Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
J Microbiol Immunol Infect ; 56(2): 257-266, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36127231

ABSTRACT

BACKGROUND: The exploration of virology knowledge was limited by the optical technology for the observation of virus. Previously, a three-dimensional multi-resolution real-time microscope system (3D-MRM) was developed to observe the uptake of HIV-1-tat peptide-modified nanoparticles in cell membrane. In this study, we labeled HIV-1 virus-like particles (VLPs) with passivated giant quantum dots (gQDs) and recorded their interactive trajectories with human Jurkat CD4 cells through 3D-MRM. METHODS: The labeled of gQDs of the HIV-1 VLPs in sucrose-gradient purified viral lysates was first confirmed by Cryo-electronic microscopy and Western blot assay. After the infection with CD4 cells, the gQD-labeled VLPs were visualized and their extracellular and intracellular trajectories were recorded by 3D-MRM. RESULTS: A total of 208 prime trajectories was identified and classified into three distinct patterns: cell-free random diffusion pattern, directional movement pattern and cell-associated movement pattern, with distributions and mean durations were 72.6%/87.6 s, 9.1%/402.7 s and 18.3%/68.7 s, respectively. Further analysis of the spatial-temporal relationship between VLP trajectories and CD4 cells revealed the three stages of interactions: (1) cell-associated (extracellular) diffusion stage, (2) cell membrane surfing stage and (3) intracellular directional movement stage. CONCLUSION: A complete trajectory of HIV-1 VLP interacting with CD4 cells was presented in animation. This encapsulating method could increase the accuracy for the observation of HIV-1-CD4 cell interaction in real time and three dimensions.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Membrane , HIV-1 , Microscopy, Electron , Quantum Dots , tat Gene Products, Human Immunodeficiency Virus , Humans , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/ultrastructure , CD4-Positive T-Lymphocytes/virology , HIV-1/physiology , HIV-1/ultrastructure , Imaging, Three-Dimensional/methods , tat Gene Products, Human Immunodeficiency Virus/physiology , Cell-Penetrating Peptides/physiology , Cell Membrane/physiology , Cell Membrane/ultrastructure , Cell Membrane/virology , Nanoparticles/ultrastructure , Nanoparticles/virology , Artificial Virus-Like Particles/physiology , Microscopy, Electron/methods
2.
Dis Markers ; 2022: 3424413, 2022.
Article in English | MEDLINE | ID: mdl-35251370

ABSTRACT

BACKGROUND: Reports on the expression of CD38 in Sézary syndrome (SS), erythrodermic primary cutaneous T cell lymphoma with leukemic involvement, are limited. The aim of the present study is the analysis of the expression of CD38 by skin-infiltrating mononuclear cells and circulating T lymphocytes in a cohort of SS patients. METHODS: SS patients diagnosed since 1985 in our clinic were retrospectively analyzed for CD38 expression in biopsy and blood samples by immunohistochemistry and flow cytometry, respectively. RESULTS: SS patients show a predominant CD38-negative phenotype on both skin and blood. A subgroup of patients was found expressing CD38 (12 cases) in either the skin (>25% cell infiltrate) or blood (CD4+CD38+ >50%), among whom 4 in the blood, 7 in the skin, and 1 in both blood and skin. CONCLUSION: The implications of these observations may be twofold: the relevance in basic science is related to a potential role in immune defense regulation, whilst in perspective CD38 may become a target for antibody therapy, considering the availability of different anti-CD38 monoclonal antibodies.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Biomarkers, Tumor/blood , Flow Cytometry , Immunohistochemistry , Membrane Glycoproteins/immunology , Sezary Syndrome , Skin Neoplasms , ADP-ribosyl Cyclase 1/genetics , Biopsy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , Female , Humans , Lymphocyte Count , Male , Membrane Glycoproteins/genetics , Middle Aged , Retrospective Studies , Sezary Syndrome/immunology , Sezary Syndrome/pathology , Skin/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/ultrastructure
3.
Diabetes ; 71(3): 566-577, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35007320

