Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Immunity ; 56(7): 1468-1484.e7, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37285842

ABSTRACT

Type 2 immune responses are critical in tissue homeostasis, anti-helminth immunity, and allergy. T helper 2 (Th2) cells produce interleukin-4 (IL-4), IL-5, and IL-13 from the type 2 gene cluster under regulation by transcription factors (TFs) including GATA3. To better understand transcriptional regulation of Th2 cell differentiation, we performed CRISPR-Cas9 screens targeting 1,131 TFs. We discovered that activity-dependent neuroprotector homeobox protein (ADNP) was indispensable for immune reactions to allergen. Mechanistically, ADNP performed a previously unappreciated role in gene activation, forming a critical bridge in the transition from pioneer TFs to chromatin remodeling by recruiting the helicase CHD4 and ATPase BRG1. Although GATA3 and AP-1 bound the type 2 cytokine locus in the absence of ADNP, they were unable to initiate histone acetylation or DNA accessibility, resulting in highly impaired type 2 cytokine expression. Our results demonstrate an important role for ADNP in promoting immune cell specialization.


Subject(s)
Histones , Transcription Factors , Histones/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation , Th2 Cells , Cytokines/metabolism , Cell Differentiation , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism
2.
Science ; 380(6651): 1258-1265, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37347855

ABSTRACT

During initiation of antiviral and antitumor T cell-mediated immune responses, dendritic cells (DCs) cross-present exogenous antigens on major histocompatibility complex (MHC) class I molecules. Cross-presentation relies on the unusual "leakiness" of endocytic compartments in DCs, whereby internalized proteins escape into the cytosol for proteasome-mediated generation of MHC I-binding peptides. Given that type 1 conventional DCs excel at cross-presentation, we searched for cell type-specific effectors of endocytic escape. We devised an assay suitable for genetic screening and identified a pore-forming protein, perforin-2 (Mpeg1), as a dedicated effector exclusive to cross-presenting cells. Perforin-2 was recruited to antigen-containing compartments, where it underwent maturation, releasing its pore-forming domain. Mpeg1-/- mice failed to efficiently prime CD8+ T cells to cell-associated antigens, revealing an important role for perforin-2 in cytosolic entry of antigens during cross-presentation.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes , Endocytosis , Pore Forming Cytotoxic Proteins , Animals , Mice , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/genetics , Cross-Priming/immunology , Dendritic Cells/immunology , Endocytosis/genetics , Endocytosis/immunology , Genetic Testing , Histocompatibility Antigens Class I , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Proteolysis
3.
Nat Immunol ; 24(1): 123-135, 2023 01.
Article in English | MEDLINE | ID: mdl-36550322

ABSTRACT

Naive CD4+ T lymphocytes initially undergo antigen-specific activation to promote a broad-spectrum response before adopting bespoke cytokine expression profiles shaped by intercellular microenvironmental cues, resulting in pathogen-focused modular cytokine responses. Interleukin (IL)-4-induced Gata3 upregulation is important for the helper type 2 T cell (TH2 cell) polarization associated with anti-helminth immunity and misdirected allergic inflammation. Whether additional microenvironmental factors participate is unclear. Using whole mouse-genome CRISPR-Cas9 screens, we discovered a previously unappreciated role for αvß3 integrin in TH2 cell differentiation. Low-level αvß3 expression by naive CD4+ T cells contributed to pan-T cell activation by promoting T-T cell clustering and IL-2/CD25/STAT5 signaling. Subsequently, IL-4/Gata3-induced selective upregulation of αvß3 licensed intercellular αvß3-Thy1 interactions among TH2 cells, enhanced mammalian target of rapamycin (mTOR) signaling, supported differentiation and promoted IL-5/IL-13 production. In mice, αvß3 was required for efficient, allergen-driven, antigen-specific lung TH2 cell responses. Thus, αvß3-expressing TH2 cells form multicellular factories to propagate and amplify TH2 cell responses.


