in English | MEDLINE | ID: mdl-28628079

ABSTRACT

In animals, certain viral proteins are targeted to peroxisomes to dampen the antiviral immune response mediated by these organelles1-3. In plants, RNA interference (RNAi) mediated by small interfering (si)RNA is the main antiviral defence mechanism. To protect themselves against the cell- and non-cell autonomous effects of RNAi, viruses produce viral suppressors of RNA silencing (VSR)4, whose study is crucial to properly understand the biological cycle of plant viruses and potentially find new solutions to control these pathogens. By combining biochemical approaches, cell-specific inhibition of RNAi movement and peroxisome isolation, we show here that one such VSR, the peanut clump virus (PCV)-encoded P15, isolates siRNA from the symplasm by delivering them into the peroxisomal matrix. Infection with PCV lacking this ability reveals that piggybacking of these VSR-bound nucleic acids into peroxisomes potentiates viral systemic movement by preventing the spread of antiviral siRNA. Collectively, these results highlight organellar confinement of antiviral molecules as a novel pathogenic strategy that may have its direct counterpart in other plant and animal viruses.


Subject(s)
Peroxisomes/metabolism , Plant Viruses/physiology , RNA Interference , RNA Viruses/physiology , RNA, Viral/metabolism , Agrobacterium/genetics , Peroxisomes/virology , Plant Diseases/virology , Plants, Genetically Modified , Nicotiana/virology , Virion/isolation & purification
11.
J Cell Mol Med ; 20(4): 750-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26865163

ABSTRACT

Hepatitis C virus (HCV) is the cause of one of the most prevalent viral infections worldwide. Upon infection, the HCV genome activates the RIG-I-MAVS signalling pathway leading to the production of direct antiviral effectors which prevent important steps in viral propagation. MAVS localizes at peroxisomes and mitochondria and coordinate the activation of an effective antiviral response: peroxisomal MAVS is responsible for a rapid but short-termed antiviral response, while the mitochondrial MAVS is associated with the activation of a stable response with delayed kinetics. The HCV NS3-4A protease was shown to specifically cleave the mitochondrial MAVS, inhibiting the downstream response. In this study, we have analysed whether HCV NS3-4A is also able to cleave the peroxisomal MAVS and whether this would have any effect on the cellular antiviral response. We show that NS3-4A is indeed able to specifically cleave this protein and release it into the cytosol, a mechanism that seems to occur at a similar kinetic rate as the cleavage of the mitochondrial MAVS. Under these conditions, RIG-I-like receptor (RLR) signalling from peroxisomes is blocked and antiviral gene expression is inhibited. Our results also show that NS3-4A is able to localize at peroxisomes in the absence of MAVS. However, mutation studies have shown that this localization pattern is preferred in the presence of a fully cleavable MAVS. These findings present evidence of a viral evasion strategy that disrupts RLR signalling on peroxisomes and provide an excellent example of how a single viral evasion strategy can block innate immune signalling from different organelles.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Fibroblasts/virology , Mitochondria/virology , Peroxisomes/virology , Viral Nonstructural Proteins/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Cell Line , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Fibroblasts/immunology , Fibroblasts/ultrastructure , Gene Expression Regulation , Gene Knockout Techniques , Hepacivirus/genetics , Hepacivirus/immunology , Humans , Immune Evasion , Kinetics , Mice , Mitochondria/immunology , Mitochondria/ultrastructure , Mutation , Peroxisomes/immunology , Peroxisomes/ultrastructure , Proteolysis , Signal Transduction/immunology , Viral Nonstructural Proteins/immunology
12.
J Virol ; 90(7): 3611-26, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26792735