ABSTRACT

Type 1 diabetes in children is heralded by a preclinical phase defined by circulating autoantibodies to pancreatic islet antigens. How islet autoimmunity is initiated and then progresses to clinical diabetes remains poorly understood. Only one study has reported gene expression in specific immune cells of children at risk associated with progression to islet autoimmunity. We analyzed gene expression with RNA sequencing in CD4+ and CD8+ T cells, natural killer (NK) cells, and B cells, and chromatin accessibility by assay for transposase-accessible chromatin sequencing (ATAC-seq) in CD4+ T cells, in five genetically at risk children with islet autoantibodies who progressed to diabetes over a median of 3 years ("progressors") compared with five children matched for sex, age, and HLA-DR who had not progressed ("nonprogressors"). In progressors, differentially expressed genes (DEGs) were largely confined to CD4+ T cells and enriched for cytotoxicity-related genes/pathways. Several top-ranked DEGs were validated in a semi-independent cohort of 13 progressors and 11 nonprogressors. Flow cytometry confirmed that progression was associated with expansion of CD4+ cells with a cytotoxic phenotype. By ATAC-seq, progression was associated with reconfiguration of regulatory chromatin regions in CD4+ cells, some linked to differentially expressed cytotoxicity-related genes. Our findings suggest that cytotoxic CD4+ T cells play a role in promoting progression to type 1 diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chromatin/chemistry , Cytotoxicity, Immunologic/genetics , Diabetes Mellitus, Type 1/immunology , Disease Progression , Gene Expression Regulation , Adolescent , Autoimmunity/genetics , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/metabolism , Child , Child, Preschool , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Humans , Islets of Langerhans/immunology , Killer Cells, Natural/metabolism , Sequence Analysis, RNA
4.
Mediators Inflamm ; 2021: 8481013, 2021.
Article in English | MEDLINE | ID: mdl-34803518

ABSTRACT

Exosomes are membrane-bound extracellular vesicles that are produced in the endosomal compartment of most eukaryotic cells. Containing proteins, RNA, and DNA, exosomes mediate intercellular communication between different cell types by transferring their contents and thus are involved in numerous physiological and pathological processes. T cells are an indispensable part of adaptive immunity, and the functions of T cell-derived exosomes have been widely studied. In the more than three decades since the discovery of exosomes, several studies have revealed that T cell-derived exosomes play a novel role in cell-to-cell signaling, especially in inflammatory responses, autoimmunity, and infectious diseases. In this review, we will summarize the function of T cell-derived exosomes and their therapeutic potential.


Subject(s)
Exosomes/physiology , T-Lymphocytes/ultrastructure , CD4-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/ultrastructure , Humans , T-Lymphocytes, Regulatory/ultrastructure
5.
Nat Immunol ; 22(12): 1551-1562, 2021 12.
Article in English | MEDLINE | ID: mdl-34811544

ABSTRACT

Misdirected immunity gives rise to the autoimmune tissue inflammation of rheumatoid arthritis, in which excess production of the cytokine tumor necrosis factor (TNF) is a central pathogenic event. Mechanisms underlying the breakdown of self-tolerance are unclear, but T cells in the arthritic joint have a distinctive metabolic signature of ATPlo acetyl-CoAhi proinflammatory effector cells. Here we show that a deficiency in the production of mitochondrial aspartate is an important abnormality in these autoimmune T cells. Shortage of mitochondrial aspartate disrupted the regeneration of the metabolic cofactor nicotinamide adenine dinucleotide, causing ADP deribosylation of the endoplasmic reticulum (ER) sensor GRP78/BiP. As a result, ribosome-rich ER membranes expanded, promoting co-translational translocation and enhanced biogenesis of transmembrane TNF. ERrich T cells were the predominant TNF producers in the arthritic joint. Transfer of intact mitochondria into T cells, as well as supplementation of exogenous aspartate, rescued the mitochondria-instructed expansion of ER membranes and suppressed TNF release and rheumatoid tissue inflammation.


