Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(10)2023 May 19.
Article in English | MEDLINE | ID: mdl-37240354

ABSTRACT

Dendritic cells (DC) are critical cellular mediators of host immunity, notably by expressing a broad panel of pattern recognition receptors. One of those receptors, the C-type lectin receptor DC-SIGN, was previously reported as a regulator of endo/lysosomal targeting through functional connections with the autophagy pathway. Here, we confirmed that DC-SIGN internalization intersects with LC3+ autophagy structures in primary human monocyte-derived dendritic cells (MoDC). DC-SIGN engagement promoted autophagy flux which coincided with the recruitment of ATG-related factors. As such, the autophagy initiation factor ATG9 was found to be associated with DC-SIGN very early upon receptor engagement and required for an optimal DC-SIGN-mediated autophagy flux. The autophagy flux activation upon DC-SIGN engagement was recapitulated using engineered DC-SIGN-expressing epithelial cells in which ATG9 association with the receptor was also confirmed. Finally, Stimulated emission depletion (STED) microscopy performed in primary human MoDC revealed DC-SIGN-dependent submembrane nanoclusters formed with ATG9, which was required to degrade incoming viruses and further limit DC-mediated transmission of HIV-1 infection to CD4+ T lymphocytes. Our study unveils a physical association between the Pattern Recognition Receptor DC-SIGN and essential components of the autophagy pathway contributing to early endocytic events and the host's antiviral immune response.


Subject(s)
HIV-1 , Humans , HIV-1/physiology , Antiviral Agents/metabolism , Dendritic Cells , Lectins, C-Type/metabolism , Autophagy
2.
iScience ; 26(2): 106042, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36818301

ABSTRACT

Mycobacterium abscessus (Mab) is an increasingly recognized pulmonary pathogen. How Mab is internalized by macrophages and establishes infection remains unknown. Here, we show that Mab uptake is significantly reduced in macrophages pre-incubated with neutralizing anti-CD81 antibodies or in cells in which the large extracellular loop (LEL) of CD81 has been deleted. Saturation of Mab with either soluble GST-CD81-LEL or CD81-LEL-derived peptides also diminished internalization of the bacilli. The mycobacterial alkyl hydroperoxide reductase C (AhpC) was unveiled as a major interactant of CD81-LEL. Pre-exposure of macrophages with soluble AhpC inhibited mycobacterial uptake whereas overexpression of AhpC in Mab enhanced its internalization. Importantly, pre-incubation of macrophages with anti-CD81-LEL antibodies inhibited phagocytosis of AhpC-coated beads, indicating that AhpC is a direct interactant of CD81-LEL. Conditional depletion of AhpC in Mab correlated with decreased internalization of Mab. These compelling data unravel a previously unexplored role for CD81/AhpC to promote uptake of pathogenic mycobacteria by host cells.

3.
EMBO J ; 42(7): e111961, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36574362

ABSTRACT

Cytosolic DNA promotes inflammatory responses upon detection by the cyclic GMP-AMP (cGAMP) synthase (cGAS). It has been suggested that cGAS downregulation is an immune escape strategy harnessed by tumor cells. Here, we used glioblastoma cells that show undetectable cGAS levels to address if alternative DNA detection pathways can promote pro-inflammatory signaling. We show that the DNA-PK DNA repair complex (i) drives cGAS-independent IRF3-mediated type I Interferon responses and (ii) that its catalytic activity is required for cGAS-dependent cGAMP production and optimal downstream signaling. We further show that the cooperation between DNA-PK and cGAS favors the expression of chemokines that promote macrophage recruitment in the tumor microenvironment in a glioblastoma model, a process that impairs early tumorigenesis but correlates with poor outcome in glioblastoma patients. Thus, our study supports that cGAS-dependent signaling is acquired during tumorigenesis and that cGAS and DNA-PK activities should be analyzed concertedly to predict the impact of strategies aiming to boost tumor immunogenicity.