ABSTRACT

UNLABELLED: Plus-stranded RNA viruses induce membrane deformations in infected cells in order to build viral replication complexes (VRCs). Tomato bushy stunt virus (TBSV) co-opts cellular ESCRT (endosomal sorting complexes required for transport) proteins to induce the formation of vesicle (spherule)-like structures in the peroxisomal membrane with tight openings toward the cytosol. In this study, using a yeast (Saccharomyces cerevisiae) vps23Δ bro1Δ double-deletion mutant, we showed that the Vps23p ESCRT-I protein (Tsg101 in mammals) and Bro1p (ALIX) ESCRT-associated protein, both of which bind to the viral p33 replication protein, play partially complementary roles in TBSV replication in cells and in cell extracts. Dual expression of dominant-negative versions of Arabidopsis homologs of Vps23p and Bro1p inhibited tombusvirus replication to greater extent than individual expression in Nicotiana benthamiana leaves. We also demonstrated the critical role of Snf7p (CHMP4), Vps20p, and Vps24p ESCRT-III proteins in tombusvirus replication in yeast and in vitro. Electron microscopic imaging of vps23Δ yeast revealed the lack of tombusvirus-induced spherule-like structures, while crescent-like structures are formed in ESCRT-III deletion yeasts replicating TBSV RNA. In addition, we also showed that the length of the viral RNA affects the sizes of spherules formed in N. benthamiana cells. The 4.8-kb genomic RNA is needed for the formation of spherules 66 nm in diameter, while spherules formed during the replication of the ∼600-nucleotide (nt)-long defective interfering RNA in the presence of p33 and p92 replication proteins are 42 nm. We propose that the viral RNA serves as a "measuring string" during VRC assembly and spherule formation. IMPORTANCE: Plant positive-strand RNA viruses, similarly to animal positive-strand RNA viruses, replicate in membrane-bound viral replicase complexes in the cytoplasm of infected cells. Identification of cellular and viral factors affecting the formation of the membrane-bound viral replication complex is a major frontier in current virology research. In this study, we dissected the functions of co-opted cellular ESCRT-I (endosomal sorting complexes required for transport I) and ESCRT-III proteins and the viral RNA in tombusvirus replicase complex formation using in vitro, yeast-based, and plant-based approaches. Electron microscopic imaging revealed the lack of tombusvirus-induced spherule-like structures in ESCRT-I or ESCRT-III deletion yeasts replicating TBSV RNA, demonstrating the requirement for these co-opted cellular factors in tombusvirus replicase formation. The work could be of broad interest in virology and beyond.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Host-Pathogen Interactions , Intracellular Membranes/virology , RNA, Viral/metabolism , Tombusvirus/physiology , Virus Replication , Arabidopsis/genetics , Arabidopsis/virology , Gene Deletion , Microscopy, Electron, Transmission , Peroxisomes/ultrastructure , Peroxisomes/virology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae/virology , Nicotiana/genetics , Nicotiana/ultrastructure , Nicotiana/virology
13.
PLoS Pathog ; 11(11): e1005264, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26588843

ABSTRACT

Sensing viruses by pattern recognition receptors (PRR) triggers the innate immune system of the host cell and activates immune signaling cascades such as the RIG-I/IRF3 pathway. Mitochondrial antiviral-signaling protein (MAVS, also known as IPS-1, Cardif, and VISA) is the crucial adaptor protein of this pathway localized on mitochondria, peroxisomes and mitochondria-associated membranes of the endoplasmic reticulum. Activation of MAVS leads to the production of type I and type III interferons (IFN) as well as IFN stimulated genes (ISGs). To refine the role of MAVS subcellular localization for the induction of type I and III IFN responses in hepatocytes and its counteraction by the hepatitis C virus (HCV), we generated various functional and genetic knock-out cell systems that were reconstituted to express mitochondrial (mito) or peroxisomal (pex) MAVS, exclusively. Upon infection with diverse RNA viruses we found that cells exclusively expressing pexMAVS mounted sustained expression of type I and III IFNs to levels comparable to cells exclusively expressing mitoMAVS. To determine whether viral counteraction of MAVS is affected by its subcellular localization we employed infection of cells with HCV, a major causative agent of chronic liver disease with a high propensity to establish persistence. This virus efficiently cleaves MAVS via a viral protease residing in its nonstructural protein 3 (NS3) and this strategy is thought to contribute to the high persistence of this virus. We found that both mito- and pexMAVS were efficiently cleaved by NS3 and this cleavage was required to suppress activation of the IFN response. Taken together, our findings indicate comparable activation of the IFN response by pex- and mitoMAVS in hepatocytes and efficient counteraction of both MAVS species by the HCV NS3 protease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endoplasmic Reticulum/metabolism , Hepacivirus , Interferons/metabolism , Mitochondria/virology , Peroxisomes/virology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Endoplasmic Reticulum/virology , Hepatocytes/metabolism , Humans , Mice , Mitochondria/metabolism , Peroxisomes/metabolism , Viral Nonstructural Proteins/metabolism
14.
J Lipid Res ; 55(7): 1357-65, 2014 07.
Article in English | MEDLINE | ID: mdl-24868094