Subject(s)
Arthritis, Rheumatoid/metabolism , Aspartic Acid/metabolism , CD4-Positive T-Lymphocytes/metabolism , Mitochondria/metabolism , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , ADP-Ribosylation , Adoptive Transfer , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoimmunity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , CD4-Positive T-Lymphocytes/ultrastructure , Case-Control Studies , Cells, Cultured , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Chaperone BiP/metabolism , Female , Humans , Male , Mice , Mitochondria/immunology , Mitochondria/transplantation , Mitochondria/ultrastructure , Synovial Membrane/immunology , Synovial Membrane/ultrastructure , Tumor Necrosis Factor-alpha/genetics
6.
Elife ; 102021 04 27.
Article in English | MEDLINE | ID: mdl-33904396

ABSTRACT

HIV-1 replication commences inside the cone-shaped viral capsid, but timing, localization, and mechanism of uncoating are under debate. We adapted a strategy to visualize individual reverse-transcribed HIV-1 cDNA molecules and their association with viral and cellular proteins using fluorescence and correlative-light-and-electron-microscopy (CLEM). We specifically detected HIV-1 cDNA inside nuclei, but not in the cytoplasm. Nuclear cDNA initially co-localized with a fluorescent integrase fusion (IN-FP) and the viral CA (capsid) protein, but cDNA-punctae separated from IN-FP/CA over time. This phenotype was conserved in primary HIV-1 target cells, with nuclear HIV-1 complexes exhibiting strong CA-signals in all cell types. CLEM revealed cone-shaped HIV-1 capsid-like structures and apparently broken capsid-remnants at the position of IN-FP signals and elongated chromatin-like structures in the position of viral cDNA punctae lacking IN-FP. Our data argue for nuclear uncoating by physical disruption rather than cooperative disassembly of the CA-lattice, followed by physical separation from the pre-integration complex.


When viruses infect human cells, they hijack the cell's machinery to produce the proteins they need to replicate. Retroviruses like HIV-1 do this by entering the nucleus and inserting their genetic information into the genome of the infected cell. This requires HIV-1 to convert its genetic material into DNA, which is then released from the protective shell surrounding it (known as the capsid) via a process called uncoating. The nucleus is enclosed within an envelope containing pores that molecules up to a certain size can pass through. Until recently these pores were thought to be smaller than the viral capsid, which led scientists to believe that the HIV-1 genome must shed this coat before penetrating the nucleus. However, recent studies have found evidence for HIV-1 capsid proteins and capsid structures inside the nucleus of some infected cells. This suggests that the capsid may not be removed before nuclear entry or that it may even play a role in helping the virus get inside the nucleus. To investigate this further, Müller et al. attached fluorescent labels to the newly made DNA of HIV-1 and some viral and cellular proteins. Powerful microscopy tools were then used to monitor the uncoating process in various cells that had been infected with the virus. Müller et al. found large amounts of capsid protein inside the nuclei of all the infected cells studied. During the earlier stages of infection, the capsid proteins were mostly associated with viral DNA and the capsid structure appeared largely intact. At later time points, the capsid structure had been broken down and the viral DNA molecules were gradually separating themselves from these remnants. These findings suggest that the HIV-1 capsid helps the virus get inside the nucleus and may protect its genetic material during conversion into DNA until right before integration into the cell's genome. Further experiments studying this process could lead to new therapeutic approaches that target the capsid as a way to prevent or treat HIV-1.


Subject(s)
Cell Nucleus/virology , DNA Replication , DNA, Viral/biosynthesis , HIV Infections/virology , HIV-1/growth & development , Virus Internalization , Virus Replication , Virus Uncoating , CD4-Positive T-Lymphocytes/ultrastructure , CD4-Positive T-Lymphocytes/virology , Capsid Proteins/metabolism , Cell Nucleus/ultrastructure , DNA, Viral/genetics , DNA, Viral/ultrastructure , HEK293 Cells , HIV Infections/pathology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , HIV-1/ultrastructure , HeLa Cells , Host-Pathogen Interactions , Humans , Macrophages/ultrastructure , Macrophages/virology , Microscopy, Electron , Microscopy, Fluorescence , Time Factors
7.
Int Immunopharmacol ; 91: 107295, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360086

