Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30270045

ABSTRACT

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Subject(s)
Capsid Proteins/immunology , Nuclear Matrix-Associated Proteins/immunology , Nuclear Matrix-Associated Proteins/physiology , Octamer Transcription Factors/immunology , Octamer Transcription Factors/physiology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/physiology , Capsid/metabolism , Capsid Proteins/metabolism , Capsid Proteins/physiology , Cell Nucleus/metabolism , DNA, Viral/genetics , DNA, Viral/immunology , DNA-Binding Proteins , Dendritic Cells/immunology , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , HIV-2/genetics , HIV-2/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate/immunology , Macrophages/immunology , Membrane Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/physiology , RNA-Binding Proteins/metabolism , Signal Transduction/immunology
2.
Cell ; 147(6): 1355-68, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22153078

ABSTRACT

Antigen (Ag) crosspresentation by dendritic cells (DCs) involves the presentation of internalized Ags on MHC class I molecules to initiate CD8+ T cell-mediated immunity in response to certain pathogens and tumor cells. Here, we identify the SNARE Sec22b as a specific regulator of Ag crosspresentation. Sec22b localizes to the ER-Golgi intermediate compartment (ERGIC) and pairs to the plasma membrane SNARE syntaxin 4, which is present in phagosomes (Phgs). Depletion of Sec22b inhibits the recruitment of ER-resident proteins to Phgs and to the vacuole containing the Toxoplasma gondii parasite. In Sec22b-deficient DCs, crosspresentation is compromised after Ag phagocytosis or endocytosis and after invasion by T. gondii. Sec22b silencing inhibited Ag export to the cytosol and increased phagosomal degradation by accelerating lysosomal recruitment. Our findings provide insight into an intracellular traffic pathway required for crosspresentation and show that Sec22b-dependent recruitment of ER proteins to Phgs critically influences phagosomal functions in DCs.


Subject(s)
Antigen Presentation , Dendritic Cells/immunology , Escherichia coli Infections/immunology , Escherichia coli , Phagosomes/immunology , R-SNARE Proteins/metabolism , Toxoplasma , Toxoplasmosis/immunology , Animals , Cross Reactions , Dendritic Cells/cytology , Mice , Mice, Inbred C57BL
3.
EMBO J ; 40(8): e105492, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33709510

ABSTRACT

Cells release diverse types of extracellular vesicles (EVs), which transfer complex signals to surrounding cells. Specific markers to distinguish different EVs (e.g. exosomes, ectosomes, enveloped viruses like HIV) are still lacking. We have developed a proteomic profiling approach for characterizing EV subtype composition and applied it to human Jurkat T cells. We generated an interactive database to define groups of proteins with similar profiles, suggesting release in similar EVs. Biochemical validation confirmed the presence of preferred partners of commonly used exosome markers in EVs: CD81/ADAM10/ITGB1, and CD63/syntenin. We then compared EVs from control and HIV-1-infected cells. HIV infection altered EV profiles of several cellular proteins, including MOV10 and SPN, which became incorporated into HIV virions, and SERINC3, which was re-routed to non-viral EVs in a Nef-dependent manner. Furthermore, we found that SERINC3 controls the surface composition of EVs. Our workflow provides an unbiased approach for identifying candidate markers and potential regulators of EV subtypes. It can be widely applied to in vitro experimental systems for investigating physiological or pathological modifications of EV release.


Subject(s)
Extracellular Vesicles/metabolism , HIV Infections/metabolism , Proteome/metabolism , Cells, Cultured , HEK293 Cells , HIV-1 , Humans , Jurkat Cells , Leukosialin/metabolism , Membrane Glycoproteins/metabolism , RNA Helicases/metabolism
4.
Proc Natl Acad Sci U S A ; 119(17): e2107394119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35439048

ABSTRACT

Tumor associated macrophages (TAMs), which differentiate from circulating monocytes, are pervasive across human cancers and comprise heterogeneous populations. The contribution of tumor-derived signals to TAM heterogeneity is not well understood. In particular, tumors release both soluble factors and extracellular vesicles (EVs), whose respective impact on TAM precursors may be different. Here, we show that triple negative breast cancer cells (TNBCs) release EVs and soluble molecules promoting monocyte differentiation toward distinct macrophage fates. EVs specifically promoted proinflammatory macrophages bearing an interferon response signature. The combination in TNBC EVs of surface CSF-1 promoting survival and cargoes promoting cGAS/STING or other activation pathways led to differentiation of this particular macrophage subset. Notably, macrophages expressing the EV-induced signature were found among patients' TAMs. Furthermore, higher expression of this signature was associated with T cell infiltration and extended patient survival. Together, this data indicates that TNBC-released CSF-1-bearing EVs promote a tumor immune microenvironment associated with a better prognosis in TNBC patients.