Subject(s)
DNA-Activated Protein Kinase , Glioblastoma , Nucleotidyltransferases , Humans , Carcinogenesis , DNA/metabolism , DNA Damage , DNA Repair , Glioblastoma/genetics , Immunity, Innate , Inflammation , Nucleotidyltransferases/metabolism , Tumor Microenvironment , DNA-Activated Protein Kinase/metabolism
4.
Front Immunol ; 13: 862053, 2022.
Article in English | MEDLINE | ID: mdl-35529884

ABSTRACT

To gain access to the brain, a so-called immune-privileged organ due to its physical separation from the blood stream, pathogens and particularly viruses have been selected throughout evolution for their use of specific mechanisms. They can enter the central nervous system through direct infection of nerves or cerebral barriers or through cell-mediated transport. Indeed, peripheral lymphoid and myeloid immune cells can interact with the blood-brain and the blood-cerebrospinal fluid barriers and allow viral brain access using the "Trojan horse" mechanism. Among immune cells, at the frontier between innate and adaptive immune responses, dendritic cells (DCs) can be pathogen carriers, regulate or exacerbate antiviral responses and neuroinflammation, and therefore be involved in viral transmission and spread. In this review, we highlight an important contribution of DCs in the development and the consequences of viral brain infections.


Subject(s)
Dendritic Cells , Virus Diseases , Brain , Central Nervous System , Humans , Myeloid Cells
5.
Emerg Microbes Infect ; 11(1): 761-774, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35191820

ABSTRACT

Usutu virus (USUV) and West Nile virus (WNV) are phylogenetically close emerging arboviruses and constitute a global public health threat. Since USUV and WNV are transmitted by mosquitoes, the first immune cells they encounter are skin-resident dendritic cells, the most peripheral outpost of immune defense. This unique network is composed of Langerhans cells (LCs) and dermal DCs, which reside in the epidermis and the dermis, respectively. Using human skin explants, we show that while both viruses can replicate in keratinocytes, they can also infect resident DCs with distinct tropism: WNV preferentially infects DCs in the dermis, whereas USUV has a greater propensity to infect LCs. Using both purified human epidermal LCs (eLCs) and monocyte derived LCs (MoLCs), we confirm that LCs sustain a faster and more efficient replication of USUV than WNV and that this correlates with a more intense innate immune response to USUV compared with WNV. Next, we show that ectopic expression of the LC-specific C-type lectin receptor (CLR), langerin, in HEK293T cells allows WNV and USUV to bind and enter, but supports the subsequent replication of USUV only. Conversely, blocking or silencing langerin in MoLCs or eLCs made them resistant to USUV infection, thus demonstrating that USUV uses langerin to enter and replicate in LCs. Altogether, our results demonstrate that LCs constitute privileged target cells for USUV in human skin, because langerin favours its entry and replication. Intriguingly, this suggests that USUV efficiently escapes the antiviral functions of langerin, which normally safeguards LCs from most viral infections.


Subject(s)
Flavivirus Infections , West Nile Fever , West Nile virus , Animals , Flavivirus , HEK293 Cells , Humans , Langerhans Cells , West Nile virus/genetics
6.
EMBO J ; 40(16): e106540, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34121210

ABSTRACT

Dendritic cells (DC) subsets, like Langerhans cells (LC), are immune cells involved in pathogen sensing. They express specific antimicrobial cellular factors that are able to restrict infection and limit further pathogen transmission. Here, we identify the alarmin S100A9 as a novel intracellular antiretroviral factor expressed in human monocyte-derived and skin-derived LC. The intracellular expression of S100A9 is decreased upon LC maturation and inversely correlates with enhanced susceptibility to HIV-1 infection of LC. Furthermore, silencing of S100A9 in primary human LC relieves HIV-1 restriction while ectopic expression of S100A9 in various cell lines promotes intrinsic resistance to both HIV-1 and MLV infection by acting on reverse transcription. Mechanistically, the intracellular expression of S100A9 alters viral capsid uncoating and reverse transcription. S100A9 also shows potent inhibitory effect against HIV-1 and MMLV reverse transcriptase (RTase) activity in vitro in a divalent cation-dependent manner. Our findings uncover an unexpected intracellular function of the human alarmin S100A9 in regulating antiretroviral immunity in Langerhans cells.