ABSTRACT

OBJECTIVES: Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a severe form of inflammatory lung disease. Its development and progression are regulated by cytokines. The purpose of this study was to determine the effects of HMGB1 involved in the regulation of Treg cells and IL-35. METHODS: A cecal ligation and puncture (CLP)-induced ALI model was used to investigate the changes in IL-35, Tregs, and the expression of RAGE and caspase-11 after HMGB1 inhibition (glycyrrhizin was used as an inhibitor of HMGB1). CD4+ naïve T cells sorted from C57BL/6 mice spleens were cultured to explore the role of HMGB1 in the differentiation from CD4+ naïve T cells to Tregs. RESULTS: HMGB1 promoted lung injury and uncontrolled inflammation in the CLP mouse model. HMGB1, NF-κB p65, RAGE, and caspase-11 expression in the lungs of CLP mice decreased significantly after pretreatment with glycyrrhizin. We found that the Treg proportion and IL-35 expression were upregulated in the serum and lung of CLP mice after inhibiting HMGB1. In our in vitro experiments, we found that recombinant HMGB1 significantly suppressed the proportion of CD4+CD25+FOXP3+Tregs differentiated from CD4+ naïve T cells. CONCLUSIONS: The inhibition of HMGB1 increased the proportion of Treg and expression of IL-35 and alleviated lung injury in the CLP-induced ALI model. Furthermore, inhibition of HMGB1 reduced caspase-11-dependent pyroptosis in the lungs of the CLP-induced ALI model.


Subject(s)
Acute Lung Injury/enzymology , CD4-Positive T-Lymphocytes/metabolism , Caspases, Initiator/metabolism , Cell Differentiation , HMGB1 Protein/metabolism , Interleukins/metabolism , Lung/enzymology , Pyroptosis , Respiratory Distress Syndrome/enzymology , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , Caspases, Initiator/genetics , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , HMGB1 Protein/genetics , Inflammation Mediators/metabolism , Interleukins/genetics , Lung/immunology , Lung/ultrastructure , Male , Mice, Inbred C57BL , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Signal Transduction
8.
Autophagy ; 16(2): 223-238, 2020 02.
Article in English | MEDLINE | ID: mdl-30982401

ABSTRACT

Obesity is associated with changes in the immune system that significantly hinder its ability to mount efficient immune responses. Previous studies have reported a dysregulation of immune responses caused by lipid challenge; however, the mechanisms underlying that dysregulation are still not completely understood. Autophagy is an essential catabolic process through which cellular components are degraded by the lysosomal machinery. In T cells, autophagy is an actively regulated process necessary to sustain homeostasis and activation. Here, we report that CD4+ T cell responses are inhibited when cells are challenged with increasing concentrations of fatty acids. Furthermore, analysis of T cells from diet-induced obese mice confirms that high lipid load inhibits activation-induced responses in T cells. We have found that autophagy is inhibited in CD4+ T cells exposed in vitro or in vivo to lipid stress, which causes decreased autophagosome formation and degradation. Supporting that inhibition of autophagy caused by high lipid load is a key mechanism that accounts for the effects on T cell function of lipid stress, we found that ATG7 (autophagy-related 7)-deficient T cells, unable to activate autophagy, did not show additional inhibitory effects on their responses to activation when subjected to lipid challenge. Our results indicate, thus, that increased lipid load can dysregulate autophagy and cause defective T cell responses, and suggest that inhibition of autophagy may underlie some of the characteristic obesity-associated defects in the T cell compartment.Abbreviations: ACTB: actin, beta; ATG: autophagy-related; CDKN1B: cyclin-dependent kinase inhibitor 1B; HFD: high-fat diet; IFNG: interferon gamma; IL: interleukin; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; MAPK8/JNK: mitogen-activated protein kinase 8; LC3-I: non-conjugated form of MAP1LC3B; LC3-II: phosphatidylethanolamine-conjugated form of MAP1LC3B; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MS: mass spectrometry; MTOR: mechanistic target of rapamycin kinase; NFATC2: nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 2; NLRP3: NLR family, pyrin domain containing 3; OA: oleic acid; PI: propidium iodide; ROS: reactive oxygen species; STAT5A: signal transducer and activator of transcription 5A; TCR: T cell receptor; TH1: T helper cell type 1.