Subject(s)
Cytokines , Th2 Cells , Mice , Animals , Cytokines/metabolism , Cell Differentiation , Allergens , Lung , Mammals/metabolism
4.
Cell Rep ; 40(7): 111205, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977488

ABSTRACT

Despite its crucial role in initiation of cytotoxic immune responses, the molecular pathways underlying antigen cross-presentation remain incompletely understood. The mechanism of antigen exit from endocytic compartments into the cytosol is a long-standing matter of controversy, confronting two main models: transfer through specific channels/transporters or rupture of endocytic membranes and leakage of luminal content. By monitoring the occurrence of intracellular damage in conventional dendritic cells (cDCs), we show that cross-presenting cDC1s display more frequent endomembrane injuries and increased recruitment of endosomal sorting complex required for transport (ESCRT)-III, the main repair system for intracellular membranes, relative to cDC2s. Silencing of CHMP2a or CHMP4b, two effector subunits of ESCRT-III, enhances cytosolic antigen export and cross-presentation. This phenotype is partially reversed by chemical inhibition of RIPK3, suggesting that endocytic damage is related to basal activation of the necroptosis pathway. Membrane repair therefore proves crucial in containing antigen export to the cytosol and cross-presentation in cDCs.


Subject(s)
Cross-Priming , Endosomal Sorting Complexes Required for Transport , Antigen Presentation , Antigens/metabolism , Cytosol/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism
5.
Trends Mol Med ; 28(4): 251-252, 2022 04.
Article in English | MEDLINE | ID: mdl-35288004

ABSTRACT

Recently, Bikorimana et al. presented a new vaccine formulation, AccumTM, capable of eliciting efficient T cell-mediated immune responses and controlling tumour growth. AccumTM is designed to facilitate cross-presentation by breaking through endo/lysosomal membranes and enhancing delivery of antigens into the cytosol of dendritic cells (DCs).


Subject(s)
Cross-Priming , Dendritic Cells , Antigen Presentation , Antigens , Humans , Lysosomes
6.
Front Immunol ; 11: 601405, 2020.
Article in English | MEDLINE | ID: mdl-33281828

ABSTRACT

Pore-forming proteins (PFPs) are present in all domains of life, and play an important role in host-pathogen warfare and in the elimination of cancers. They can be employed to deliver specific effectors across membranes, to disrupt membrane integrity interfering with cell homeostasis, and to lyse membranes either destroying intracellular organelles or entire cells. Considering the destructive potential of PFPs, it is perhaps not surprising that mechanisms controlling their activity are remarkably complex, especially in multicellular organisms. Mammalian PFPs discovered to date include the complement membrane attack complex (MAC), perforins, as well as gasdermins. While the primary function of perforin-1 and gasdermins is to eliminate infected or cancerous host cells, perforin-2 and MAC can target pathogens directly. Yet, all mammalian PFPs are in principle capable of generating pores in membranes of healthy host cells which-if uncontrolled-could have dire, and potentially lethal consequences. In this review, we will highlight the strategies employed to protect the host from destruction by endogenous PFPs, while enabling timely and efficient elimination of target cells.


Subject(s)
Cytotoxicity, Immunologic , Immune System/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Adaptive Immunity , Animals , Complement Membrane Attack Complex/metabolism , Humans , Immune System/immunology , Immunity, Innate , Perforin/metabolism , Pore Forming Cytotoxic Proteins/genetics , Signal Transduction , Transcription, Genetic
7.
Cell Rep ; 32(2): 107905, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32668257

ABSTRACT

Cross-presentation of antigens by dendritic cells (DCs) is critical for initiation of anti-tumor immune responses. Yet, key steps involved in trafficking of antigens taken up by DCs remain incompletely understood. Here, we screen 700 US Food and Drug Administration (FDA)-approved drugs and identify 37 enhancers of antigen import from endolysosomes into the cytosol. To reveal their mechanism of action, we generate proteomic organellar maps of control and drug-treated DCs (focusing on two compounds, prazosin and tamoxifen). By combining organellar mapping, quantitative proteomics, and microscopy, we conclude that import enhancers undergo lysosomal trapping leading to membrane permeation and antigen release. Enhancing antigen import facilitates cross-presentation of soluble and cell-associated antigens. Systemic administration of prazosin leads to reduced growth of MC38 tumors and to a synergistic effect with checkpoint immunotherapy in a melanoma model. Thus, inefficient antigen import into the cytosol limits antigen cross-presentation, restraining the potency of anti-tumor immune responses and efficacy of checkpoint blockers.