Subject(s)
Alarmins/genetics , Calgranulin B/genetics , HIV-1/physiology , Langerhans Cells/virology , Moloney murine leukemia virus/physiology , Retroviridae Infections/prevention & control , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cricetulus , HIV-1/genetics , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Leukemia, Experimental/prevention & control , Mice , Moloney murine leukemia virus/genetics , Reverse Transcription , Transforming Growth Factor beta/immunology , Tumor Virus Infections/prevention & control , Virus Replication
7.
Proc Natl Acad Sci U S A ; 117(24): 13708-13718, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32482853

ABSTRACT

The Q fever agent Coxiella burnetii uses a defect in organelle trafficking/intracellular multiplication (Dot/Icm) type 4b secretion system (T4SS) to silence the host innate immune response during infection. By investigating C. burnetii effector proteins containing eukaryotic-like domains, here we identify NopA (nucleolar protein A), which displays four regulator of chromosome condensation (RCC) repeats, homologous to those found in the eukaryotic Ras-related nuclear protein (Ran) guanine nucleotide exchange factor (GEF) RCC1. Accordingly, NopA is found associated with the chromatin nuclear fraction of cells and uses the RCC-like domain to interact with Ran. Interestingly, NopA triggers an accumulation of Ran-GTP, which accumulates at nucleoli of transfected or infected cells, thus perturbing the nuclear import of transcription factors of the innate immune signaling pathway. Accordingly, qRT-PCR analysis on a panel of cytokines shows that cells exposed to the C. burnetii nopA::Tn or a Dot/Icm-defective dotA::Tn mutant strain present a functional innate immune response, as opposed to cells exposed to wild-type C. burnetii or the corresponding nopA complemented strain. Thus, NopA is an important regulator of the innate immune response allowing Coxiella to behave as a stealth pathogen.


Subject(s)
Bacterial Proteins/metabolism , Coxiella burnetii/metabolism , Q Fever/immunology , Animals , Bacterial Proteins/genetics , Coxiella burnetii/genetics , Female , Host-Pathogen Interactions , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, SCID , Q Fever/genetics , Q Fever/microbiology
8.
Viruses ; 12(6)2020 06 11.
Article in English | MEDLINE | ID: mdl-32545337

ABSTRACT

Death domain-associated protein 6 (Daxx) is a multifunctional, ubiquitously expressed and highly conserved chaperone protein involved in numerous cellular processes, including apoptosis, transcriptional repression, and carcinogenesis. In 2015, we identified Daxx as an antiretroviral factor that interfered with HIV-1 replication by inhibiting the reverse transcription step. In the present study, we sought to unravel the molecular mechanism of Daxx-mediated restriction and, in particular, to identify the protein(s) that Daxx targets in order to achieve its antiviral activity. First, we show that the SUMO-interacting motif (SIM) located at the C-terminus of the protein is strictly required for Daxx to inhibit HIV-1 reverse transcription. By performing a quantitative proteomic screen combined with classical biochemical analyses, we found that Daxx associated with incoming HIV-1 cores through a SIM-dependent interaction with cyclophilin A (CypA) and capsid (CA). Daxx was found to reside within a multiprotein complex associated with viral capsids, also containing TNPO3, TRIM5α, and TRIM34. Given the well-known influence of these cellular factors on the stability of HIV-1 cores, we investigated the effect of Daxx on the cytoplasmic fate of incoming cores and found that Daxx prevented HIV-1 uncoating in a SIM-dependent manner. Altogether, our findings suggest that, by recruiting TNPO3, TRIM5α, and TRIM34 and possibly other proteins onto incoming HIV-1 cores through a SIM-dependent interaction with CA-bound CypA, Daxx increases their stability, thus preventing uncoating and reverse transcription. Our study uncovers a previously unknown function of Daxx in the early steps of HIV-1 infection and further illustrates how reverse transcription and uncoating are two tightly interdependent processes.


