Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
Nature ; 599(7883): 147-151, 2021 11.
Article in English | MEDLINE | ID: mdl-34616045

ABSTRACT

Understanding cellular architecture is essential for understanding biology. Electron microscopy (EM) uniquely visualizes cellular structures with nanometre resolution. However, traditional methods, such as thin-section EM or EM tomography, have limitations in that they visualize only a single slice or a relatively small volume of the cell, respectively. Focused ion beam-scanning electron microscopy (FIB-SEM) has demonstrated the ability to image small volumes of cellular samples with 4-nm isotropic voxels1. Owing to advances in the precision and stability of FIB milling, together with enhanced signal detection and faster SEM scanning, we have increased the volume that can be imaged with 4-nm voxels by two orders of magnitude. Here we present a volume EM atlas at such resolution comprising ten three-dimensional datasets for whole cells and tissues, including cancer cells, immune cells, mouse pancreatic islets and Drosophila neural tissues. These open access data (via OpenOrganelle2) represent the foundation of a field of high-resolution whole-cell volume EM and subsequent analyses, and we invite researchers to explore this atlas and pose questions.


Subject(s)
Datasets as Topic , Information Dissemination , Microscopy, Electron, Scanning , Organelles/ultrastructure , Animals , Cell Line , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Interphase , Islets of Langerhans/cytology , Male , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Scanning/standards , Microtubules/ultrastructure , Neuroglia/ultrastructure , Neurons/ultrastructure , Open Access Publishing , Ovarian Neoplasms/immunology , Ovarian Neoplasms/ultrastructure , Ribosomes/ultrastructure , Synaptic Vesicles/ultrastructure , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
2.
Cell Cycle ; 17(10): 1220-1234, 2018.
Article in English | MEDLINE | ID: mdl-30019620

ABSTRACT

Dendritic cells (DCs) play a predominant role in initiating cell immune responses. Here we generated a DC-targeting lentiviral vector (LVDC-UbHBcAg-LIGHT) and evaluated its capacity to elicit HBV-specific cytotoxic T lymphocyte (CTL) responses. DC-SIGN-mediated specific transduction using this construct was confirmed in DC-SIGN-expressing 293T cells and ex vivo-cultured bone marrow cells. LVDC-UbHBcAg-LIGHT-loaded DCs were highly effective in inducing HBV-specific CTLs. Mechanistic studies demonstrated autophagy blocking led to a significant increase in apoptosis and obvious inhibition of CD8 + T cells entry into S-phase, correspondingly attenuated LVDC-UbHBcAg-LIGHT-loaded DC-induced T cell responses. This observation was supported by accumulation of pro-apoptotic proteins and the main negative cell cycle regulator-CDKN1B that otherwise would be degraded in activated T cells where autophagy preferentially occured. Our findings revealed an important role of autophagy in the activation of T cells and suggested LVDC-UbHBcAg-LIGHT may potentially be used as a therapeutic strategy to combat persistent HBV infection with higher security.


Subject(s)
Autophagy , Dendritic Cells/metabolism , Genetic Vectors/metabolism , Hepatitis B virus/immunology , Lentivirus/genetics , T-Lymphocytes, Cytotoxic/immunology , Transduction, Genetic , Up-Regulation , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion Molecules/metabolism , Dendritic Cells/drug effects , Genetic Engineering , Hepatitis B Core Antigens/metabolism , Humans , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Receptors, Cell Surface/metabolism , S Phase/drug effects , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/ultrastructure , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Ubiquitin/metabolism , Up-Regulation/drug effects
3.
Eur J Immunol ; 47(9): 1562-1572, 2017 09.
Article in English | MEDLINE | ID: mdl-28688208