Subject(s)
Autophagy/drug effects , Lipids/pharmacology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , Cell Proliferation/drug effects , Cytokines/metabolism , Diet, High-Fat , Down-Regulation/drug effects , Female , Homeostasis/drug effects , Humans , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Obesity/immunology , Oleic Acid/pharmacology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects
9.
Cytometry B Clin Cytom ; 98(3): 259-269, 2020 05.
Article in English | MEDLINE | ID: mdl-31571372

ABSTRACT

BACKGROUND: Several methods exist for flow-cytometric estimation of human peripheral blood CD4+ T regulatory cells (CD4+ Tregs). METHODS: We report our experience with the estimation of human CD4+ Tregs via three different characterizations using flow cytometry (CD25high FoxP3+ , CD25high CD127low/- FoxP3+ , and CD4+ CD25high/int CD45ROFoxP3+ ) in normal subjects. We have used these methods on the control populations from two studies (32 and 36 subjects, respectively), the latter two methods retrospectively on the subjects of the first study. The six CD4+ T cell fractions obtained by the third method were differentially colored to ascertain the distribution of these cell fractions in the CD25/FoxP3, CD45RO/FoxP3, and CD25/CD127 dot plots from CD4/CD25/CD45RO/FoxP3 and CD4/CD25/CD45RO/CD127 panels. RESULTS: Each approach gives significantly different estimates of Tregs (expressed as percentage of CD4+ T cells), with the second almost invariably yielding higher percentages than the other two. Only the third approach can distinguish among effector and naïve Tregs and FoxP3+ non-Tregs. Analysis of CD25/CD127 dot plots reveals that Treg delineation via the widely used definition of CD4+ CD25high CD127low/- cells unavoidably yields a mixture of nearly all effector and most of naïve Tregs, as well as FoxP3+ non-Tregs plus other cells. Delineation of effector/naïve Tregs and FoxP3+ non-Tregs is possible via CD45RO/CD25 dot plots but not by CD45RO/FoxP3 counterparts (as done previously) because of overlapping FoxP3 intensities among Tregs and non-Tregs. CONCLUSION: Our comparison shows that CD4/CD25/CD45RO/FoxP3 panels are an objective means of estimating effector and naïve Tregs via colored dot plots, aiding thus in Treg delineation in health and detecting aberrations in disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Leukocytes, Mononuclear/ultrastructure , T-Lymphocytes, Regulatory/immunology , Adult , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/ultrastructure , Female , Forkhead Transcription Factors/genetics , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Leukocyte Common Antigens/genetics , Leukocytes, Mononuclear/immunology , Male , T-Lymphocytes, Regulatory/ultrastructure
10.
Cell Rep ; 29(6): 1610-1620.e4, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31693899

ABSTRACT

CD4+ T cells play critical roles in directing immunity, both as T helper and as regulatory T (Treg) cells. Here, we demonstrate that hepatocytes can modulate T cell populations through engulfment of live CD4+ lymphocytes. We term this phenomenon enclysis to reflect the specific enclosure of CD4+ T cells in hepatocytes. Enclysis is selective for CD4+ but not CD8+ cells, independent of antigen-specific activation, and occurs in human hepatocytes in vitro, ex vivo, and in vivo. Intercellular adhesion molecule 1 (ICAM-1) facilitates T cell early adhesion and internalization, whereas hepatocytes form membrane lamellipodia or blebs to mediate engulfment. T cell internalization is unaffected by wortmannin and Rho kinase inhibition. Hepatocytes engulf Treg cells more efficiently than non-Treg cells, but Treg cell-containing vesicles preferentially acidify overnight. Thus, enclysis is a biological process with potential effects on immunomodulation and opens a new field for research to fully understand CD4+ T cell dynamics in liver inflammation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endocytosis/immunology , Endosomes/immunology , Hepatocytes/metabolism , Intercellular Adhesion Molecule-1/metabolism , T-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion/genetics , Cell Line , Endocytosis/genetics , Endosomes/genetics , Forkhead Transcription Factors/metabolism , Humans , Immune Tolerance , Intercellular Adhesion Molecule-1/genetics , Liver/immunology , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Microscopy, Electron, Scanning , Pinocytosis , T-Lymphocytes, Regulatory/ultrastructure , beta Catenin/genetics , beta Catenin/metabolism
12.
Cell Rep ; 27(1): 129-141.e4, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943396