Subject(s)
Co-Repressor Proteins/metabolism , HIV Infections/metabolism , HIV-1/genetics , Molecular Chaperones/metabolism , SUMO-1 Protein/metabolism , Virus Uncoating , Amino Acid Motifs , Capsid/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/genetics , HIV Infections/genetics , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Reverse Transcription , SUMO-1 Protein/genetics , beta Karyopherins/genetics , beta Karyopherins/metabolism
10.
Sci Adv ; 5(11): eaax3511, 2019 11.
Article in English | MEDLINE | ID: mdl-31799391

ABSTRACT

Plasmacytoid dendritic cells (pDCs) play a crucial role in antiviral innate immunity through their unique capacity to produce large amounts of type I interferons (IFNs) upon viral detection. Tripartite motif (TRIM) proteins have recently come forth as important modulators of innate signaling, but their involvement in pDCs has not been investigated. Here, we performed a rationally streamlined small interfering RNA (siRNA)-based screen of TRIM proteins in human primary pDCs to identify those that are critical for the IFN response. Among candidate hits, TRIM8 emerged as an essential regulator of IFN regulatory factor 7 (IRF7) function. Mechanistically, TRIM8 protects phosphorylated IRF7 (pIRF7) from proteasomal degradation in an E3 ubiquitin ligase-independent manner by preventing its recognition by the peptidyl-prolyl isomerase Pin1. Our findings uncover a previously unknown regulatory mechanism of type I IFN production in pDCs by which TRIM8 and Pin1 oppositely regulate the stability of pIRF7.


Subject(s)
Carrier Proteins/metabolism , Chikungunya virus/immunology , Dendritic Cells/immunology , HIV-1/immunology , Influenza A Virus, H3N2 Subtype/immunology , Interferon Type I/immunology , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nerve Tissue Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Line , HEK293 Cells , Humans , Immunity, Innate/immunology , Interferon Regulatory Factor-7/metabolism , Nerve Tissue Proteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/immunology , Ubiquitin-Protein Ligases/metabolism , Zebrafish
11.
J Invest Dermatol ; 139(1): 157-166, 2019 01.
Article in English | MEDLINE | ID: mdl-30048652

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) propagates within and between individuals via cell-to-cell transmission, and primary infection typically occurs across juxtaposed mucosal surfaces during breastfeeding or sexual intercourse. It is therefore likely that dendritic cells (DCs) are among the first potential targets for HTLV-1. However, it remains unclear how DCs contribute to virus transmission and dissemination in the early stages of infection. We show that an HTLV-1-infected cell line (MT-2) and naturally infected CD4+ T cells transfer p19+ viral particles to the surface of allogeneic DCs via cell-to-cell contacts. Similarly organized cell-to-cell contacts also facilitate DC-mediated transfer of HTLV-1 to autologous CD4+ T cells. These findings shed light on the cellular structures involved in anterograde and retrograde transmission and suggest a key role for DCs in the natural history and pathogenesis of HTLV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Dendritic Cells/virology , Human T-lymphotropic virus 1/physiology , Leukemia, T-Cell/pathology , Virus Replication , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Humans , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/virology , Microscopy, Electron, Scanning , Tumor Cells, Cultured
12.
J Invest Dermatol ; 136(10): 1981-1989, 2016 10.
Article in English | MEDLINE | ID: mdl-27375111