Subject(s)
Antineoplastic Agents/pharmacology , Cytosol/metabolism , Endosomes/metabolism , Immunity , Neoplasms/immunology , Small Molecule Libraries/pharmacology , Animals , Antigens/metabolism , Biological Transport/drug effects , Cross-Priming/drug effects , Cytosol/drug effects , Dendritic Cells/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , Endosomes/drug effects , Immunity/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/drug therapy , Permeability , Prazosin/pharmacology , Quinazolines/pharmacology , Tamoxifen/pharmacology , beta-Lactamases/metabolism
8.
Proc Natl Acad Sci U S A ; 115(41): 10440-10445, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30209217

ABSTRACT

Adenovirus has enormous potential as a gene-therapy vector, but preexisting immunity limits its widespread application. What is responsible for this immune block is unclear because antibodies potently inhibit transgene expression without impeding gene transfer into target cells. Here we show that antibody prevention of adenoviral gene delivery in vivo is mediated by the cytosolic antibody receptor TRIM21. Genetic KO of TRIM21 or a single-antibody point mutation is sufficient to restore transgene expression to near-naïve immune levels. TRIM21 is also responsible for blocking cytotoxic T cell induction by vaccine vectors, preventing a protective response against subsequent influenza infection and an engrafted tumor. Furthermore, adenoviral preexisting immunity can lead to an augmented immune response upon i.v. administration of the vector. Transcriptomic analysis of vector-transduced tissue reveals that TRIM21 is responsible for the specific up-regulation of hundreds of immune genes, the majority of which are components of the intrinsic or innate response. Together, these data define a major mechanism underlying the preimmune block to adenovirus gene therapy and demonstrate that TRIM21 efficiently blocks gene delivery in vivo while simultaneously inducing a rapid program of immune transcription.


Subject(s)
Adenoviridae Infections/therapy , Adenoviridae/immunology , Antibodies/immunology , Fibrosarcoma/therapy , Genetic Therapy , Ribonucleoproteins/physiology , Vaccination , Adenoviridae Infections/genetics , Adenoviridae Infections/immunology , Animals , Fibrosarcoma/genetics , Fibrosarcoma/immunology , Gene Transfer Techniques , Genetic Vectors , Mice , Mice, Inbred C57BL , Mice, Knockout , Transgenes , Tumor Cells, Cultured
10.
J Exp Med ; 214(8): 2231-2241, 2017 Aug 07.
Article in English | MEDLINE | ID: mdl-28663435

ABSTRACT

CD8+ T cells mediate antigen-specific immune responses that can induce rejection of solid tumors. In this process, dendritic cells (DCs) are thought to take up tumor antigens, which are processed into peptides and loaded onto MHC-I molecules, a process called "cross-presentation." Neither the actual contribution of cross-presentation to antitumor immune responses nor the intracellular pathways involved in vivo are clearly established because of the lack of experimental tools to manipulate this process. To develop such tools, we generated mice bearing a conditional DC-specific mutation in the sec22b gene, a critical regulator of endoplasmic reticulum-phagosome traffic required for cross-presentation. DCs from these mice show impaired cross-presentation ex vivo and defective cross-priming of CD8+ T cell responses in vivo. These mice are also defective for antitumor immune responses and are resistant to treatment with anti-PD-1. We conclude that Sec22b-dependent cross-presentation in DCs is required to initiate CD8+ T cell responses to dead cells and to induce effective antitumor immune responses during anti-PD-1 treatment in mice.