ABSTRACT

The actin-binding protein profilin1 (PFN1) plays a central role in actin dynamics, which is essential for cytotoxic T lymphocyte (CTL) functions. The functional role of PFN1 in CTLs, however still remains elusive. Here, we identify PFN1 as the only member of the profilin family expressed in primary human CD8+ T cells. Using in vitro assays, we find that PFN1 is a negative regulator of CTL-mediated elimination of target cells. Furthermore, PFN1 is involved in activation-induced lytic granule (LG) release, CTL migration and modulation of actin structures at the immunological synapse (IS). During CTL migration, PFN1 modulates the velocity, protrusion formation patterns and protrusion sustainability. In contrast, PFN1 does not significantly affect migration persistence and the rates of protrusion emergence and retraction. Under in vitro conditions mimicking a tumor microenvironment, we show that PFN1 downregulation promotes CTL invasion into a 3D matrix, without affecting the viability of CTLs in a hydrogen peroxide-enriched microenvironment. Highlighting its potential relevance in cancer, we find that in pancreatic cancer patients, PFN1 expression is substantially decreased in peripheral CD8+ T cells. Taken together, we conclude that PFN1 is a negative regulator for CTL-mediated cytotoxicity and may have an impact on CTL functionality in a tumor-related context.


Subject(s)
Cell Movement , Cell Surface Extensions/ultrastructure , Extracellular Matrix/metabolism , Immunological Synapses/ultrastructure , Pancreatic Neoplasms/immunology , Profilins/metabolism , T-Lymphocytes, Cytotoxic/immunology , Actin Cytoskeleton/ultrastructure , CD8 Antigens/metabolism , Cells, Cultured , Cytotoxicity, Immunologic , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Hydrogen Peroxide/metabolism , Lymphocyte Activation , Pancreatic Neoplasms/genetics , Profilins/immunology , T-Lymphocytes, Cytotoxic/ultrastructure , Tumor Microenvironment
4.
Vet Immunol Immunopathol ; 177: 24-34, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27436441

ABSTRACT

Feline large granular lymphocyte lymphomas are rare but very aggressive tumors with a poor prognosis. In this study, a cell line from an abdominal effusion of a cat with large granular lymphoma was characterized. Immunophenotype staining was positive for CD3 and CD45R, and negative for CD4, CD8, CD56, CD79α, BLA.36 and NK1. A TCR γ gene rearrangement was detectable by PARR. Neither FeLV antigen nor exogenous FeLV provirus could be detected. A chromosomal instability associated with a centrosome hyperamplification could also be determined. The cell line is able to lyse target cells without antigen presentation or interaction with antigen presenting cells. Therefore, these cells were classified as genetically instable non-MHC-restricted cytotoxic T cells with large granular lymphocyte morphology.


Subject(s)
Cat Diseases/genetics , Cat Diseases/immunology , Cats/genetics , Cats/immunology , Lymphoma/veterinary , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Genes, p53 , Genomic Instability , Immunophenotyping , Leukemia Virus, Feline/isolation & purification , Lymphoma/genetics , Lymphoma/immunology , Microscopy, Electron, Transmission , RNA, Messenger/genetics , T-Lymphocytes, Cytotoxic/ultrastructure
5.
Nat Rev Immunol ; 16(7): 421-32, 2016 07.
Article in English | MEDLINE | ID: mdl-27265595

ABSTRACT

Cytotoxic T lymphocytes (CTLs) kill virus-infected and tumour cells with remarkable specificity. Upon recognition, CTLs form a cytolytic immune synapse with their target cell, and marked reorganization of both the actin and the microtubule cytoskeletons brings the centrosome up to the plasma membrane to the point of T cell receptor signalling. Secretory granules move towards the centrosome and are delivered to this focal point of secretion. Such centrosomal docking at the plasma membrane also occurs during ciliogenesis; indeed, striking similarities exist between the cytolytic synapse and the primary cilium that throw light on the possible origins of immune synapses.


Subject(s)
Cytotoxicity, Immunologic/immunology , Immunological Synapses/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Humans , Immunological Synapses/ultrastructure , T-Lymphocytes, Cytotoxic/ultrastructure
6.
Curr Biol ; 25(24): 3239-44, 2015 Dec 21.
Article in English | MEDLINE | ID: mdl-26670998