Subject(s)
Extracellular Vesicles , Triple Negative Breast Neoplasms , Extracellular Vesicles/physiology , Humans , Macrophages , Triple Negative Breast Neoplasms/pathology
5.
Immunity ; 43(6): 1087-100, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26682983

ABSTRACT

The initiation of cytotoxic immune responses by dendritic cells (DCs) requires the presentation of antigenic peptides derived from phagocytosed microbes and infected or dead cells to CD8(+) T cells, a process called cross-presentation. Antigen cross-presentation by non-activated DCs, however, is not sufficient for the effective induction of immune responses. Additionally, DCs need to be activated through innate receptors, like Toll-like receptors (TLRs). During DC maturation, cross-presentation efficiency is first upregulated and then turned off. Here we show that during this transient phase of enhanced cross-presentation, phago-lysosome fusion was blocked by the topological re-organization of lysosomes into perinuclear clusters. LPS-induced lysosomal clustering, inhibition of phago-lysosome fusion and enhanced cross-presentation, all required expression of the GTPase Rab34. We conclude that TLR4 engagement induces a Rab34-dependent re-organization of lysosomal distribution that delays antigen degradation to transiently enhance cross-presentation, thereby optimizing the priming of CD8(+) T cell responses against pathogens.


Subject(s)
Antigen Presentation/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Toll-Like Receptor 4/immunology , Animals , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Lysosomes/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagosomes/immunology , RNA, Small Interfering , Transfection , rab GTP-Binding Proteins/immunology
6.
Proc Natl Acad Sci U S A ; 116(43): 21685-21693, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31591213

ABSTRACT

The human dendritic cell (DC) lineage has recently been unraveled by high-dimensional mapping, revealing the existence of a discrete new population of blood circulating DC precursors (pre-DCs). Whether this new DC population possesses specific functional features as compared to the other blood DC subset upon pathogen encounter remained to be evaluated. A unique feature of pre-DCs among blood DCs is their constitutive expression of the viral adhesion receptor Siglec-1. Here, we show that pre-DCs, but not other blood DC subsets, are susceptible to infection by HIV-1 in a Siglec-1-dependent manner. Siglec-1 mediates pre-DC infection of CCR5- and CXCR4-tropic strains. Infection of pre-DCs is further enhanced in the presence of HIV-2/SIVmac Vpx, indicating that Siglec-1 does not counteract restriction factors such as SAMHD1. Instead, Siglec-1 promotes attachment and fusion of viral particles. HIV-1-infected pre-DCs produce new infectious viral particles that accumulate in intracellular compartments reminiscent of the virus-containing compartment of macrophages. Pre-DC activation by toll-like receptor (TLR) ligands induces an antiviral state that inhibits HIV-1 fusion and infection, but Siglec-1 remains functional and mediates replication-independent transfer of HIV-1 to activated primary T lymphocytes. Altogether, Siglec-1-mediated susceptibility to HIV-1 infection of pre-DCs constitutes a unique functional feature that might represent a preferential relationship of this emerging cell type with viruses.


Subject(s)
Dendritic Cells/virology , HIV Infections/transmission , Sialic Acid Binding Ig-like Lectin 1/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Dendritic Cells/cytology , HIV Infections/pathology , HIV Infections/virology , HIV-1/immunology , Humans , Sialic Acid Binding Ig-like Lectin 1/biosynthesis , Signal Transduction/immunology , Virus Attachment
7.
EMBO J ; 36(20): 3012-3028, 2017 10 16.
Article in English | MEDLINE | ID: mdl-28923825