ABSTRACT

Lymphocyte activation gene-3 (LAG-3) is an inhibitory receptor expressed by CD4+ T cells and tempers their homeostatic expansion. Because CD4+ T cell proliferation is tightly coupled to bioenergetics, we investigate the role of LAG-3 in modulating naive CD4+ T cell metabolism. LAG-3 deficiency enhances the metabolic profile of naive CD4+ T cells by elevating levels of mitochondrial biogenesis. In vivo, LAG-3 blockade partially restores expansion and the metabolic phenotype of wild-type CD4+ T cells to levels of Lag3-/- CD4+ T cells, solidifying that LAG-3 controls these processes. Lag3-/- CD4+ T cells also demonstrate greater signal transducer and activator of transcription 5 (STAT5) activation, enabling resistance to interleukin-7 (IL-7) deprivation. These results implicate this pathway as a target of LAG-3-mediated inhibition. Additionally, enhancement of STAT5 activation, as a result of LAG-3 deficiency, contributes to greater activation potential in these cells. These results identify an additional mode of regulation elicited by LAG-3 in controlling CD4+ T cell responses.


Subject(s)
Antigens, CD/physiology , CD4-Positive T-Lymphocytes , Energy Metabolism/genetics , Mitochondria/physiology , Organelle Biogenesis , Animals , Antigens, CD/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/ultrastructure , Cells, Cultured , Female , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Resting Phase, Cell Cycle/genetics , Lymphocyte Activation Gene 3 Protein
14.
Nat Commun ; 9(1): 3630, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30194420

ABSTRACT

Microvilli on T cells have been proposed to survey surfaces of antigen-presenting cells (APC) or facilitate adhesion under flow; however, whether they serve essential functions during T cell activation remains unclear. Here we show that antigen-specific T cells deposit membrane particles derived from microvilli onto the surface of cognate antigen-bearing APCs. Microvilli carry T cell receptors (TCR) at all stages of T cell activation and are released as large TCR-enriched, T cell microvilli particles (TMP) in a process of trogocytosis. These microvilli exclusively contain protein arrestin-domain-containing protein 1, which is directly involved in membrane budding and, in combination with vacuolar protein-sorting-associated protein 4, transforms large TMPs into smaller, exosome-sized TMPs. Notably, TMPs from CD4+ T cells are enriched with LFA-2/CD2 and various cytokines involved in activating dendritic cells. Collectively, these results demonstrate that T cell microvilli constitute "immunological synaptosomes" that carry T cell messages to APCs.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Microvilli/physiology , Animals , Antigen-Presenting Cells , CD4-Positive T-Lymphocytes/ultrastructure , Cell-Derived Microparticles/physiology , Dendritic Cells/physiology , HEK293 Cells , Humans , Jurkat Cells , Mice , Receptors, Antigen, T-Cell/metabolism , Synaptosomes
15.
Sci Signal ; 11(545)2018 08 28.
Article in English | MEDLINE | ID: mdl-30154103

ABSTRACT

The cytokine receptor subunit γc provides critical signals for T cell survival and differentiation. We investigated the molecular mechanism that controls the cell surface abundance of γc during T cell development in the thymus. We found that the amount of γc was low on CD4+CD8+ double-positive (DP) thymocytes before their positive selection to become mature T cells. The transcription factor RORγt was abundant in immature DP thymocytes, and its loss resulted in an increase in the abundance of surface γc, specifically on preselection DP cells. Rather than directly repressing expression of the gene encoding γc, RORγt acted through the antiapoptotic protein Bcl-xL to reduce the abundance of surface γc, which resulted in decreased cytokine signaling and was associated with inhibition of cell metabolism and mitochondrial biogenesis. Accordingly, overexpression of Bcl-xL in RORγt-deficient thymocytes restored the amount of surface γc to that present on normal preselection DP cells. Together, these data highlight a previously unappreciated role for RORγt and Bcl-xL in limiting γc abundance at the cell surface and reveal a signaling circuit in which survival factors control cytokine signaling by limiting the abundance and surface distribution of a receptor subunit shared by several cytokines.