ABSTRACT

Cytotoxic T lymphocytes (CTLs) are highly effective serial killers capable of destroying virally infected and cancerous targets by polarized release from secretory lysosomes. Upon target contact, the CTL centrosome rapidly moves to the immunological synapse, focusing microtubule-directed release at this point [1-3]. Striking similarities have been noted between centrosome polarization at the synapse and basal body docking during ciliogenesis [1, 4-8], suggesting that CTL centrosomes might dock with the plasma membrane during killing, in a manner analogous to primary cilia formation [1, 4]. However, questions remain regarding the extent and function of centrosome polarization at the synapse, and recent reports have challenged its role [9, 10]. Here, we use high-resolution transmission electron microscopy (TEM) tomography analysis to show that, as in ciliogenesis, the distal appendages of the CTL mother centriole contact the plasma membrane directly during synapse formation. This is functionally important as small interfering RNA (siRNA) targeting of the distal appendage protein, Cep83, required for membrane contact during ciliogenesis [11], impairs CTL secretion. Furthermore, the regulatory proteins CP110 and Cep97, which must dissociate from the mother centriole to allow cilia formation [12], remain associated with the mother centriole in CTLs, and neither axoneme nor transition zone ciliary structures form. Moreover, complete centrosome docking can occur in proliferating CTLs with multiple centriole pairs. Thus, in CTLs, centrosomes dock transiently with the membrane, within the cell cycle and without progression into ciliogenesis. We propose that this transient centrosome docking without cilia formation is important for CTLs to deliver rapid, repeated polarized secretion directed by the centrosome.


Subject(s)
Centrioles/physiology , Cilia/physiology , Immunological Synapses/physiology , T-Lymphocytes, Cytotoxic/physiology , Animals , Cells, Cultured , Mice, Inbred C57BL , Microscopy, Electron, Transmission , T-Lymphocytes, Cytotoxic/ultrastructure
7.
J Neurooncol ; 125(1): 23-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26290143

ABSTRACT

Malignant glioma is among the most challenging of all cancers to treat successfully. Despite recent advances in surgery, radiotherapy and chemotherapy, current treatment regimens have only a marginal impact on patient survival. In this study, we constructed a novel nanoparticle containing neuritin peptide with grp170. The nanoparticle could elicit a neuritin-specific cytotoxic T lymphocyte response to lyse glioma cells in vitro. In addition, the nanoparticle could inhibit tumor growth and improve the lifespan of tumor-bearing mice in vivo. Taken together, the results demonstrated that the nanoparticle can inhibit tumor growth and represents a promising therapy for glioma.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Glycoproteins/therapeutic use , HSP70 Heat-Shock Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Animals , Brain Neoplasms/immunology , Cell Line, Tumor , Chromium/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glioma/immunology , Humans , Lymphocyte Activation/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Microscopy, Electron, Transmission , Nanoparticles/administration & dosage , Nanoparticles/ultrastructure , Peptides/therapeutic use , T-Lymphocytes, Cytotoxic/ultrastructure
8.
PLoS One ; 10(8): e0135994, 2015.
Article in English | MEDLINE | ID: mdl-26296096

ABSTRACT

Killing of virally infected cells or tumor cells by cytotoxic T lymphocytes requires targeting of lytic granules to the junction between the CTL and its target. We used whole-cell patch clamp to measure the cell capacitance at fixed intracellular [Ca2+] to study fusion of lytic granules in human CTLs. Expression of a fluorescently labeled human granzyme B construct allowed identification of lytic granule fusion using total internal reflection fluorescence microscopy. In this way capacitance steps due to lytic granule fusion were identified. Our goal was to determine the size of fusing lytic granules and to describe their behavior at the plasma membrane. On average, 5.02 ± 3.09 (mean ± s.d.) lytic granules were released per CTL. The amplitude of lytic granule fusion events was ~ 3.3 fF consistent with a diameter of about 325 nm. Fusion latency was biphasic with time constants of 15.9 and 106 seconds. The dwell time of fusing lytic granules was exponentially distributed with a mean dwell time of 28.5 seconds. Fusion ended in spite of the continued presence of granules at the immune synapse. The mobility of fusing granules at the membrane was indistinguishable from that of lytic granules which failed to fuse. While dwelling at the plasma membrane lytic granules exhibit mobility consistent with docking interspersed with short periods of greater mobility. The failure of lytic granules to fuse when visible in TIRF at the membrane may indicate that a membrane-confined reaction is rate limiting.