ABSTRACT

Exosomes, nano-sized secreted extracellular vesicles (EVs), are actively studied for their diagnostic and therapeutic potential. In particular, exosomes secreted by dendritic cells (DCs) have been shown to carry MHC-peptide complexes allowing efficient activation of T lymphocytes, thus displaying potential as promoters of adaptive immune responses. DCs also secrete other types of EVs of different size, subcellular origin and protein composition, whose immune capacities have not been yet compared to those of exosomes. Here, we show that large EVs (lEVs) released by human DCs are as efficient as small EVs (sEVs), including exosomes, to induce CD4+ T-cell activation in vitro When released by immature DCs, however, lEVs and sEVs differ in their capacity to orient T helper (Th) cell responses, the former favouring secretion of Th2 cytokines, whereas the latter promote Th1 cytokine secretion (IFN-γ). Upon DC maturation, however, these functional differences are abolished, and all EVs become able to induce IFN-γ. Our results highlight the need to comprehensively compare the functionalities of EV subtypes in all patho/physiological systems where exosomes are claimed to perform critical roles.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Extracellular Vesicles/metabolism , Lymphocyte Activation , Humans
8.
Proc Natl Acad Sci U S A ; 113(8): E968-77, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858453

ABSTRACT

Extracellular vesicles (EVs) have become the focus of rising interest because of their numerous functions in physiology and pathology. Cells release heterogeneous vesicles of different sizes and intracellular origins, including small EVs formed inside endosomal compartments (i.e., exosomes) and EVs of various sizes budding from the plasma membrane. Specific markers for the analysis and isolation of different EV populations are missing, imposing important limitations to understanding EV functions. Here, EVs from human dendritic cells were first separated by their sedimentation speed, and then either by their behavior upon upward floatation into iodixanol gradients or by immuno-isolation. Extensive quantitative proteomic analysis allowing comparison of the isolated populations showed that several classically used exosome markers, like major histocompatibility complex, flotillin, and heat-shock 70-kDa proteins, are similarly present in all EVs. We identified proteins specifically enriched in small EVs, and define a set of five protein categories displaying different relative abundance in distinct EV populations. We demonstrate the presence of exosomal and nonexosomal subpopulations within small EVs, and propose their differential separation by immuno-isolation using either CD63, CD81, or CD9. Our work thus provides guidelines to define subtypes of EVs for future functional studies.


Subject(s)
Antigens, CD/metabolism , Cell-Derived Microparticles/metabolism , HSP70 Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Proteomics , Biomarkers/metabolism , Humans
9.
Int J Cancer ; 142(1): 133-144, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28884480

ABSTRACT

Human blood monocytes are very potent to take up antigens. Like macrophages in tissue, they efficiently degrade exogenous protein and are less efficient than dendritic cells (DCs) at cross-presenting antigens to CD8+ T cells. Although it is generally accepted that DCs take up tissue antigens and then migrate to lymph nodes to prime T cells, the mechanisms of presentation of antigens taken up by monocytes are poorly documented so far. In the present work, we show that monocytes loaded in vitro with MelanA long peptides retain the capacity to stimulate antigen-specific CD8+ T cell clones after 5 days of differentiation into monocytes-derived dendritic cells (MoDCs). Tagged-long peptides can be visualized in electron-dense endocytic compartments distinct from lysosomes, suggesting that antigens can be protected from degradation for extended periods of time. To address the pathophysiological relevance of these findings, we screened blood monocytes from 18 metastatic melanoma patients and found that CD14+ monocytes from two patients effectively activate a MelanA-specific CD8 T cell clone after in vitro differentiation into MoDCs. This in vivo sampling of tumor antigen by circulating monocytes might alter the tumor-specific immune response and should be taken into account for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , MART-1 Antigen/immunology , Melanoma/immunology , Monocytes/immunology , Antigen Presentation/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Humans , Monocytes/cytology
10.
Blood ; 124(15): 2411-20, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25114264