Subject(s)
Interleukin Receptor Common gamma Subunit/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Thymocytes/immunology , bcl-X Protein/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/ultrastructure , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/ultrastructure , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression/immunology , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Thymocytes/metabolism , Thymocytes/ultrastructure , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
16.
Exp Mol Pathol ; 105(1): 150-152, 2018 08.
Article in English | MEDLINE | ID: mdl-30009774

ABSTRACT

The nature of the immunologic synapse in autoimmune hepatitis is defined. This process involves the T cell receptor (TCR) which binds to the hepatocyte antigen presenting major histocompatibility complex (MHC) on the plasma membrane. This complex is quickly removed from the liver cell and taken into the T cell cytoplasm to be digested by the lysosome. The liver cell is gradually diminished to the point of its total removal by the lymphocytes binding to it.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis, Autoimmune/pathology , Histocompatibility Antigens Class II/immunology , Immunological Synapses/immunology , Receptors, Antigen, T-Cell/immunology , CD4-Positive T-Lymphocytes/ultrastructure , Hepatitis, Autoimmune/immunology , Humans , Necrosis
17.
Mol Ther ; 26(9): 2107-2118, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30017878

ABSTRACT

Extracellular vesicles (EVs) play a major role in cell-to-cell communication in physiological and pathological conditions, and their manipulation may represent a promising therapeutic strategy. Microglia, the parenchymal mononuclear phagocytes of the brain, modulate neighboring cells also through the release of EVs. The production of custom EVs filled with desired molecules, possibly targeted to make their uptake cell specific, and their administration in biological fluids may represent a valid approach for drug delivery. We engineered a murine microglia cell line, BV-2, to release EVs overexpressing the endogenous "eat me" signal Lactadherin (Mfg-e8) on the surface to target phagocytes and containing the anti-inflammatory cytokine IL-4. A single injection of 107 IL-4+Mfg-e8+ EVs into the cisterna magna modulated established neuroinflammation and significantly reduced clinical signs in the mouse model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Injected IL-4+Mfg-e8+ EVs target mainly phagocytes (i.e., macrophages and microglia) surrounding liquoral spaces, and their cargo promote the upregulation of anti-inflammatory markers chitinase 3-like 3 (ym1) and arginase-1 (arg1), significantly reducing tissue damage. Engineered EVs may represent a biological drug delivery tool able to deliver multiple functional molecules simultaneously to treat neuroinflammatory diseases.


Subject(s)
Extracellular Vesicles/metabolism , Interleukin-4/metabolism , Multiple Sclerosis/metabolism , Animals , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/ultrastructure , Cell Line , Cells, Cultured , Disease Models, Animal , Extracellular Vesicles/ultrastructure , Female , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Rats , Reverse Transcriptase Polymerase Chain Reaction
18.
Br J Nutr ; 119(2): 163-175, 2018 01.
Article in English | MEDLINE | ID: mdl-29249211

ABSTRACT

Cell membrane fatty acids influence fundamental properties of the plasma membrane, including membrane fluidity, protein functionality, and lipid raft signalling. Evidence suggests that dietary n-3 PUFA may target the plasma membrane of immune cells by altering plasma membrane lipid dynamics, thereby regulating the attenuation of immune cell activation and suppression of inflammation. As lipid-based immunotherapy might be a promising new clinical strategy for the treatment of inflammatory disorders, we conducted in vitro and in vivo experiments to examine the effects of n-3 PUFA on CD4+ T cell membrane order, mitochondrial bioenergetics and lymphoproliferation. n-3 PUFA were incorporated into human primary CD4+ T cells phospholipids in vitro in a dose-dependent manner, resulting in a reduction in whole cell membrane order, oxidative phosphorylation and proliferation. At higher doses, n-3 PUFA induced unique phase separation in T cell-derived giant plasma membrane vesicles. Similarly, in a short-term human pilot study, supplementation of fish oil (4 g n-3 PUFA/d) for 6 weeks in healthy subjects significantly elevated EPA (20 : 5n-3) levels in CD4+ T cell membrane phospholipids, and reduced membrane lipid order. These results demonstrate that the dynamic reshaping of human CD4+ T cell plasma membrane organisation by n-3 PUFA may modulate down-stream clonal expansion.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , Cell Membrane/drug effects , Fatty Acids, Omega-3/pharmacology , Aged , Aged, 80 and over , Cell Membrane/chemistry , Cell Membrane/physiology , Dietary Fats/administration & dosage , Dietary Supplements , Eicosapentaenoic Acid/blood , Energy Metabolism/drug effects , Fatty Acids/blood , Female , Fish Oils/administration & dosage , Humans , Lymphocyte Activation/drug effects , Male , Membrane Lipids/blood , Membrane Lipids/chemistry , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Phospholipids/blood , Phospholipids/chemistry , Pilot Projects
19.
mBio ; 8(5)2017 09 19.
Article in English | MEDLINE | ID: mdl-28928214