ABSTRACT

Influenza virus acquires a host-derived lipid envelope during budding, yet a convergent view on the role of host lipid metabolism during infection is lacking. Using a mass spectrometry-based lipidomics approach, we provide a systems-scale perspective on membrane lipid dynamics of infected human lung epithelial cells and purified influenza virions. We reveal enrichment of the minor peroxisome-derived ether-linked phosphatidylcholines relative to bulk ester-linked phosphatidylcholines in virions as a unique pathogenicity-dependent signature for influenza not found in other enveloped viruses. Strikingly, pharmacological and genetic interference with peroxisomal and ether lipid metabolism impaired influenza virus production. Further integration of our lipidomics results with published genomics and proteomics data corroborated altered peroxisomal lipid metabolism as a hallmark of influenza virus infection in vitro and in vivo. Influenza virus may therefore tailor peroxisomal and particularly ether lipid metabolism for efficient replication.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Peroxisomes/metabolism , Phosphatidylcholines/metabolism , Virus Replication/physiology , A549 Cells , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Humans , Madin Darby Canine Kidney Cells , Peroxisomes/virology
15.
Virology ; 452-453: 133-42, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24606690

ABSTRACT

Tombusviruses replicate on pre-existing organelles such as peroxisomes or mitochondria, the membranes of which become extensively reorganized into multivesicular bodies (MVBs) during the infection process. Cucumber necrosis virus (CNV) has previously been shown to replicate in association with peroxisomes in yeast. We show that CNV induces MVBs from peroxisomes in infected plants and that GFP-tagged p33 auxiliary replicase protein colocalizes with YFP(SKL), a peroxisomal marker. Most remarkably, the ER of CNV infected Nicotiana benthamiana 16C plants undergoes a dramatic reorganization producing numerous new peroxisome-like structures that associate with CNV p33, thus likely serving as a new site for viral RNA replication. We also show that plants agroinfiltrated with p33 develop CNV-like necrotic symptoms which are associated with increased levels of peroxide. Since peroxisomes are a site for peroxide catabolism, and peroxide is known to induce plant defense responses, we suggest that dysfunctional peroxisomes contribute to CNV induced necrosis.


Subject(s)
Endoplasmic Reticulum/virology , Nicotiana/virology , Peroxisomes/virology , RNA-Dependent RNA Polymerase/metabolism , Tombusvirus/enzymology , Tombusvirus/physiology , Viral Proteins/metabolism , Inclusion Bodies, Viral/virology , Protein Transport , RNA-Dependent RNA Polymerase/genetics , Tombusvirus/genetics , Viral Proteins/genetics , Virus Replication
16.
Subcell Biochem ; 69: 67-75, 2013.
Article in English | MEDLINE | ID: mdl-23821143

ABSTRACT

Cell biology and microbiology are some of the oldest areas of scientific inquiry. Despite the depth of knowledge we now have in these respective fields, much remains unclear about how microorganisms interact with host intracellular organelles. Perhaps nowhere is this statement more accurate than in the role of peroxisomes in microbial infections. Peroxisomes were one of the first organelles discovered by Christian De Duve over 50 years ago (de Duve Ann N Y Acad Sci 386:1-4, 1982). These organelles are ubiquitously found in eukaryotic cells, where they serve several well-defined functions in lipid and oxygen homeostasis (Waterham and Wanders Biochim Biophys Acta 1822:1325, 2012). This chapter will discuss the emerging evidence that indicates that in addition to their functions in cellular metabolism, peroxisomes play an important role in viral infections.