Subject(s)
Cell Membrane/metabolism , Cytoplasmic Granules/metabolism , Immunological Synapses/metabolism , Membrane Fusion/immunology , T-Lymphocytes, Cytotoxic/metabolism , Calcium/immunology , Calcium/metabolism , Cell Membrane/immunology , Cell Membrane/ultrastructure , Cytoplasmic Granules/immunology , Cytoplasmic Granules/ultrastructure , Cytotoxicity, Immunologic , Electric Capacitance , Electroporation , Exocytosis , Gene Expression , Granzymes/genetics , Granzymes/immunology , Granzymes/metabolism , Humans , Immunological Synapses/immunology , Immunological Synapses/ultrastructure , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Primary Cell Culture , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure , Time Factors , Red Fluorescent Protein
9.
Gastroenterology ; 149(3): 681-91.e10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26001928

ABSTRACT

BACKGROUND & AIMS: The mechanisms of tissue destruction during progression of celiac disease are poorly defined. It is not clear how tissue stress and adaptive immunity contribute to the activation of intraepithelial cytotoxic T cells and the development of villous atrophy. We analyzed epithelial cells and intraepithelial cytotoxic T cells in family members of patients with celiac disease, who were without any signs of adaptive antigluten immunity, and in potential celiac disease patients, who have antibodies against tissue transglutaminase 2 in the absence of villous atrophy. METHODS: We collected blood and intestinal biopsy specimens from 268 patients at tertiary medical centers in the United States and Italy from 2004 to 2012. All subjects had normal small intestinal histology. Study groups included healthy individuals with no family history of celiac disease or antibodies against tissue transglutaminase 2 (controls), healthy family members of patients with celiac disease, and potential celiac disease patients. Intraepithelial cytotoxic T cells were isolated and levels of inhibitory and activating natural killer (NK) cells were measured by flow cytometry. Levels of heat shock protein (HSP) and interleukin 15 were measured by immunohistochemistry, and ultrastructural alterations in intestinal epithelial cells (IECs) were assessed by electron microscopy. RESULTS: IECs from subjects with a family history of celiac disease, but not from subjects who already had immunity to gluten, expressed higher levels of HS27, HSP70, and interleukin-15 than controls; their IECs also had ultrastructural alterations. Intraepithelial cytotoxic T cells from relatives of patients with celiac disease expressed higher levels of activating NK receptors than cells from controls, although at lower levels than patients with active celiac disease, and without loss of inhibitory receptors for NK cells. Intraepithelial cytotoxic T cells from potential celiac disease patients failed to up-regulate activating NK receptors. CONCLUSIONS: A significant subset of healthy family members of patients with celiac disease with normal intestinal architecture had epithelial alterations, detectable by immunohistochemistry and electron microscopy. The adaptive immune response to gluten appears to act in synergy with epithelial stress to allow intraepithelial cytotoxic T cells to kill epithelial cells and induce villous atrophy in patients with active celiac disease.


Subject(s)
Adaptive Immunity , Celiac Disease/immunology , Cell Communication , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Intestine, Small/immunology , Stress, Physiological , T-Lymphocytes, Cytotoxic/immunology , Autoantibodies/blood , Case-Control Studies , Celiac Disease/blood , Celiac Disease/pathology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , GTP-Binding Proteins/immunology , HSP27 Heat-Shock Proteins/immunology , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Interleukin-15/immunology , Interleukin-15/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Italy , Molecular Chaperones , Phenotype , Protein Glutamine gamma Glutamyltransferase 2 , Risk Factors , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/ultrastructure , Transglutaminases/immunology , United States
11.
Neurobiol Dis ; 59: 194-205, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23899663

ABSTRACT

Axon injury is a central determinant of irreversible neurological deficit and disease progression in patients with multiple sclerosis (MS). CD8(+) lymphocytes (CTLs) within inflammatory demyelinated MS lesions correlate with acute axon injury and neurological deficits. The mechanisms of these correlations are unknown. We interrogated CTL-mediated axon injury using the transgenic OT-I antigen-specific CTL model system in conjunction with a chambered cortical neuron culture platform that permitted the isolated manipulation of axons independent of neuron cell bodies and glia. Interferon gamma upregulated, through a dose dependent mechanism, the axonal expression of functional major histocompatibility complex class I (MHC I) molecules competent to present immunologically-relevant antigens derived from endogenously expressed proteins. Antigen-specific CTLs formed cytotoxic immune synapses with and directly injured axons expressing antigen-loaded MHC I molecules. CTL-mediated axon injury was mechanistically dependent upon axonal MHC I antigen presentation, T cell receptor specificity and axoplasmic granzyme B activity. Despite extensive distal CTL-mediated axon injury, acute neuron cell body apoptosis was not observed. These findings present a novel model of immune-mediated axon injury and offer anti-axonal CTLs and granzyme B as targets for the therapeutic protection of axons and prevention of neurological deficits in MS patients.