Subject(s)
Cross-Priming/immunology , Neoplasms/immunology , R-SNARE Proteins/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Death/immunology , Dendritic Cells/immunology , Female , Immunity, Cellular/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , R-SNARE Proteins/genetics , RAW 264.7 Cells
11.
Proc Natl Acad Sci U S A ; 114(29): E5910-E5919, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28679634

ABSTRACT

Although antigen cross-presentation in dendritic cells (DCs) is critical to the initiation of most cytotoxic immune responses, the intracellular mechanisms and traffic pathways involved are still unclear. One of the most critical steps in this process, the export of internalized antigen to the cytosol, has been suggested to be mediated by Sec61. Sec61 is the channel that translocates signal peptide-bearing nascent polypeptides into the endoplasmic reticulum (ER), and it was also proposed to mediate protein retrotranslocation during ER-associated degradation (a process called ERAD). Here, we used a newly identified Sec61 blocker, mycolactone, to analyze Sec61's contribution to antigen cross-presentation, ERAD, and transport of internalized antigens into the cytosol. As shown previously in other cell types, mycolactone prevented protein import into the ER of DCs. Mycolactone-mediated Sec61 blockade also potently suppressed both antigen cross-presentation and direct presentation of synthetic peptides to CD8+ T cells. In contrast, it did not affect protein export from the ER lumen or from endosomes into the cytosol, suggesting that the inhibition of cross-presentation was not related to either of these trafficking pathways. Proteomic profiling of mycolactone-exposed DCs showed that expression of mediators of antigen presentation, including MHC class I and ß2 microglobulin, were highly susceptible to mycolactone treatment, indicating that Sec61 blockade affects antigen cross-presentation indirectly. Together, our data suggest that the defective translocation and subsequent degradation of Sec61 substrates is the cause of altered antigen cross-presentation in Sec61-blocked DCs.


Subject(s)
Antigen Presentation/physiology , Endosomes/metabolism , Macrolides/pharmacology , SEC Translocation Channels/metabolism , Animals , Antigen Presentation/drug effects , Cell Line , Cytosol/drug effects , Cytosol/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endoplasmic Reticulum-Associated Degradation/drug effects , Endosomes/drug effects , HEK293 Cells , Humans , Mice , Protein Transport/drug effects , SEC Translocation Channels/antagonists & inhibitors
12.
PLoS One ; 10(10): e0140404, 2015.
Article in English | MEDLINE | ID: mdl-26466362

ABSTRACT

The HIV-1-encoded protein, Nef, plays a key role in the development of AIDS. One of Nef's functions is to keep MHC class I off the surface of infected cells, a process that requires the host proteins clathrin and AP-1. To identify other proteins involved in this pathway, we carried out a genome-wide siRNA library screen on HeLa cells co-expressing HLA-A2 and an inducible form of Nef. Out of 21,121 siRNA pools, 100 were selected for further analysis, based on their ability to either inhibit or enhance downregulation of MHC-I by Nef. When cells were treated with the same siRNA pools as those used in the screen, 79% produced a similar phenotype. However, when the cells were treated with different siRNA reagents targeting the same genes, only 16% produced a similar phenotype. This indicates that most of the hits found in the original screen are likely to have been off-target, an important concern that is often not taken into account in siRNA screening studies. Nevertheless, we identified novel host factors involved in Nef-induced downregulation of MHC-I, including four genes, MIIP, CAMSAP3, SLC6A3, and KCTD19, where multiple reagents produced a strong inhibitory effect on Nef activity. Other hits slightly below our very high stringency cutoff point may also deserve further study. Thus, our dataset is a valuable resource for scientists investigating the pathogenesis of HIV.


Subject(s)
Down-Regulation , HLA-A2 Antigen/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Genome-Wide Association Study , HLA-A2 Antigen/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Potassium Channels/genetics , Potassium Channels/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
13.
PLoS One ; 8(5): e64514, 2013.
Article in English | MEDLINE | ID: mdl-23741335