ABSTRACT

Sterile alpha motif (SAM) and histidine-aspartic (HD) domains protein 1 (SAMHD1) was previously identified as a critical post-entry restriction factor to HIV-1 infection in myeloid dendritic cells. Here we show that SAMHD1 is also expressed in epidermis-isolated Langerhans cells (LC), but degradation of SAMHD1 does not rescue HIV-1 or vesicular stomatitis virus G-pseudotyped lentivectors infection in LC. Strikingly, using Langerhans cells model systems (mutz-3-derived LC, monocyte-derived LC [MDLC], and freshly isolated epidermal LC), we characterize previously unreported post-entry restriction activity to HIV-1 in these cells, which acts at HIV-1 reverse transcription, but remains independent of restriction factors SAMHD1 and myxovirus resistance 2 (MX2). We demonstrate that transforming growth factor-ß signaling confers this potent HIV-1 restriction in MDLC during their differentiation and blocking of mothers against decapentaplegic homolog 2 (SMAD2) signaling in MDLC restores cells' infectivity. Interestingly, maturation of MDLC with a toll-like receptor 2 agonist or transforming growth factor-α significantly increases cells' susceptibility to HIV-1 infection, which may explain why HIV-1 acquisition is increased during coinfection with sexually transmitted infections. In conclusion, we report a SAMHD1-independent post-entry restriction in MDLC and LC isolated from epidermis, which inhibits HIV-1 replication. A better understanding of HIV-1 restriction and propagation from LC to CD4(+) T cells may help in the development of new microbicides or vaccines to curb HIV-1 infection at its earliest stages during mucosal transmission.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Langerhans Cells/virology , Monomeric GTP-Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Cell Line , Humans , Monocytes/metabolism , Myxovirus Resistance Proteins/metabolism , SAM Domain and HD Domain-Containing Protein 1 , Transforming Growth Factor alpha/metabolism , Virus Replication/physiology
13.
J Immunol ; 197(2): 517-32, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27288536

ABSTRACT

It is widely assumed that CD4(+) T cells recognize antigenic peptides (epitopes) derived solely from incoming, exogenous, viral particles or proteins. However, alternative sources of MHC class II (MHC-II)-restricted Ags have been described, in particular epitopes derived from newly synthesized proteins (so-called endogenous). In this study, we show that HIV-infected dendritic cells (DC) present MHC-II-restricted endogenous viral Ags to HIV-specific (HS) CD4(+) T cells. This endogenous pathway functions independently of the exogenous route for HIV Ag presentation and offers a distinct possibility for the immune system to activate HS CD4(+) T cells. We examined the implication of autophagy, which plays a crucial role in endogenous viral Ag presentation and thymic selection of CD4(+) T cells, in HIV endogenous presentation. We show that infected DC do not use autophagy to process MHC-II-restricted HIV Ags. This is unlikely to correspond to a viral escape from autophagic degradation, as infecting DC with Nef- or Env-deficient HIV strains did not impact HS T cell activation. However, we demonstrate that, in DC, specific targeting of HIV Ags to autophagosomes using a microtubule-associated protein L chain 3 (LC3) fusion protein effectively enhances and broadens HS CD4(+) T cell responses, thus favoring an endogenous MHC-II-restricted presentation. In summary, in DC, multiple endogenous presentation pathways lead to the activation of HS CD4(+) T cell responses. These findings will help in designing novel strategies to activate HS CD4(+) T cells that are required for CTL activation/maintenance and B cell maturation.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , HIV Infections/immunology , Lymphocyte Activation/immunology , Autophagy/immunology , Blotting, Western , Dendritic Cells/virology , HIV-1/immunology , Histocompatibility Antigens Class II/immunology , Humans , Microscopy, Confocal
14.
J Invest Dermatol ; 136(6): 1172-1181, 2016 06.
Article in English | MEDLINE | ID: mdl-26896775

ABSTRACT

Delivery of vaccine formulations into the dermis using antigen-coated microneedle patches is a promising and safe approach because of efficient antigen delivery and safety. We evaluated an intradermal vaccine using HIV-1 p24 Gag peptide-conjugated polypropylene sulfide nanoparticles to induce immunity against HIV-1. This peptide-conjugated polypropylene sulfide nanoparticle formulation did not accelerate the maturation of blood- or skin-derived subsets of dendritic cells, either generated in vitro or purified ex vivo, despite efficient uptake in the absence of adjuvant. Moreover, dendritic cell-mediated capture of particulate antigen in this form induced potent HIV-1-specific CD4(+) T-cell responses, as well as B-cell-mediated antibody production. Nanoparticle-based intradermal antigen delivery may therefore provide a new option in the global effort to develop an effective vaccine against HIV-1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Drug Delivery Systems/methods , HIV-1/immunology , Immunity, Cellular/drug effects , Vaccines/administration & dosage , Animals , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Dendritic Cells/drug effects , HIV Infections/prevention & control , HIV-1/drug effects , Humans , Nanoparticles/administration & dosage , Polypropylenes/pharmacology , Sulfides/pharmacology
15.
Nat Commun ; 6: 8620, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26503418

ABSTRACT

Autophagy is a conserved homeostatic process active in all human cells and affecting a spectrum of diseases. Here we use a pharmaceutical screen to discover new mechanisms for activation of autophagy. We identify a subset of pharmaceuticals inducing autophagic flux with effects in diverse cellular systems modelling specific stages of several human diseases such as HIV transmission and hyperphosphorylated tau accumulation in Alzheimer's disease. One drug, flubendazole, is a potent inducer of autophagy initiation and flux by affecting acetylated and dynamic microtubules in a reciprocal way. Disruption of dynamic microtubules by flubendazole results in mTOR deactivation and dissociation from lysosomes leading to TFEB (transcription factor EB) nuclear translocation and activation of autophagy. By inducing microtubule acetylation, flubendazole activates JNK1 leading to Bcl-2 phosphorylation, causing release of Beclin1 from Bcl-2-Beclin1 complexes for autophagy induction, thus uncovering a new approach to inducing autophagic flux that may be applicable in disease treatment.


Subject(s)
Alzheimer Disease/physiopathology , Autophagy/drug effects , Drug Evaluation, Preclinical , Small Molecule Libraries/pharmacology , Acetylation/drug effects , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mebendazole/analogs & derivatives , Mebendazole/pharmacology , Microtubules/drug effects , Microtubules/metabolism
16.
J Virol ; 89(1): 615-25, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25339774

ABSTRACT

UNLABELLED: Autophagy is a ubiquitous mechanism involved in the lysosomal-mediated degradation of cellular components when they are engulfed in vacuoles called autophagosomes. Autophagy is also recognized as an important regulator of the innate and adaptive immune responses against numerous pathogens, which have, therefore, developed strategies to block or use the autophagy machinery to their own benefit. Upon human immunodeficiency virus type 1 (HIV-1) infection, viral envelope (Env) glycoproteins induce autophagy-dependent apoptosis of uninfected bystander CD4(+) T lymphocytes, a mechanism likely contributing to the loss of CD4(+) T cells. In contrast, in productively infected CD4(+) T cells, HIV-1 is able to block Env-induced autophagy in order to avoid its antiviral effect. To date, nothing is known about how autophagy restricts HIV-1 infection in CD4(+) T lymphocytes. Here, we report that autophagy selectively degrades the HIV-1 transactivator Tat, a protein essential for viral transcription and virion production. We demonstrated that this selective autophagy-mediated degradation of Tat relies on its ubiquitin-independent interaction with the p62/SQSTM1 adaptor. Taken together, our results provide evidence that the anti-HIV effect of autophagy is specifically due to the degradation of the viral transactivator Tat but that this process is rapidly counteracted by the virus to favor its replication and spread. IMPORTANCE: Autophagy is recognized as one of the most ancient and conserved mechanisms of cellular defense against invading pathogens. Cross talk between HIV-1 and autophagy has been demonstrated depending on the virally challenged cell type, and HIV-1 has evolved strategies to block this process to replicate efficiently. However, the mechanisms by which autophagy restricts HIV-1 infection remain to be elucidated. Here, we report that the HIV-1 transactivator Tat, a protein essential for viral replication, is specifically degraded by autophagy in CD4(+) T lymphocytes. Both Tat present in infected cells and incoming Tat secreted from infected cells are targeted for autophagy degradation through a ubiquitin-independent interaction with the autophagy receptor p62/SQSTM1. This study is the first to demonstrate that selective autophagy can be an antiviral process by degrading a viral transactivator. In addition, the results could help in the design of new therapies against HIV-1 by specifically targeting this mechanism.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV-1/immunology , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Humans , Sequestosome-1 Protein
17.
Redox Biol ; 4: 40-7, 2015.
Article in English | MEDLINE | ID: mdl-25498966

ABSTRACT

12/15-Lipoxygenase (LOX) enzymatically generates oxidized phospholipids in monocytes and macrophages. Herein, we show that cells deficient in 12/15-LOX contain defective mitochondria and numerous cytoplasmic vacuoles containing electron dense material, indicating defects in autophagy or membrane processing, However, both LC3 expression and lipidation were normal both basally and on chloroquine treatment. A LOX-derived oxidized phospholipid, 12-hydroxyeicosatetraenoic acid-phosphatidylethanolamine (12-HETE-PE) was found to be a preferred substrate for yeast Atg8 lipidation, versus native PE, while both native and oxidized PE were effective substrates for LC3 lipidation. Last, phospholipidomics demonstrated altered levels of several phospholipid classes. Thus, we show that oxidized phospholipids generated by 12/15-LOX can act as substrates for key proteins required for effective autophagy and that cells deficient in this enzyme show evidence of autophagic dysfunction. The data functionally link phospholipid oxidation with autophagy for the first time.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Autophagy/genetics , Lipid Metabolism/genetics , Phospholipids/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/analogs & derivatives , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Animals , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 15-Lipoxygenase/genetics , Autophagy-Related Protein 8 Family , Macrophages/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism
18.
PLoS Pathog ; 10(5): e1004058, 2014 May.
Article in English | MEDLINE | ID: mdl-24787765

ABSTRACT

NKG2D plays a major role in controlling immune responses through the regulation of natural killer (NK) cells, αß and γδ T-cell function. This activating receptor recognizes eight distinct ligands (the MHC Class I polypeptide-related sequences (MIC) A andB, and UL16-binding proteins (ULBP)1-6) induced by cellular stress to promote recognition cells perturbed by malignant transformation or microbial infection. Studies into human cytomegalovirus (HCMV) have aided both the identification and characterization of NKG2D ligands (NKG2DLs). HCMV immediate early (IE) gene up regulates NKGDLs, and we now describe the differential activation of ULBP2 and MICA/B by IE1 and IE2 respectively. Despite activation by IE functions, HCMV effectively suppressed cell surface expression of NKGDLs through both the early and late phases of infection. The immune evasion functions UL16, UL142, and microRNA(miR)-UL112 are known to target NKG2DLs. While infection with a UL16 deletion mutant caused the expected increase in MICB and ULBP2 cell surface expression, deletion of UL142 did not have a similar impact on its target, MICA. We therefore performed a systematic screen of the viral genome to search of addition functions that targeted MICA. US18 and US20 were identified as novel NK cell evasion functions capable of acting independently to promote MICA degradation by lysosomal degradation. The most dramatic effect on MICA expression was achieved when US18 and US20 acted in concert. US18 and US20 are the first members of the US12 gene family to have been assigned a function. The US12 family has 10 members encoded sequentially through US12-US21; a genetic arrangement, which is suggestive of an 'accordion' expansion of an ancestral gene in response to a selective pressure. This expansion must have be an ancient event as the whole family is conserved across simian cytomegaloviruses from old world monkeys. The evolutionary benefit bestowed by the combinatorial effect of US18 and US20 on MICA may have contributed to sustaining the US12 gene family.


Subject(s)
Cytomegalovirus , Histocompatibility Antigens Class I/metabolism , Immune Evasion , Killer Cells, Natural/immunology , Lysosomes/metabolism , Proteolysis , Viral Proteins/physiology , Adult , Bacterial Proteins/metabolism , Cells, Cultured , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Enzyme Inhibitors/pharmacology , Humans , Immune Evasion/drug effects , Killer Cells, Natural/drug effects , Leupeptins/pharmacology , Luminescent Proteins/metabolism , Lysosomes/drug effects , Macrolides/pharmacology , NK Cell Lectin-Like Receptor Subfamily K/physiology , Proteolysis/drug effects , Recombinant Proteins/metabolism
19.
PLoS One ; 8(9): e73780, 2013.
Article in English | MEDLINE | ID: mdl-24040067

ABSTRACT

Ribosome-inactivating proteins (RIPs) are endowed with several medicinal properties, including antiviral activity. We demonstrate here that the recently identified type I RIP from Momordica balsamina also possesses antiviral activity, as determined by viral growth curve assays and single-round infection experiments. Importantly, this activity is at play even as doses where the RIP has no cytotoxic effect. In addition, balsamin inhibits HIV-1 replication not only in T cell lines but also in human primary CD4(+) T cells. This antiviral compound exerts its activity at a viral replicative step occurring later than reverse-transcription, most likely on viral protein translation, prior to viral budding and release. Finally, we demonstrate that balsamin antiviral activity is broad since it also impedes influenza virus replication. Altogether our results demonstrate that type I RIP can exert a potent anti-HIV-1 activity which paves the way for new therapeutic avenues for the treatment of viral infections.


Subject(s)
HIV-1/drug effects , Momordica/metabolism , Plant Proteins/pharmacology , Ribosome Inactivating Proteins/pharmacology , Virus Replication/drug effects , Animals , CD4-Positive T-Lymphocytes/virology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Resistance, Viral/genetics , HIV-1/genetics , HIV-1/physiology , Host-Pathogen Interactions/drug effects , Humans , Inhibitory Concentration 50 , Jurkat Cells , Mutation , T-Lymphocytes/pathology , T-Lymphocytes/virology
20.
Retrovirology ; 10: 80, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23899341

ABSTRACT

BACKGROUND: Newly synthesized HIV-1 particles assemble at the plasma membrane of infected cells, before being released as free virions or being transferred through direct cell-to-cell contacts to neighboring cells. Localization of HIV-1 Gag precursor at the cell membrane is necessary and sufficient to trigger viral assembly, whereas the GagPol precursor is additionally required to generate a fully matured virion. HIV-1 Nef is an accessory protein that optimizes viral replication through partly defined mechanisms. Whether Nef modulates Gag and/or GagPol localization and assembly at the membrane and facilitates viral cell-to-cell transfer has not been extensively characterized so far. RESULTS: We report that Nef increases the total amount of Gag proteins present in infected cells, and promotes Gag localization at the cell membrane. Moreover, the processing of p55 into p24 is improved in the presence of Nef. We also examined the effect of Nef during HIV-1 cell-to-cell transfer. We show that without Nef, viral transfer through direct contacts between infected cells and target cells is impaired. With a nef-deleted virus, the number of HIV-1 positive target cells after a short 2h co-culture is reduced, and viral material transferred to uninfected cells is less matured. At later time points, this defect is associated with a reduction in the productive infection of new target cells. CONCLUSIONS: Our results highlight a previously unappreciated role of Nef during the viral replication cycle. Nef promotes HIV-1 Gag membrane localization and processing, and facilitates viral cell-to-cell transfer.


Subject(s)
Cell Membrane/virology , HIV-1/physiology , Virus Assembly , Virus Release , gag Gene Products, Human Immunodeficiency Virus/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Humans , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...