ABSTRACT

The indomitable aspect of HIV-1 infection is not that HIV-1 proviral DNA is integrated into host DNA but that it can also turn itself off, remaining invisible to drug or immune surveillance. Thus, the goals of eradication include ways to precisely excise HIV-1 DNA or wake up the silent HIV-1 provirus and eliminate the infected cells thus identified. Methods to identify and fish out the latently infected cells or to delineate their characteristics are being rapidly developed. In 2016, Baxter et al. (A. E. Baxter, J. Niessl, R. Fromentin, J. Richard, F. Porichis, R. Charlebois, M. Massanella, N. Brassard, N. Alsahafi, G. G. Delgado, J. P. Routy, B. D. Walker, A. Finzi, N. Chomont, and D. E. Kaufmann, Cell Host Microbe 20:368-380, 2016, https://doi.org/10.1016/j.chom.2016.07.015) and Martrus et al. (G. Martrus, A. Niehrs, R. Cornelis, A. Rechtien, W. García-Beltran, M. Lütgehetmann, C. Hoffmann, and M. Altfeld, J Virol 90:9018-9028, 2016, https://doi.org/10.1128/JVI.01448-16) reported using the fluorescence in situ hybridization-flow cytometry technique to identify and quantify cells expressing HIV-1 RNA and Gag protein, as well as bearing unique cell surface markers. In a recent article in mBio, Grau-Expósito et al. (J. Grau-Expósito, C. Serra-Peinado, L. Miguel, J. Navarro, A. Curran, J. Burgos, I. Ocaña, E. Ribera, A. Torrella, B. Planas, R. Badía, J. Castellví, V. Falcó, M. Crespo, and M. J. Buzon, mBio 8:e00876-17, 2017, https://doi.org/10.1128/mBio.00876-17) reported a similar method that they claim to be more sensitive. With these methods, researchers are one step closer to measuring latent reservoirs and eliminating critical barriers to HIV eradication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/virology , HIV-1/physiology , In Situ Hybridization, Fluorescence/methods , Virus Latency , Animals , CD4-Positive T-Lymphocytes/ultrastructure , Cell Count , HIV Infections/diagnosis , HIV-1/genetics , HIV-1/ultrastructure , Humans , Proviruses/physiology , Proviruses/ultrastructure , RNA, Viral/analysis
20.
AIDS Res Ther ; 14(1): 40, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-28893281

ABSTRACT

Despite the tremendous success of anti-retroviral therapy (ART) no current treatment can eradicate latent HIV reservoirs from HIV-infected individuals or generate, effective HIV-specific immunity. Technological limitations have hampered the identification and characterization of both HIV-infected cells and HIV-specific responses in clinical samples directly ex vivo. RNA-flow cytometric fluorescence in situ hybridisation (RNA Flow-FISH) is a powerful technique, which enables detection of mRNAs in conjunction with proteins at a single-cell level. Here, we describe how we are using this technology to address some of the major questions remaining in the HIV field in the era of ART. We discuss how CD4 T cell responses to HIV antigens, both following vaccination and HIV infection, can be characterized by measurement of cytokine mRNAs. We describe how our development of a dual HIV mRNA/protein assay (HIVRNA/Gag assay) enables high sensitivity detection of very rare HIV-infected cells and aids investigations into the translation-competent latent reservoir in the context of HIV cure.


Subject(s)
Flow Cytometry/methods , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/immunology , In Situ Hybridization, Fluorescence/methods , RNA/analysis , AIDS Vaccines , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/ultrastructure , CD4-Positive T-Lymphocytes/virology , Cytokines/genetics , Cytokines/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Vaccination , Virus Latency
SELECTION OF CITATIONS
SEARCH DETAIL
...