ABSTRACT

The ontogeny of human Langerhans cells (LCs) remains poorly characterized, in particular the nature of LC precursors and the factors that may drive LC differentiation. Here we report that thymic stromal lymphopoietin (TSLP), a keratinocyte-derived cytokine involved in epithelial inflammation, cooperates with transforming growth factor (TGF)-ß for the generation of LCs. We show that primary human blood BDCA-1(+), but not BDCA-3(+), dendritic cells (DCs) stimulated with TSLP and TGF-ß harbor a typical CD1a(+)Langerin(+) LC phenotype. Electron microscopy established the presence of Birbeck granules, an intracellular organelle specific to LCs. LC differentiation was not observed from tonsil BDCA-1(+) and BDCA-3(+) subsets. TSLP + TGF-ß LCs had a mature phenotype with high surface levels of CD80, CD86, and CD40. They induced a potent CD4(+) T-helper (Th) cell expansion and differentiation into Th2 cells with increased production of tumor necrosis factor-α and interleukin-6 compared with CD34-derived LCs. Our findings establish a novel LC differentiation pathway from BDCA-1(+) blood DCs with potential implications in epithelial inflammation. Therapeutic targeting of TSLP may interfere with tissue LC repopulation from circulating precursors.


Subject(s)
Antigens, CD1/metabolism , Cell Differentiation/drug effects , Cytokines/pharmacology , Dendritic Cells/cytology , Glycoproteins/metabolism , Langerhans Cells/cytology , Transforming Growth Factor beta/pharmacology , Cell Differentiation/genetics , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Gene Expression Profiling , Humans , Langerhans Cells/drug effects , Langerhans Cells/metabolism , Macrophages/drug effects , Macrophages/metabolism , Phenotype , Skin/metabolism , Th2 Cells/cytology , Th2 Cells/drug effects , Th2 Cells/metabolism , Thymic Stromal Lymphopoietin
11.
Nat Commun ; 15(1): 3749, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702311

ABSTRACT

Regulatory T cells (Tregs) are plastic cells playing a pivotal role in the maintenance of immune homeostasis. Tregs actively adapt to the microenvironment where they reside; as a consequence, their molecular and functional profiles differ among tissues and pathologies. In tumors, the features acquired by Tregs remains poorly characterized. Here, we observe that human tumor-infiltrating Tregs selectively overexpress CD74, the MHC class II invariant chain. CD74 has been previously described as a regulator of antigen-presenting cell biology, however its function in Tregs remains unknown. CD74 genetic deletion in human primary Tregs reveals that CD74KO Tregs exhibit major defects in the organization of their actin cytoskeleton and intracellular organelles. Additionally, intratumoral CD74KO Tregs show a decreased activation, a drop in Foxp3 expression, a low accumulation in the tumor, and consistently, they are associated with accelerated tumor rejection in preclinical models in female mice. These observations are unique to tumor conditions as, at steady state, CD74KO-Treg phenotype, survival, and suppressive capacity are unaffected in vitro and in vivo. CD74 therefore emerges as a specific regulator of tumor-infiltrating Tregs and as a target to interfere with Treg anti-tumor activity.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Histocompatibility Antigens Class II , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/genetics , Humans , Female , Mice , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Knockout
12.
J Extracell Vesicles ; 11(7): e12242, 2022 07.
Article in English | MEDLINE | ID: mdl-35790086

ABSTRACT

Eukaryotic cells, including cancer cells, secrete highly heterogeneous populations of extracellular vesicles (EVs). EVs could have different subcellular origin, composition and functional properties, but tools to distinguish between EV subtypes are scarce. Here, we tagged CD63- or CD9-positive EVs secreted by triple negative breast cancer cells with Nanoluciferase enzyme, to set-up a miniaturized method to quantify secretion of these two EV subtypes directly in the supernatant of cells. We performed a cell-based high-content screening to identify clinically-approved drugs able to affect EV secretion. One of the identified hits is Homosalate, an anti-inflammatory drug found in sunscreens which robustly increased EVs' release. Comparing EVs induced by Homosalate with those induced by Bafilomycin A1, we demonstrate that: (1) the two drugs act on EVs generated in distinct subcellular compartments, and (2) EVs released by Homosalate-, but not by Bafilomycin A1-treated cells enhance resistance to anchorage loss in another recipient epithelial tumour cell line. In conclusion, we identified a new drug modifying EV release and demonstrated that under influence of different drugs, triple negative breast cancer cells release EV subpopulations from different subcellular origins harbouring distinct functional properties.