ABSTRACT

CALM (clathrin assembly lymphoid myeloid leukemia protein) is a cargo-selective adaptor for the post-Golgi R-SNAREs VAMPs 2, 3, and 8, and it also regulates the size of clathrin-coated pits and vesicles at the plasma membrane. The present study has two objectives: to determine whether CALM can sort additional VAMPs, and to investigate whether VAMP sorting contributes to CALM-dependent vesicle size regulation. Using a flow cytometry-based endocytosis efficiency assay, we demonstrate that CALM is also able to sort VAMPs 4 and 7, even though they have sorting signals for other clathrin adaptors. CALM homologues are present in nearly every eukaryote, suggesting that the CALM family may have evolved as adaptors for retrieving all post-Golgi VAMPs from the plasma membrane. Using a knockdown/rescue system, we show that wild-type CALM restores normal VAMP sorting in CALM-depleted cells, but that two non-VAMP-binding mutants do not. However, when we assayed the effect of CALM depletion on coated pit morphology, using a fluorescence microscopy-based assay, we found that the two mutants were as effective as wild-type CALM. Thus, we can uncouple the sorting function of CALM from its structural role.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Monomeric Clathrin Assembly Proteins/metabolism , R-SNARE Proteins/metabolism , Amino Acid Sequence , Coated Pits, Cell-Membrane/ultrastructure , Endocytosis , Gene Expression , Golgi Apparatus/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Monomeric Clathrin Assembly Proteins/antagonists & inhibitors , Monomeric Clathrin Assembly Proteins/genetics , Mutation , Protein Sorting Signals , Protein Transport , R-SNARE Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Alignment , Signal Transduction , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure
14.
Nat Cell Biol ; 15(1): 50-60, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23263279

ABSTRACT

Clathrin-mediated endocytosis is essential for a wide range of cellular functions. We used a multi-step siRNA-based screening strategy to identify regulators of the first step in clathrin-mediated endocytosis, formation of clathrin-coated vesicles (CCVs) at the plasma membrane. A primary genome-wide screen identified 334 hits that caused accumulation of CCV cargo on the cell surface. A secondary screen identified 92 hits that inhibited cargo uptake and/or altered the morphology of clathrin-coated structures. The hits include components of four functional complexes: coat proteins, V-ATPase subunits, spliceosome-associated proteins and acetyltransferase subunits. Electron microscopy revealed that V-ATPase depletion caused the cell to form aberrant non-constricted clathrin-coated structures at the plasma membrane. The V-ATPase-knockdown phenotype was rescued by addition of exogenous cholesterol, indicating that the knockdown blocks clathrin-mediated endocytosis by preventing cholesterol from recycling from endosomes back to the plasma membrane.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Coated Pits, Cell-Membrane/metabolism , Endocytosis/genetics , Genome, Human , Vacuolar Proton-Translocating ATPases/metabolism , Cholesterol/physiology , Clathrin-Coated Vesicles/ultrastructure , Coated Pits, Cell-Membrane/ultrastructure , Filipin/metabolism , Gene Knockdown Techniques , HeLa Cells , Humans , Hydrogen-Ion Concentration , Macrolides/pharmacology , RNA Interference , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/physiology
15.
J Cell Biol ; 197(1): 141-60, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22472443

ABSTRACT

Despite recent advances in mass spectrometry, proteomic characterization of transport vesicles remains challenging. Here, we describe a multivariate proteomics approach to analyzing clathrin-coated vesicles (CCVs) from HeLa cells. siRNA knockdown of coat components and different fractionation protocols were used to obtain modified coated vesicle-enriched fractions, which were compared by stable isotope labeling of amino acids in cell culture (SILAC)-based quantitative mass spectrometry. 10 datasets were combined through principal component analysis into a "profiling" cluster analysis. Overall, 136 CCV-associated proteins were predicted, including 36 new proteins. The method identified >93% of established CCV coat proteins and assigned >91% correctly to intracellular or endocytic CCVs. Furthermore, the profiling analysis extends to less well characterized types of coated vesicles, and we identify and characterize the first AP-4 accessory protein, which we have named tepsin. Finally, our data explain how sequestration of TACC3 in cytosolic clathrin cages causes the severe mitotic defects observed in auxilin-depleted cells. The profiling approach can be adapted to address related cell and systems biological questions.