Subject(s)
Axons/metabolism , CD8 Antigens/metabolism , Granzymes/metabolism , Histocompatibility Antigens Class I/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Axons/pathology , Axons/ultrastructure , Cerebral Cortex/cytology , Embryo, Mammalian , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Granzymes/genetics , Histocompatibility Antigens Class I/genetics , Interferon-gamma/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuroglia/cytology , Neuroglia/ultrastructure , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Ovalbumin/genetics , Ovalbumin/metabolism , Ovalbumin/pharmacology , Peptide Fragments/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
12.
Nat Commun ; 4: 1439, 2013.
Article in English | MEDLINE | ID: mdl-23385584

ABSTRACT

Cytotoxic T lymphocytes kill virus-infected and tumorigenic target cells through the release of perforin and granzymes via fusion of lytic granules at the contact site, the immunological synapse. It has been postulated that this fusion process is mediated by non-neuronal members of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor complex protein family. Here, using a synaptobrevin2-monomeric red fluorescence protein knock-in mouse we demonstrate that, surprisingly, the major neuronal v-SNARE synaptobrevin2 is expressed in cytotoxic T lymphocytes and exclusively localized on granzyme B-containing lytic granules. Cleavage of synaptobrevin2 by tetanus toxin or ablation of the synaptobrevin2 gene leads to a complete block of lytic granule exocytosis while leaving upstream events unaffected, identifying synaptobrevin2 as the v-SNARE responsible for the fusion of lytic granules at the immunological synapse.


Subject(s)
Cytoplasmic Granules/metabolism , Cytotoxicity, Immunologic , Membrane Fusion , SNARE Proteins/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Animals , Blotting, Western , Cell Degranulation/drug effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/ultrastructure , Cytotoxicity, Immunologic/drug effects , Flow Cytometry , Gene Knock-In Techniques , Green Fluorescent Proteins/metabolism , Immunological Synapses/drug effects , Immunological Synapses/metabolism , Membrane Fusion/drug effects , Mice , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/ultrastructure , Tetanus Toxin/pharmacology
13.
Eur J Immunol ; 42(8): 2132-41, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22736282

ABSTRACT

Cytotoxic T lymphocytes (CTLs) kill tumorigenic and virally infected cells by targeted secretion of lytic granule contents. The precise point at which secretion occurs is directed by the centrosome docking at the immunological synapse (IS). The centrosome is highly dynamic in CTLs, lagging behind the nucleus in the uropod of migrating CTLs, but translocating across the entire length of the cell to dock at the IS when a target cell is recognized. While in most cell types, the centrosome is always closely associated with the nuclear membrane, in CTLs, it often appears to be dissociated from the nucleus, both in migrating cells and when forming an IS. We asked whether this dissociation is required for CTL killing, by expressing GFP-BICD2-NT-nesprin-3, which tethers the centrosome to the nucleus irreversibly. Immunofluorescence microscopy revealed that the centrosome polarized successfully to the central supramolecular activation complex (cSMAC) of the synapse in GFP-BICD2-NT-nesprin-3-expressing CTLs, with the centrosome and nucleus migrating together to the IS. CTLs in which the centrosome was "glued" to the nucleus were able to dock and release granules at the IS as effectively as mock-treated cells. These data demonstrate that CTL cytotoxicity is independent of centrosomal dissociation from the nuclear envelope.