Subject(s)
Extracellular Vesicles , Triple Negative Breast Neoplasms , Dietary Supplements , Humans , Salicylates , Triple Negative Breast Neoplasms/drug therapy
13.
Cell Rep ; 36(13): 109763, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34592156

ABSTRACT

The human immunodeficiency virus (HIV) enters the nucleus to establish infection, but the role of nuclear envelope proteins in this process is incompletely understood. Inner nuclear transmembrane proteins SUN1 and SUN2 connect nuclear lamins to the cytoskeleton and participate in the DNA damage response (DDR). Increased levels of SUN1 or SUN2 potently restrict HIV infection through an unresolved mechanism. Here, we find that the antiviral activities of SUN1 and SUN2 are distinct. HIV-1 and HIV-2 are preferentially inhibited by SUN1 and SUN2, respectively. We identify DNA damage inducers that stimulate HIV-1 infection and show that SUN1, but not SUN2, neutralizes this effect. Finally, we show that chromatin movements and nuclear rotations are associated with the effects of SUN proteins and Lamin A/C on infection. These results reveal an emerging role of chromatin dynamics and the DDR in the control of HIV infection by structural components of the nuclear envelope.


Subject(s)
Chromatin/metabolism , HIV Infections/virology , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Envelope/metabolism , Cell Nucleus/metabolism , Humans , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Envelope/virology , Nuclear Proteins/metabolism
14.
Nat Commun ; 12(1): 4389, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34282141

ABSTRACT

Despite their roles in intercellular communications, the different populations of extracellular vesicles (EVs) and their secretion mechanisms are not fully characterized: how and to what extent EVs form as intraluminal vesicles of endocytic compartments (exosomes), or at the plasma membrane (PM) (ectosomes) remains unclear. Here we follow intracellular trafficking of the EV markers CD9 and CD63 from the endoplasmic reticulum to their residency compartment, respectively PM and late endosomes. We observe transient co-localization at both places, before they finally segregate. CD9 and a mutant CD63 stabilized at the PM are more abundantly released in EVs than CD63. Thus, in HeLa cells, ectosomes are more prominent than exosomes. By comparative proteomic analysis and differential response to neutralization of endosomal pH, we identify a few surface proteins likely specific of either exosomes (LAMP1) or ectosomes (BSG, SLC3A2). Our work sets the path for molecular and functional discrimination of exosomes and small ectosomes in any cell type.


Subject(s)
Exosomes/metabolism , Tetraspanin 29/metabolism , Tetraspanin 30/metabolism , Cell Communication , Cell Membrane/metabolism , Endosomes/metabolism , Extracellular Vesicles/metabolism , Fusion Regulatory Protein 1, Heavy Chain , Gene Knockout Techniques , HeLa Cells , Humans , Membrane Proteins/metabolism , Protein Transport , Proteomics
15.
Sci Signal ; 14(697)2021 08 24.
Article in English | MEDLINE | ID: mdl-34429383

ABSTRACT

Modified vaccinia Ankara (MVA) is a live, attenuated human smallpox vaccine and a vector for the development of new vaccines against infectious diseases and cancer. Efficient activation of the immune system by MVA partially relies on its encounter with dendritic cells (DCs). MVA infection of DCs leads to multiple outcomes, including cytokine production, activation of costimulatory molecules for T cell stimulation, and cell death. Here, we examined how these diverse responses are orchestrated in human DCs. Single-cell analyses revealed that the response to MVA infection in DCs was limited to early viral gene expression. In response to the early events in the viral cycle, we found that DCs grouped into three distinct clusters. A cluster of infected cells sensed the MVA genome by the intracellular innate immunity pathway mediated by cGAS, STING, TBK1, and IRF3 and subsequently produced inflammatory cytokines. In response to these cytokines, a cluster of noninfected bystander cells increased costimulatory molecule expression. A separate cluster of infected cells underwent caspase-dependent apoptosis. Induction of apoptosis persisted after inhibition of innate immunity pathway mediators independently of previously described IRF-dependent or replication-dependent pathways and was a response to early MVA gene expression. Together, our study identified multiple mechanisms that underlie the interactions of MVA with human DCs.