Subject(s)
Peroxisomes/virology , Virus Diseases/virology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Host-Pathogen Interactions , Humans , Immunity, Innate , Mitochondria/immunology , Mitochondria/metabolism , Mitochondria/virology , Peroxisomes/immunology , Peroxisomes/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Virus Diseases/immunology , Virus Diseases/metabolism
17.
PLoS One ; 7(1): e29291, 2012.
Article in English | MEDLINE | ID: mdl-22235281

ABSTRACT

BACKGROUND: Innate immune responses have recently been appreciated to play an important role in the pathogenesis of HIV infection. Whereas inadequate innate immune sensing of HIV during acute infection may contribute to failure to control and eradicate infection, persistent inflammatory responses later during infection contribute in driving chronic immune activation and development of immunodeficiency. However, knowledge on specific HIV PAMPs and cellular PRRs responsible for inducing innate immune responses remains sparse. METHODS/PRINCIPAL FINDINGS: Here we demonstrate a major role for RIG-I and the adaptor protein MAVS in induction of innate immune responses to HIV genomic RNA. We found that secondary structured HIV-derived RNAs induced a response similar to genomic RNA. In primary human peripheral blood mononuclear cells and primary human macrophages, HIV RNA induced expression of IFN-stimulated genes, whereas only low levels of type I IFN and tumor necrosis factor α were produced. Furthermore, secondary structured HIV-derived RNA activated pathways to NF-κB, MAP kinases, and IRF3 and co-localized with peroxisomes, suggesting a role for this organelle in RIG-I-mediated innate immune sensing of HIV RNA. CONCLUSIONS/SIGNIFICANCE: These results establish RIG-I as an innate immune sensor of cytosolic HIV genomic RNA with secondary structure, thereby expanding current knowledge on HIV molecules capable of stimulating the innate immune system.


Subject(s)
DEAD-box RNA Helicases/metabolism , Genome, Viral/immunology , HIV-1/genetics , Immunity, Innate , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , DEAD Box Protein 58 , HIV-1/immunology , HIV-1/metabolism , Humans , Interferon Regulatory Factors/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , Peroxisomes/metabolism , Peroxisomes/virology , Protein Transport , Receptors, Immunologic , Signal Transduction/immunology , Viral Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Curr Opin Microbiol ; 14(4): 458-69, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21824805

ABSTRACT

Viruses that are of great importance for global public health, including HIV, influenza and rotavirus, appear to exploit a remarkable organelle, the peroxisome, during intracellular replication in human cells. Peroxisomes are sites of lipid biosynthesis and catabolism, reactive oxygen metabolism, and other metabolic pathways. Viral proteins are targeted to peroxisomes (the spike protein of rotavirus) or interact with peroxisomal proteins (HIV's Nef and influenza's NS1) or use the peroxisomal membrane for RNA replication. The Nef interaction correlates strongly with the crucial Nef function of CD4 downregulation. Viral exploitation of peroxisomal lipid metabolism appears likely. Mostly, functional significance and mechanisms remain to be elucidated. Recently, peroxisomes were discovered to play a crucial role in the innate immune response by signaling the presence of intracellular virus, leading to the first rapid antiviral response. This review unearths, interprets and connects old data, in the hopes of stimulating new and promising research.