Subject(s)
Clathrin-Coated Vesicles/chemistry , Proteomics , Adaptor Proteins, Vesicular Transport , Cell Division , Cells, Cultured , Chromatography, Liquid , Clathrin-Coated Vesicles/metabolism , Computational Biology , HeLa Cells , Humans , Mass Spectrometry , Tandem Mass Spectrometry
16.
Traffic ; 11(6): 843-55, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20214754

ABSTRACT

Sorting signals for cargo selection into coated vesicles are usually in the form of short linear motifs. Three motifs for clathrin-mediated endocytosis have been identified: YXXPhi, [D/E]XXXL[L/I] and FXNPXY. To search for new endocytic motifs, we made a library of CD8 chimeras with random sequences in their cytoplasmic tails, and used a novel fluorescence-activated cell sorting (FACS)-based assay to select for endocytosed constructs. Out of the five tails that were most efficiently internalized, only one was found to contain a conventional motif. Two contain dileucine-like sequences that appear to be variations on the [D/E]XXXL[L/I] motif. Another contains a novel internalization signal, YXXXPhiN, which is able to function in cells expressing a mutant mu2 that cannot bind YXXPhi, indicating that it is not a variation on the YXXPhi motif. Similar sequences are present in endogenous proteins, including a functional YXXXPhiN (in addition to a classical YXXPhi) in cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Thus, the repertoire of endocytic motifs is more extensive than the three well-characterized sorting signals.


Subject(s)
Endocytosis , Transcription Factor AP-2/chemistry , Amino Acid Motifs , Animals , Antigens, CD/chemistry , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen , Cell Separation , Flow Cytometry , HeLa Cells , Humans , Mice , Microscopy, Fluorescence/methods , Mutation , T-Lymphocytes, Cytotoxic/immunology
17.
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
18.
Proc Natl Acad Sci U S A ; 101(52): 18018-23, 2004 Dec 28.
Article in English | MEDLINE | ID: mdl-15596717

ABSTRACT

According to the cisternal maturation hypothesis, endoplasmic reticulum (ER)-derived membranes nucleate new Golgi cisternae. The yeast Saccharomyces cerevisiae offers a unique opportunity to test this idea because small buds contain both ER and Golgi structures early in the cell cycle. We previously predicted that mutants defective in ER inheritance also would show defects in Golgi inheritance. Surprisingly, studies of S. cerevisiae have not revealed the expected link between ER and Golgi inheritance. Here, we revisit this issue by generating mutant strains in which many of the small buds are devoid of detectable ER. These strains also show defects in the inheritance of both early and late Golgi cisternae. Strikingly, virtually all of the buds that lack ER also lack early Golgi cisternae. Our results fit with the idea that membranes exported from the ER coalesce with vesicles derived from existing Golgi compartments to generate new Golgi cisternae. This basic mechanism of Golgi inheritance may be conserved from yeast to vertebrate cells.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/physiology , Saccharomyces cerevisiae/physiology , Alleles , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Cell Cycle , Codon , Fungal Proteins/physiology , Golgi Apparatus/metabolism , Green Fluorescent Proteins/metabolism , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Mutation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Time Factors , Vesicular Transport Proteins
19.
Biol Chem ; 385(9): 763-78, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15493870

ABSTRACT

MHC (major histocompatibility complex) class I molecules bind intracellular virus-derived peptides in the endoplasmic reticulum (ER) and present them at the cell surface to cytotoxic T lymphocytes. Peptide-free class I molecules at the cell surface, however, could lead to aberrant T cell killing. Therefore, cells ensure that class I molecules bind high-affinity ligand peptides in the ER, and restrict the export of empty class I molecules to the Golgi apparatus. For both of these safeguard mechanisms, the MHC class I loading complex (which consists of the peptide transporter TAP, the chaperones tapasin and calreticulin, and the protein disulfide isomerase ERp57) plays a central role. This article reviews the actions of accessory proteins in the biogenesis of class I molecules, specifically the functions of the loading complex in high-affinity peptide binding and localization of class I molecules, and the known connections between these two regulatory mechanisms. It introduces new models for the mode of action of tapasin, the role of the class I loading complex in peptide editing, and the intracellular localization of class I molecules.


Subject(s)
Antiporters/chemistry , HLA Antigens/physiology , Histocompatibility Antigens Class I/physiology , Immunoglobulins/chemistry , Models, Molecular , Molecular Chaperones/chemistry , Animals , Antiporters/metabolism , Antiporters/physiology , HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Humans , Immunoglobulins/metabolism , Immunoglobulins/physiology , Membrane Transport Proteins , Molecular Chaperones/metabolism , Molecular Chaperones/physiology , Protein Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...