Subject(s)
Cell Nucleus/metabolism , Centrosome/metabolism , Cytoplasmic Granules/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Cell Polarity , Cells, Cultured , Gene Transfer Techniques , Immunological Synapses , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Nuclear Envelope , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , T-Lymphocytes, Cytotoxic/ultrastructure
14.
J Immunol ; 187(11): 5824-33, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22043013

ABSTRACT

The cytoskeletal adaptor protein paxillin localizes to the microtubule organizing center (MTOC) in T cells and, upon target cell binding, is recruited to the supramolecular activation complex (SMAC). We mapped the region of paxillin that associates with both the MTOC and SMAC to the leucine-aspartic acid (LD) domains and showed that a protein segment containing LD2-4 was sufficient for MTOC and SMAC recruitment. Examination of the localization of paxillin at the SMAC revealed that paxillin localizes to the peripheral area of the SMAC along with LFA-1, suggesting that LFA-1 may contribute to its recruitment. LFA-1 or CD3 engagement alone was insufficient for paxillin recruitment because there was no paxillin accumulation at the site of CTL contact with anti-LFA-1- or anti-CD3-coated beads. In contrast, paxillin accumulation was detected when beads coated with both anti-CD3 and anti-LFA-1 were bound to CTL, suggesting that signals from both the TCR and LFA-1 are required for paxillin accumulation. Paxillin was shown to be phosphorylated downstream of ERK, but when we generated a mutation (S83A/S130A) that abolished the mobility shift as a result of phosphorylation, we found that paxillin still bound to the MTOC and was recruited to the SMAC. Furthermore, ERK was not absolutely required for MTOC reorientation in CTL that require ERK for killing. Finally, expression of the LD2-4 region of paxillin substantially reduced MTOC reorientation. These studies demonstrated that paxillin is recruited, through its LD domains, to sites of integrin engagement and may contribute to MTOC reorientation required for directional degranulation.


Subject(s)
Lymphocyte Activation/physiology , Microtubule-Organizing Center/metabolism , Microtubules/metabolism , Paxillin/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Blotting, Western , Cell Degranulation/physiology , Immunological Synapses/chemistry , Immunological Synapses/immunology , Immunological Synapses/metabolism , Immunoprecipitation , Leucine/chemistry , Leucine/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microtubule-Organizing Center/chemistry , Microtubule-Organizing Center/ultrastructure , Mutagenesis, Site-Directed , Paxillin/chemistry , Paxillin/immunology , Polymerase Chain Reaction , Protein Structure, Tertiary , Protein Transport/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
15.
BMC Biol ; 9: 45, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21711522

ABSTRACT

BACKGROUND: Cytolytic cells of the immune system destroy pathogen-infected cells by polarised exocytosis of secretory lysosomes containing the pore-forming protein perforin. Precise delivery of this lethal hit is essential to ensuring that only the target cell is destroyed. In cytotoxic T lymphocytes (CTLs), this is accomplished by an unusual movement of the centrosome to contact the plasma membrane at the centre of the immunological synapse formed between killer and target cells. Secretory lysosomes are directed towards the centrosome along microtubules and delivered precisely to the point of target cell recognition within the immunological synapse, identified by the centrosome. We asked whether this mechanism of directing secretory lysosome release is unique to CTL or whether natural killer (NK) and invariant NKT (iNKT) cytolytic cells of the innate immune system use a similar mechanism to focus perforin-bearing lysosome release. RESULTS: NK cells were conjugated with B-cell targets lacking major histocompatibility complex class I 721.221 cells, and iNKT cells were conjugated with glycolipid-pulsed CD1-bearing targets, then prepared for thin-section electron microscopy. High-resolution electron micrographs of the immunological synapse formed between NK and iNKT cytolytic cells with their targets revealed that in both NK and iNKT cells, the centrioles could be found associated (or 'docked') with the plasma membrane within the immunological synapse. Secretory clefts were visible within the synapses formed by both NK and iNKT cells, and secretory lysosomes were polarised along microtubules leading towards the docked centrosome. The Golgi apparatus and recycling endosomes were also polarised towards the centrosome at the plasma membrane within the synapse. CONCLUSIONS: These results reveal that, like CTLs of the adaptive immune system, the centrosomes of NK and iNKT cells (cytolytic cells of the innate immune system) direct secretory lysosomes to the immunological synapse. Morphologically, the overall structure of the immunological synapses formed by NK and iNKT cells are very similar to those formed by CTLs, with both exocytic and endocytic organelles polarised towards the centrosome at the plasma membrane, which forms a focal point for exocytosis and endocytosis within the immunological synapse. We conclude that centrosomal polarisation provides a rapid, responsive and precise mechanism for secretory lysosome delivery to the immunological synapse in CTLs, NK cells and iNKT cells.


Subject(s)
Centrioles/metabolism , Centrioles/ultrastructure , Immune System/metabolism , Immunological Synapses/metabolism , Immunological Synapses/ultrastructure , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/ultrastructure , Adaptive Immunity/physiology , Cells, Cultured , Humans , Immune System/ultrastructure , Microscopy, Electron, Transmission
16.
J Cell Biol ; 192(4): 663-74, 2011 Feb 21.
Article in English | MEDLINE | ID: mdl-21339332

ABSTRACT

Docking of the centrosome at the plasma membrane directs lytic granules to the immunological synapse. To identify signals controlling centrosome docking at the synapse, we have studied cytotoxic T lymphocytes (CTLs) in which expression of the T cell receptor-activated tyrosine kinase Lck is ablated. In the absence of Lck, the centrosome is able to translocate around the nucleus toward the immunological synapse but is unable to dock at the plasma membrane. Lytic granules fail to polarize and release their contents, and target cells are not killed. In CTLs deficient in both Lck and the related tyrosine kinase Fyn, centrosome translocation is impaired, and the centrosome remains on the distal side of the nucleus relative to the synapse. These results show that repositioning of the centrosome in CTLs involves at least two distinct steps, with Lck signaling required for the centrosome to dock at the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Centrosome/metabolism , Immunological Synapses/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , T-Lymphocytes, Cytotoxic/enzymology , Animals , Cell Membrane/ultrastructure , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Centrosome/ultrastructure , Cytoplasmic Granules/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Proto-Oncogene Proteins c-fyn/physiology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/physiology , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
17.
Int J Oncol ; 37(3): 645-53, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664933

ABSTRACT

Umbilical cord blood cells (UCBC) are a rich source of immature immune effector and accessory cells, including dendritic cells. UCBC-derived cytotoxic T lymphocytes (CTLs) generated against human breast cancer or neuroblastoma have shown an increased tumor-specific cytotoxicity compared to peripheral blood (PB)-derived CTLs. The precise mechanism of this increased cytotoxicity is not known. Since dendritic cells (DCs) play a central role in the immunostimulation, we compared the ultrastructure and antigen presenting nature of DCs from UCBC, PB and bone marrow (BM) at various stages of maturation using scanning and transmission electron microscopy as well as fluorescent microscopy to elucidate the mechanism underlying the increased cytotoxicity of UCBC-derived CTLs. DCs were examined for their immunophenotype nuclear morphology, dendritic processes and cytoplasmic endosomal vesicles after 0, 3, 7 and 10 days in culture with antigen priming on day 6. Results showed that there were smaller and more vesicles in UCB-DCs compared to DCs from the other two sources, while the endosomal vesicles in PB-DCs were heterogenous in size. The antigen processing ability of the UCB-DCs showed an increase in antigen-positive endosomes compared to PB-DCs as determined by the fluorescent microscopy. Thus, our results provided the comparative analyses of DCs from cord blood, peripheral blood and bone marrow, and suggested that UCBC-DCs might have better antigen presenting ability leading to increased CTL-mediated antitumor cytotoxicity.


Subject(s)
Bone Marrow Cells/immunology , Fetal Blood/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigen Presentation/immunology , Bone Marrow Cells/ultrastructure , Cell Line, Tumor , Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Fetal Blood/cytology , Humans , Microscopy, Electron, Transmission , Microscopy, Fluorescence , T-Lymphocytes, Cytotoxic/ultrastructure
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 25(11): 980-3, 2009 Nov.
Article in Chinese | MEDLINE | ID: mdl-19900361

ABSTRACT

AIM: To prepare Nano-Liposome encapsulated MAGE3/HSP70(NL M3H) and study its character and antitumor immunity in mouse. METHODS: NL M3H was prepared by the thin film-dispersion ultrasonic. The shape and size of NL M3H were detected by electron microscope. The encapsulation rate, drug-carrying capacity, stability and the releasing character were tested by Sephedex-G100 gel filtration. The mouse was immunized by NL M3H, and the antitumor immunity was detected by ELISPOT and LDH release assay. RESULTS: The mean size of NL M3H was lower than 100 nm. The encapsulation rate was 38%.The drug content was 0.038 g/L. NL M3H has good stability after stored in 4 degrees C for 6 months. The releasing profile showed that 74 percent of proteins was released during the first 24 hours in saline. The results of ELISPOT and LDH release assay showed that NL M3H generated tumor specific cytotoxic T lymphocyte(CTL)to damage tumor cell. CONCLUSION: NL M3H has novel characters, it can generate specific CTL to kill tumor cell, and can be used as new kind of vaccine against tumor.


Subject(s)
Antigens, Neoplasm/blood , Antigens, Neoplasm/immunology , Cancer Vaccines/blood , Cancer Vaccines/immunology , Liposomes , Nanostructures , Animals , Antigens, Neoplasm/metabolism , Cancer Vaccines/metabolism , Cancer Vaccines/pharmacokinetics , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , HSP70 Heat-Shock Proteins/metabolism , Interferon-gamma/metabolism , Liposomes/chemistry , Mice , Microscopy, Electron , Nanostructures/chemistry , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/ultrastructure , Time Factors
19.
Immunity ; 31(4): 621-31, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19833087

ABSTRACT

Killing by cytotoxic T lymphocytes (CTLs) is mediated by the secretion of lytic granules. The centrosome plays a key role in granule delivery, polarizing to the central supramolecular activation complex (cSMAC) within the immunological synapse upon T cell receptor (TCR) activation. Although stronger TCR signals lead to increased target cell death than do weaker signals, it is not known how the strength of TCR signal controls polarization of the centrosome and lytic granules. By using TCR transgenic OT-I CTLs, we showed that both high- and low-avidity interactions led to centrosome polarization to the cSMAC. However, only high-avidity interactions, which induced a higher threshold of intracellular signaling, gave rise to granule recruitment to the polarized centrosome at the synapse. By controlling centrosome and granule polarization independently, the centrosome is able to respond rapidly to weak signals so that CTLs are poised and ready for the trigger for granule delivery.


Subject(s)
Cell Polarity/immunology , Centrosome/metabolism , Cytoplasmic Granules/metabolism , Immunological Synapses/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/metabolism , Animals , Cell Degranulation/immunology , Centrosome/immunology , Centrosome/ultrastructure , Cytoplasmic Granules/immunology , Cytoplasmic Granules/ultrastructure , Cytotoxicity, Immunologic/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Immunological Synapses/immunology , Immunological Synapses/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Ovalbumin/immunology , Peptide Fragments/immunology , Phosphorylation/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure , src-Family Kinases/immunology , src-Family Kinases/metabolism
20.
Biotechnol Bioeng ; 101(6): 1123-8, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18563847

ABSTRACT

Preparation of human immune T cells containing iron-oxide nanoparticles was carried out for the development of magnetically mediated immunotherapy. Peripheral blood lymphocytes (PBLs) after the incubation with magnetite nanoparticles were found to contain measurable ferric ions, which suggested the incorporation of magnetite nanoparticles. Transmission electron microscopic (TEM) study indicated that the incorporation of magnetite nanoparticles was mediated by endocytosis of PBLs. Furthermore, the effects of dosages and diameter of magnetite nanoparticles on the magnetite incorporation were investigated, and it was demonstrated that the increase in dosage promoted the incorporation of nanoparticles and the uptake into PBLs was more effective for magnetite nanoparticles, which formed smaller aggregations in medium. Finally, the demonstration of magnetite incorporation into enriched T cells and tumor antigen-specific cytotoxic T lymphocyte (CTL) line promises the achievement of magnetically mediated immunotherapy with tumor-specific CTLs containing magnetic nanoparticles.


Subject(s)
Ferric Compounds/metabolism , Nanoparticles , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Cells, Cultured , Cytoplasm/ultrastructure , Endocytosis , Ferrosoferric Oxide/metabolism , Humans , Microscopy, Electron, Transmission , T-Lymphocytes, Cytotoxic/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...