Subject(s)
Host-Pathogen Interactions , Peroxisomes/virology , Signal Transduction , Virus Replication , Animals , CD4 Antigens/metabolism , Capsid Proteins/metabolism , HIV/metabolism , HIV/pathogenicity , HIV/physiology , Humans , Immunity, Innate , Intracellular Membranes/metabolism , Orthomyxoviridae/immunology , Orthomyxoviridae/metabolism , Orthomyxoviridae/pathogenicity , Orthomyxoviridae/physiology , Palmitoyl-CoA Hydrolase/metabolism , Peroxisomes/immunology , Peroxisomes/metabolism , Plant Viruses/metabolism , Plant Viruses/pathogenicity , Plant Viruses/physiology , Rotavirus/immunology , Rotavirus/metabolism , Rotavirus/pathogenicity , Rotavirus/physiology , Viral Nonstructural Proteins/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism
19.
Arch Virol ; 154(10): 1695-8, 2009.
Article in English | MEDLINE | ID: mdl-19768632

ABSTRACT

Five new isolates of carnation Italian ringspot virus (CIRV) from cherry trees, Gypsophila and surface water differ from the original carnation isolate (CIRV-car) and also from Pelargonium necrotic spot virus (PelNSV) by having an ORF 1/ORF1-RT with a typical tombusvirus-like 5'end and by inducing the formation of peroxisome- rather than mitochondrion-derived multivesicular bodies (MVBs). This supports with natural isolates earlier conclusions reached by others with artificially produced hybrid viruses that the 5'end of ORF 1 determines from which organelle the MBVs will be derived. CIRV-car might have resulted from a natural recombination event with genome elements of a PelNSV-like virus.


Subject(s)
Multivesicular Bodies/virology , Peroxisomes/virology , Tombusvirus/genetics , Dianthus/virology , Genome, Viral/genetics , Mitochondria/virology , Molecular Sequence Data , Open Reading Frames/genetics , Prunus/virology , Terminator Regions, Genetic/genetics , Tombusvirus/physiology , Trans-Activators/genetics , Virus Replication/genetics
20.
Virology ; 379(2): 294-305, 2008 Sep 30.
Article in English | MEDLINE | ID: mdl-18684480

ABSTRACT

Replication of Tomato bushy stunt virus (TBSV) RNA takes place on the cytosolic membrane surface of peroxisomes in plants and in yeast, a model host. To identify the host proteins involved in assisting the peroxisomal localization of the tombusvirus p33 replication protein, we tested if p33 could bind directly to yeast proteins involved in peroxisomal transport in vitro. This work has led to the demonstration of Pex19p-p33 interaction via pull-down and co-purification experiments. Pex19p was also detected in the tombusvirus replicase after protein cross-linking, suggesting that Pex19p transiently binds to the replicase as could be expected from a transporter. To validate the importance of Pex19p-p33 interaction in TBSV replication in yeast, we re-targeted Pex19p to the mitochondria, which resulted in the re-distribution of a large fraction of p33 to the mitochondria. The expression of the mitochondrial-targeted Pex19p inhibited TBSV RNA accumulation by 2-4-fold in vivo and reduced the in vitro activity of the tombusvirus replicase by 80%. These data support the model that Pex19p is a cellular transporter for localization of p33 replication protein to the host peroxisomal membranes.


Subject(s)
Membrane Proteins/metabolism , Peroxisomes/metabolism , Peroxisomes/virology , Saccharomyces cerevisiae Proteins/metabolism , Tombusvirus/physiology , Viral Proteins/physiology , Base Sequence , Cross-Linking Reagents , DNA Primers/genetics , DNA, Fungal/genetics , Formaldehyde , Host-Pathogen Interactions , Intracellular Membranes/metabolism , Intracellular Membranes/virology , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mitochondria/metabolism , Mitochondria/virology , Models, Biological , Protein Binding , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/virology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/isolation & purification , Tombusvirus/genetics , Tombusvirus/pathogenicity , Viral Proteins/isolation & purification , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...