Subject(s)
Vaccinia , Viral Vaccines , Dendritic Cells , Humans , Single-Cell Analysis , Vaccines, DNA
16.
Retrovirology ; 7: 29, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20374631

ABSTRACT

The molecular mechanisms involved in the assembly of newly synthesized Human Immunodeficiency Virus (HIV) particles are poorly understood. Most of the work on HIV-1 assembly has been performed in T cells in which viral particle budding and assembly take place at the plasma membrane. In contrast, few studies have been performed on macrophages, the other major target of HIV-1. Infected macrophages represent a viral reservoir and probably play a key role in HIV-1 physiopathology. Indeed macrophages retain infectious particles for long periods of time, keeping them protected from anti-viral immune response or drug treatments. Here, we present an overview of what is known about HIV-1 assembly in macrophages as compared to T lymphocytes or cell lines.Early electron microscopy studies suggested that viral assembly takes place at the limiting membrane of an intracellular compartment in macrophages and not at the plasma membrane as in T cells. This was first considered as a late endosomal compartment in which viral budding seems to be similar to the process of vesicle release into multi-vesicular bodies. This view was notably supported by a large body of evidence involving the ESCRT (Endosomal Sorting Complex Required for Transport) machinery in HIV-1 budding, the observation of viral budding profiles in such compartments by immuno-electron microscopy, and the presence of late endosomal markers associated with macrophage-derived virions. However, this model needs to be revisited as recent data indicate that the viral compartment has a neutral pH and can be connected to the plasma membrane via very thin micro-channels. To date, the exact nature and biogenesis of the HIV assembly compartment in macrophages remains elusive. Many cellular proteins potentially involved in the late phases of HIV-1 cycle have been identified; and, recently, the list has grown rapidly with the publication of four independent genome-wide screens. However, their respective roles in infected cells and especially in macrophages remain to be characterized. In summary, the complete process of HIV-1 assembly is still poorly understood and will undoubtedly benefit from the ongoing explosion of new imaging techniques allowing better time-lapse and quantitative studies.


Subject(s)
HIV-1/physiology , Macrophages/virology , Monocytes/virology , Virus Assembly , CD4-Positive T-Lymphocytes/virology , Host-Pathogen Interactions , Humans
17.
Front Microbiol ; 11: 1603, 2020.
Article in English | MEDLINE | ID: mdl-32754142

ABSTRACT

A significant proportion of HIV-2-infected patients exhibit natural virological control that is generally absent from HIV-1-infected patients. Along with CD4+ T cells, HIV-1 targets macrophages which may contribute to viral spreading and the latent reservoir. We have studied the relationship between macrophages and HIV-2, focusing on post-entry steps. HIV-2-infected monocyte-derived macrophages (MDMs) produced substantial amounts of viral particles that were largely harbored intracellularly. New viruses assembled at the limiting membrane of internal compartments similar to virus-containing compartments (VCCs) described for HIV-1. VCCs from MDMs infected with either virus shared protein composition and morphology. Strikingly, HIV-2 Gag was mostly absent from the cytosol and almost exclusively localized to the VCCs, whereas HIV-1 Gag was distributed in both locations. Ultrastructural analyses of HIV-2-infected MDMs revealed the presence of numerous VCCs containing both immature and mature particles in the lumen. HIV-2 particles produced de novo by MDMs were poorly infectious in reporter cells and in transmission to activated T cells through a process that appeared independent of BST2 restriction. Rather than being involved in viral spreading, HIV-2-infected macrophages may represent a cell-associated source of viral antigens that can participate in the immune control of HIV-2 infection.

18.
J Virol ; 82(17): 8373-82, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18596106

ABSTRACT

The lentiviral Nef protein has been studied extensively for its ability to induce the downregulation of several immunoreceptors on the surfaces of infected cells. However, Nef expression is unique in inducing highly effective upregulation of the major histocompatibility complex class II-associated chaperone invariant (Ii) chain complexes in different cell types. Under normal conditions, endocytosis of the Ii chain and other molecules, like the transferrin receptor and CD4, is rapid and AP-2 dependent. Human immunodeficiency virus type 1 (HIV-1) Nef expression strongly reduces the internalization of the Ii chain, enhances that of CD4, and does not modify transferrin uptake. The mutation of AP-2 binding motifs LL164 and DD174 in Nef leads to the inhibition of Ii chain upregulation. In AP-2-depleted cells, surface levels of the Ii chain are high and remain unmodified by Nef expression, further indicating that Nef regulates Ii chain internalization via the AP-2 pathway. Immunoprecipitation experiments revealed that the Ii chain can interact with Nef in a dileucine-dependent manner. Importantly, we have shown that Nef-induced CD4 downregulation and Ii chain upregulation are genetically distinguishable. We have identified natural nef alleles that have lost one of the two functions but not the other one. Moreover, we have characterized Nef mutant forms possessing a similar phenotype in the context of HIV-1 infection. Therefore, the Nef-induced accumulation of Ii chain complexes at the cell surface probably results from a complex mechanism leading to the impairment of AP-2-mediated endocytosis rather than from direct competition between Nef and the Ii chain for binding AP-2.


Subject(s)
Gene Products, nef/metabolism , HIV-1/physiology , Histocompatibility Antigens Class II/physiology , Transcription Factor AP-2/physiology , Antigens, Differentiation, B-Lymphocyte , Cell Line , Electroporation , Endocytosis , Gene Expression Regulation, Viral , HIV-1/genetics , HeLa Cells , Humans , Jurkat Cells , Kidney/cytology , RNA, Small Interfering/metabolism , Transfection
19.
J Extracell Vesicles ; 8(1): 1628592, 2019.
Article in English | MEDLINE | ID: mdl-31303981

ABSTRACT

Acetylcholinesterase (AChE) activity is found in abundance in reticulocytes and neurons and was developed as a marker of reticulocyte EVs in the 1970s. Easily, quickly, and cheaply assayed, AChE activity has more recently been proposed as a generic marker for small extracellular vesicles (sEV) or exosomes, and as a negative marker of HIV-1 virions. To evaluate these proposed uses of AChE activity, we examined data from different EV and virus isolation methods using T-lymphocytic (H9, PM1 and Jurkat) and promonocytic (U937) cell lines grown in culture conditions that differed by serum content. When EVs were isolated by differential ultracentrifugation, no correlation between AChE activity and particle count was observed. AChE activity was detected in non-conditioned medium when serum was added, and most of this activity resided in soluble fractions and could not be pelleted by centrifugation. The serum-derived pelletable AChE protein was not completely eliminated from culture medium by overnight ultracentrifugation; however, a serum "extra-depletion" protocol, in which a portion of the supernatant was left undisturbed during harvesting, achieved near-complete depletion. In conditioned medium also, only small percentages of AChE activity could be pelleted together with particles. Furthermore, no consistent enrichment of AChE activity in sEV fractions was observed. Little if any AChE activity is produced by the cells we examined, and this activity was mainly present in non-vesicular structures, as shown by electron microscopy. Size-exclusion chromatography and iodixanol gradient separation showed that AChE activity overlaps only minimally with EV-enriched fractions. AChE activity likely betrays exposure to blood products and not EV abundance, echoing the MISEV 2014 and 2018 guidelines and other publications. Additional experiments may be merited to validate these results for other cell types and biological fluids other than blood.

20.
Nat Commun ; 10(1): 2864, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253807

ABSTRACT

The T cell immune synapse is a site of intense vesicular trafficking. Here we show that the golgin GMAP210, known to capture vesicles and organize membrane traffic at the Golgi, is involved in the vesicular transport of LAT to the immune synapse. Upon activation, more GMAP210 interact with LAT-containing vesicles and go together with LAT to the immune synapse. Regulating LAT recruitment and LAT-dependent signaling, GMAP210 controls T cell activation. Using a rerouting and capture assay, we show that GMAP210 captures VAMP7-decorated vesicles. Overexpressing different domains of GMAP210, we also show that GMAP210 allows their specific delivery to the immune synapse by tethering LAT-vesicles to the Golgi. Finally, in a model of ectopic expression of LAT in ciliated cells, we show that GMAP210 tethering activity controls the delivery of LAT to the cilium. Hence, our results reveal a function for the golgin GMAP210 conveying specific vesicles to the immune synapse.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Golgi Apparatus/physiology , Leukocytes, Mononuclear/physiology , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Transport Vesicles/physiology , Cell Line , Cytoskeletal Proteins , Female , Gene Expression Regulation , Humans , Male , Nuclear Proteins/genetics , R-SNARE Proteins/genetics , R-SNARE Proteins/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Signal Transduction , T-Lymphocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL