Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
J Cell Sci ; 133(17)2020 09 09.
Article in English | MEDLINE | ID: mdl-32907931

ABSTRACT

T cell entry into inflamed tissue requires firm adhesion, cell spreading, and migration along and through the endothelial wall. These events require the T cell integrins LFA-1 and VLA-4 and their endothelial ligands ICAM-1 and VCAM-1, respectively. T cells migrate against the direction of shear flow on ICAM-1 and with the direction of shear flow on VCAM-1, suggesting that these two ligands trigger distinct cellular responses. However, the contribution of specific signaling events downstream of LFA-1 and VLA-4 has not been explored. Using primary mouse T cells, we found that engagement of LFA-1, but not VLA-4, induces cell shape changes associated with rapid 2D migration. Moreover, LFA-1 ligation results in activation of the phosphoinositide 3-kinase (PI3K) and ERK pathways, and phosphorylation of multiple kinases and adaptor proteins, whereas VLA-4 ligation triggers only a subset of these signaling events. Importantly, T cells lacking Crk adaptor proteins, key LFA-1 signaling intermediates, or the ubiquitin ligase cCbl (also known as CBL), failed to migrate against the direction of shear flow on ICAM-1. These studies identify novel signaling differences downstream of LFA-1 and VLA-4 that drive T cell migratory behavior.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Actins , Lymphocyte Function-Associated Antigen-1 , Animals , Cell Adhesion , Intercellular Adhesion Molecule-1/genetics , Mice , Phosphatidylinositol 3-Kinases , Polymerization , T-Lymphocytes , Vascular Cell Adhesion Molecule-1
2.
PLoS Pathog ; 16(8): e1008685, 2020 08.
Article in English | MEDLINE | ID: mdl-32745153

ABSTRACT

Smallpox and monkeypox pose severe threats to human health. Other orthopoxviruses are comparably virulent in their natural hosts, including ectromelia, the cause of mousepox. Disease severity is linked to an array of immunomodulatory proteins including the B22 family, which has homologs in all pathogenic orthopoxviruses but not attenuated vaccine strains. We demonstrate that the ectromelia B22 member, C15, is necessary and sufficient for selective inhibition of CD4+ but not CD8+ T cell activation by immunogenic peptide and superantigen. Inhibition is achieved not by down-regulation of surface MHC- II or co-stimulatory protein surface expression but rather by interference with antigen presentation. The appreciable outcome is interference with CD4+ T cell synapse formation as determined by imaging studies and lipid raft disruption. Consequently, CD4+ T cell activating stimulus shifts to uninfected antigen-presenting cells that have received antigen from infected cells. This work provides insight into the immunomodulatory strategies of orthopoxviruses by elucidating a mechanism for specific targeting of CD4+ T cell activation, reflecting the importance of this cell type in control of the virus.


Subject(s)
Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Histocompatibility Antigens Class II/immunology , Viral Proteins/immunology , Animals , Ectromelia, Infectious/metabolism , Ectromelia, Infectious/virology , Female , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Viral Proteins/metabolism , Virulence
3.
Blood ; 131(15): 1743-1754, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29348127

ABSTRACT

Improved diagnostic and treatment methods are needed for chronic graft-versus-host disease (cGVHD), the leading cause of late nonrelapse mortality (NRM) in long-term survivors of allogenic hematopoietic cell transplantation. Validated biomarkers that facilitate disease diagnosis and classification generally are lacking in cGVHD. Here, we conducted whole serum proteomics analysis of a well-established murine multiorgan system cGVHD model. We discovered 4 upregulated proteins during cGVHD that are targetable by genetic ablation or blocking antibodies, including the RAS and JUN kinase activator, CRKL, and CXCL7, CCL8, and CCL9 chemokines. Donor T cells lacking CRK/CRKL prevented the generation of cGVHD, germinal center reactions, and macrophage infiltration seen with wild-type T cells. Whereas antibody blockade of CCL8 or CXCL7 was ineffective in treating cGVHD, CCL9 blockade reversed cGVHD clinical manifestations, histopathological changes, and immunopathological hallmarks. Mechanistically, elevated CCL9 expression was present predominantly in vascular smooth muscle cells and uniquely seen in cGVHD mice. Plasma concentrations of CCL15, the human homolog of mouse CCL9, were elevated in a previously published cohort of 211 cGVHD patients compared with controls and associated with NRM. In a cohort of 792 patients, CCL15 measured at day +100 could not predict cGVHD occurring within the next 3 months with clinically relevant sensitivity/specificity. Our findings demonstrate for the first time the utility of preclinical proteomics screening to identify potential new targets for cGVHD and specifically CCL15 as a diagnosis marker for cGVHD. These data warrant prospective biomarker validation studies.


Subject(s)
Chemokines, CC/blood , Graft vs Host Disease/blood , Macrophage Inflammatory Proteins/blood , Proteome/metabolism , Animals , Biomarkers/blood , Chemokines, CC/genetics , Chronic Disease , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Humans , Macrophage Inflammatory Proteins/genetics , Mice , Proteome/genetics , Proteomics
4.
Immunity ; 34(6): 825-7, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703536

ABSTRACT

Movement of immunoreceptor microclusters tunes lymphocyte activation, but the underlying mechanisms are incompletely understood. In this issue of Immunity, Schnyder et al. (2011) and Hashimoto-Tane et al. (2011) show that cytoplasmic dynein drives microcluster centralization along microtubules.

5.
J Immunol ; 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28794231

ABSTRACT

Sustained Ca2+ signaling, known as store-operated calcium entry (SOCE), occurs downstream of immunoreceptor engagement and is critical for cytotoxic lymphocyte signaling and effector function. CD8+ T cells require sustained Ca2+ signaling for inflammatory cytokine production and the killing of target cells; however, much less is known about its role in NK cells. In this study, we use mice deficient in stromal interacting molecules 1 and 2, which are required for SOCE, to examine the contribution of sustained Ca2+ signaling to murine NK cell function. Surprisingly, we found that, although SOCE is required for NK cell IFN-γ production in an NFAT-dependent manner, NK cell degranulation/cytotoxicity and tumor rejection in vivo remained intact in the absence of sustained Ca2+ signaling. Our data suggest that mouse NK cells use different signaling mechanisms for cytotoxicity compared with other cytotoxic lymphocytes.

6.
Immunity ; 28(6): 732-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18549795

ABSTRACT

Integrin engagement costimulates T cell receptor signaling, but the underlying mechanisms are poorly understood. In this issue of Immunity, Nguyen et al. (2008) show that engagement of VLA-4 promotes sustained signaling by altering the dynamics of actin filaments and signaling molecules at the immunological synapse.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Integrin alpha4beta1/metabolism , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Animals , Antigen Presentation , Humans , Lymphocyte Activation , Receptors, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/metabolism
7.
J Immunol ; 194(9): 4362-70, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25821220

ABSTRACT

Regulatory T cells (Tregs) are a subset of CD4(+) T cells that maintain immune tolerance in part by their ability to inhibit the proliferation of conventional CD4(+) T cells (Tconvs). The role of the TCR and the downstream signaling pathways required for this suppressive function of Tregs are not fully understood. To yield insight into how TCR-mediated signals influence Treg suppressive function, we assessed the ability of Tregs with altered TCR-mediated signaling capacity to inhibit Tconv proliferation. Mature Tregs deficient in Src homology 2 domain containing leukocyte protein of 76 kDa (SLP-76), an adaptor protein that nucleates the proximal signaling complex downstream of the TCR, were unable to inhibit Tconv proliferation, suggesting that TCR signaling is required for Treg suppressive function. Moreover, Tregs with defective phospholipase C γ (PLCγ) activation due to a Y145F mutation of SLP-76 were also defective in their suppressive function. Conversely, enhancement of diacylglycerol-mediated signaling downstream of PLCγ by genetic ablation of a negative regulator of diacylglycerol kinase ζ increased the suppressive ability of Tregs. Because SLP-76 is also important for integrin activation and signaling, we tested the role of integrin activation in Treg-mediated suppression. Tregs lacking the adaptor proteins adhesion and degranulation promoting adapter protein or CT10 regulator of kinase/CT10 regulator of kinase-like, which are required for TCR-mediated integrin activation, inhibited Tconv proliferation to a similar extent as wild-type Tregs. Together, these data suggest that TCR-mediated PLCγ activation, but not integrin activation, is required for Tregs to inhibit Tconv proliferation.


Subject(s)
Immunomodulation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Diglycerides/metabolism , Integrins/metabolism , Mice , Mice, Transgenic , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell/genetics
8.
Immunol Rev ; 256(1): 80-94, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117814

ABSTRACT

Ca(2+) mobilization and cytoskeletal reorganization are key hallmarks of T-cell activation, and their interdependence has long been recognized. Recent advances in the field have elucidated the molecular pathways that underlie these events and have revealed several points of intersection. Ca(2+) signaling can be divided into two phases: initial events leading to release of Ca(2+) from endoplasmic reticulum stores, and a second phase involving STIM 1 (stromal interaction molecule 1) clustering and CRAC (calcium release activated calcium) channel activation. Cytoskeletal dynamics promote both phases. During the first phase, the actin cytoskeleton promotes mechanotransduction and serves as a dynamic scaffold for microcluster assembly. Proteins that drive actin polymerization such as WASp (Wiskott-Aldrich syndrome protein) and HS1 (hematopoietic lineage cell-specific protein 1) promote signaling through PLCγ1 (phospholipase Cγ1) and release of Ca(2+) from endoplasmic reticulum stores. During the second phase, the WAVE (WASP-family verprolin homologous protein) complex and the microtubule cytoskeleton promote STIM 1 clustering at sites of plasma membrane apposition, opening Orai channels. In addition, gross cell shape changes and organelle movements buffer local Ca(2+) levels, leading to sustained Ca(2+) mobilization. Conversely, elevated intracellular Ca(2+) activates cytoskeletal remodeling. This can occur indirectly, via calpain activity, and directly, via Ca(2+) -dependent cytoskeletal regulatory proteins such as myosin II and L-plastin. While it is true that the cytoskeleton regulates Ca(2+) responses and vice versa, interdependence between Ca(2+) and the cytoskeleton also encompasses signaling events that occur in parallel, downstream of shared intermediates. Inositol cleavage by PLCγ1 simultaneously triggers both endoplasmic reticulum store release and diacylglycerol-dependent microtubule organizing center reorientation, while depleting the pool of phosphatidylinositol-4,5-bisphosphate, an activator of multiple actin-regulatory proteins. The close interdependence of Ca(2+) signaling and cytoskeletal dynamics in T cells provides positive feedback mechanisms for T-cell activation and allows for finely tuned responses to extracellular cues.


Subject(s)
Calcium Signaling , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Actins/metabolism , Animals , Calcium/metabolism , Cell Movement , Humans , Organelles/metabolism , Receptors, Antigen, T-Cell/metabolism
9.
J Immunol ; 188(9): 4145-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22467651

ABSTRACT

Clonal selection of a T cell for use in the immune response appears to necessitate proliferative expansion and terminal effector differentiation of some cellular progeny, while reserving other progeny as less-differentiated memory cells. It has been suggested that asymmetric cell division may promote initial cell diversification. Stem cell-like models of adaptive immunity might predict that subsequent encounters with a pathogen would evoke reiterative, self-renewing, asymmetric division by memory T cells. In this study, we show that murine memory CD8(+) T cells can divide asymmetrically in response to secondary encounter with pathogen. Critical regulators of signaling and transcription are partitioned to one side of the mitotic spindle in rechallenged memory T cells, and two phenotypically distinct populations of daughter cells are evident from the earliest divisions. Memory T cells may thus use asymmetric cell division to generate cellular heterogeneity when faced with pathogen rechallenge.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Signal Transduction/immunology , Animals , Cell Division/genetics , Lymphocytic Choriomeningitis/genetics , Mice , Mice, Transgenic , Signal Transduction/genetics , Transcription, Genetic/genetics , Transcription, Genetic/immunology
10.
J Immunol ; 188(12): 6135-44, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22573807

ABSTRACT

The Ras GTPase-activating-like protein IQGAP1 is a multimodular scaffold that controls signaling and cytoskeletal regulation in fibroblasts and epithelial cells. However, the functional role of IQGAP1 in T cell development, activation, and cytoskeletal regulation has not been investigated. In this study, we show that IQGAP1 is dispensable for thymocyte development as well as microtubule organizing center polarization and cytolytic function in CD8(+) T cells. However, IQGAP1-deficient CD8(+) T cells as well as Jurkat T cells suppressed for IQGAP1 were hyperresponsive, displaying increased IL-2 and IFN-γ production, heightened LCK activation, and augmented global phosphorylation kinetics after TCR ligation. In addition, IQGAP1-deficient T cells exhibited increased TCR-mediated F-actin assembly and amplified F-actin velocities during spreading. Moreover, we found that discrete regions of IQGAP1 regulated cellular activation and F-actin accumulation. Taken together, our data suggest that IQGAP1 acts as a dual negative regulator in T cells, limiting both TCR-mediated activation kinetics and F-actin dynamics via distinct mechanisms.


Subject(s)
Actin Cytoskeleton/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Signal Transduction/immunology , ras GTPase-Activating Proteins/metabolism , Actins/immunology , Actins/metabolism , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cytoskeleton/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Jurkat Cells , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , ras GTPase-Activating Proteins/immunology
11.
bioRxiv ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38798563

ABSTRACT

Osteoclasts are multinucleated cells unique in their ability to resorb bone. Osteoclastogenesis involves several steps of actin-driven rearrangements that participate not only in the cell-cell fusion process, but also in the formation of the sealing zone, the adhesive structure determining the resorption area. Despite the importance of these actin cytoskeleton-based processes, their precise mechanisms of regulation are still poorly characterized. Here, we found that moesin, a member of the Ezrin/Radixin/Moesin (ERM) protein family, is activated during osteoclast maturation and plays an instrumental role for both osteoclast fusion and function. In mouse and human osteoclast precursors, moesin is negatively regulated to potentiate their ability to fuse and degrade bone. Accordingly, we demonstrated that moesin depletion decreases membrane-to-cortex attachment and enhances formation of tunneling nanotubes (TNTs), F-actin-containing intercellular bridges that we revealed to trigger osteoclast fusion. In addition, via a ß3-integrin/RhoA/SLK pathway and independently of its role in fusion, moesin regulates the number and organization of sealing zones in mature osteoclast, and thus participates in the control of bone resorption. Supporting these findings, we found that moesin-deficient mice are osteopenic with a reduced density of trabecular bones and increased osteoclast abundance and activity. These findings provide a better understanding of the regulation of osteoclast biology, and open new opportunities to specifically target osteoclast activity in bone disease therapy.

12.
J Immunol ; 186(5): 2850-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21263068

ABSTRACT

Dendritic cells (DCs) play central roles in innate and adaptive immunity. Upon maturation, DCs assemble numerous veil-like membrane protrusions, disassemble podosomes, and travel from the peripheral tissues to lymph nodes to present Ags to T cells. These alterations in morphology and motility are closely linked to the primary function of DCs, Ag presentation. However, it is unclear how and what cytoskeletal proteins control maturation-associated alterations, in particular, the change in cell migration. Fascin1, an actin-bundling protein, is specifically and greatly induced upon maturation, suggesting a unique role for fascin1 in mature DCs. To determine the physiological roles of fascin1, we characterized bone marrow-derived, mature DCs from fascin1 knockout mice. We found that fascin1 is critical for cell migration: fascin1-null DCs exhibit severely decreased membrane protrusive activity. Importantly, fascin1-null DCs have lower chemotactic activity toward CCL19 (a chemokine for mature DCs) in vitro, and in vivo, Langerhans cells show reduced emigration into draining lymph nodes. Morphologically, fascin1-null mature DCs are flatter and fail to disassemble podosomes, a specialized structure for cell-matrix adhesion. Expression of exogenous fascin1 in fascin1-null DCs rescues the defects in membrane protrusive activity, as well as in podosome disassembly. These results indicate that fascin1 positively regulates migration of mature DCs into lymph nodes, most likely by increasing dynamics of membrane protrusions, as well as by disassembling podosomes.


Subject(s)
Cell Differentiation/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Microfilament Proteins/physiology , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Movement/genetics , Cell Surface Extensions/immunology , Cell Surface Extensions/pathology , Cell Surface Extensions/ultrastructure , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Female , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Receptors, Odorant
13.
J Immunol ; 187(11): 5952-63, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22031761

ABSTRACT

The hematopoietic actin regulatory protein hematopoietic lineage cell-specific protein 1 (HS1) is required for cell spreading and signaling in lymphocytes, but the scope of HS1 function in Ag presentation has not been addressed. We show that dendritic cells (DCs) from HS1(-/-) mice differentiate normally and display normal LPS-induced upregulation of surface markers and cytokines. Consistent with their normal expression of MHC and costimulatory molecules, HS1(-/-) DCs present OVA peptide efficiently to CD4(+) T cells. However, presentation of OVA protein is defective. Similarly, MHC class I-dependent presentation of VSV8 peptide to CD8(+) T cells occurs normally, but cross-presentation of GRP94/VSV8 complexes is defective. Analysis of Ag uptake pathways shows that HS1 is required for receptor-mediated endocytosis, but not for phagocytosis or macropinocytosis. HS1 interacts with dynamin 2, a protein involved in scission of endocytic vesicles. However, HS1(-/-) DCs showed decreased numbers of endocytic invaginations, whereas dynamin-inhibited cells showed accumulation of these endocytic intermediates. Taken together, these studies show that HS1 promotes an early step in the endocytic pathway that is required for efficient Ag presentation of exogenous Ag by DCs.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Endocytosis/immunology , Granulocyte Colony-Stimulating Factor/immunology , Animals , Blotting, Western , Cell Separation , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Flow Cytometry , Granulocyte Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Ovalbumin/immunology
14.
J Immunol ; 186(8): 4805-18, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21398607

ABSTRACT

Dendritic cells (DCs) are professional APCs that reside in peripheral tissues and survey the body for pathogens. Upon activation by inflammatory signals, DCs undergo a maturation process and migrate to lymphoid organs, where they present pathogen-derived Ags to T cells. DC migration depends on tight regulation of the actin cytoskeleton to permit rapid adaptation to environmental cues. We investigated the role of hematopoietic lineage cell-specific protein 1 (HS1), the hematopoietic homolog of cortactin, in regulating the actin cytoskeleton of murine DCs. HS1 localized to lamellipodial protrusions and podosomes, actin-rich structures associated with adhesion and migration. DCs from HS1(-/-) mice showed aberrant lamellipodial dynamics. Moreover, although these cells formed recognizable podosomes, their podosome arrays were loosely packed and improperly localized within the cell. HS1 interacts with Wiskott-Aldrich syndrome protein (WASp), another key actin-regulatory protein, through mutual binding to WASp-interacting protein. Comparative analysis of DCs deficient for HS1, WASp or both proteins revealed unique roles for these proteins in regulating podosomes with WASp being essential for podosome formation and with HS1 ensuring efficient array organization. WASp recruitment to podosome cores was independent of HS1, whereas HS1 recruitment required Src homology 3 domain-dependent interactions with the WASp/WASp-interacting protein heterodimer. In migration assays, the phenotypes of HS1- and WASp-deficient DCs were related, but distinct. WASp(-/y) DCs migrating in a chemokine gradient showed a large decrease in velocity and diminished directional persistence. In contrast, HS1(-/-) DCs migrated faster than wild-type cells, but directional persistence was significantly reduced. These studies show that HS1 functions in concert with WASp to fine-tune DC cytoarchitecture and direct cell migration.


Subject(s)
Chemotaxis/immunology , Dendritic Cells/immunology , Granulocyte Colony-Stimulating Factor/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Actins/genetics , Actins/metabolism , Animals , Antigen Presentation/immunology , Blotting, Western , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Movement/immunology , Cells, Cultured , Cytoskeleton/immunology , Cytoskeleton/metabolism , Dendritic Cells/metabolism , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Protein Binding , Pseudopodia/immunology , Pseudopodia/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
15.
Immunol Rev ; 231(1): 148-59, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19754895

ABSTRACT

In the last few years, great progress has been made in understanding how stromal interacting molecule 1 (STIM1), a protein containing a calcium sensor that is located in the endoplasmic reticulum, and Orai1, a protein that forms a calcium channel in the plasma membrane, interact and give rise to store-operated calcium entry. Pharmacological depletion of calcium stores leads to the formation of clusters containing STIM and Orai that appear to be sites for calcium influx. Similar puncta are also produced in response to physiological stimuli in immune cells. In T cells engaged with antigen-presenting cells, clusters containing STIM and Orai accumulate at the immunological synapse. We recently discovered that in activated T cells, STIM1 and Orai1 also accumulate in cap-like structures opposite the immune synapse at the distal pole of the cell. Both caps and puncta are long-lived stable structures containing STIM1 and Orai1 in close proximity. The function of puncta as sites of calcium influx is clear. We speculate that the caps may provide a secondary site of calcium entry. Alternatively, they may serve as a source of preformed channel complexes that move to new immune synapses as T cells repeatedly engage antigen-presenting cells.


Subject(s)
Calcium Channels/metabolism , Membrane Proteins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Calcium/metabolism , Calcium Channels/chemistry , Humans , Membrane Proteins/chemistry , Protein Binding , Signal Transduction , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
17.
Immunol Rev ; 256(1): 5-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117809
18.
J Clin Invest ; 132(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35579963

ABSTRACT

In lymphopenic environments, secondary lymphoid organs regulate the size of B and T cell compartments by supporting the homeostatic proliferation of mature lymphocytes. The molecular mechanisms underlying these responses and their functional consequences remain incompletely understood. To evaluate homeostasis of the mature B cell pool during lymphopenia, we turned to an adoptive transfer model of purified follicular B cells into Rag2-/- mouse recipients. Highly purified follicular B cells transdifferentiated into marginal zone-like B cells when transferred into Rag2-/- lymphopenic hosts but not into wild-type hosts. In lymphopenic spleens, transferred B cells gradually lost their follicular phenotype and acquired characteristics of marginal zone B cells, as judged by cell surface phenotype, expression of integrins and chemokine receptors, positioning close to the marginal sinus, and an ability to rapidly generate functional plasma cells. Initiation of follicular to marginal zone B cell transdifferentiation preceded proliferation. Furthermore, the transdifferentiation process was dependent on Notch2 receptors in B cells and expression of Delta-like 1 Notch ligands by splenic Ccl19-Cre+ fibroblastic stromal cells. Gene expression analysis showed rapid induction of Notch-regulated transcripts followed by upregulated Myc expression and acquisition of broad transcriptional features of marginal zone B cells. Thus, naive mature B cells are endowed with plastic transdifferentiation potential in response to increased stromal Notch ligand availability during lymphopenia.


Subject(s)
Lymphopenia , Animals , B-Lymphocytes/metabolism , Cell Proliferation , Homeostasis , Lymphopenia/genetics , Mice , Mice, Inbred C57BL
19.
J Immunol ; 182(2): 1021-32, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124745

ABSTRACT

The highly homologous proteins ezrin, radixin, and moesin link proteins to the actin cytoskeleton. The two family members expressed in T cells, ezrin and moesin, are implicated in promoting T cell activation and polarity. To elucidate the contributions of ezrin and moesin, we conducted a systematic analysis of their function during T cell activation. In response to TCR engagement, ezrin and moesin were phosphorylated in parallel at the regulatory threonine, and both proteins ultimately localized to the distal pole complex (DPC). However, ezrin exhibited unique behaviors, including tyrosine phosphorylation and transient localization to the immunological synapse before movement to the DPC. To ask whether these differences reflect unique requirements for ezrin vs moesin in T cell signaling, we generated mice with conditional deletion of ezrin in mature T cells. Ezrin-/- T cells exhibited normal immunological synapse organization based upon localization of protein kinase C-theta, talin, and phospho-ZAP70. DPC localization of CD43 and RhoGDP dissociation inhibitor, as well as the novel DPC protein Src homology region 2 domain-containing phosphatase-1, was also unaffected. However, recruitment of three novel DPC proteins, ezrin binding protein of 50 kDa, Csk binding protein, and the p85 subunit of PI3K was partially perturbed. Biochemical analysis of ezrin-/- T cells or T cells suppressed for moesin using small interfering RNA showed intact early TCR signaling, but diminished levels of IL-2. The defects in IL-2 production were more pronounced in T cells deficient for both ezrin and moesin. These cells also exhibited diminished phospholipase C-gamma1 phosphorylation and calcium flux. We conclude that despite their unique movement and phosphorylation patterns, ezrin and moesin function together to promote T cell activation.


Subject(s)
Cytoskeletal Proteins/physiology , Lymphocyte Activation/immunology , Microfilament Proteins/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Cell Line , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Humans , Immunological Synapses/genetics , Jurkat Cells , Lymphocyte Activation/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Receptors, Antigen, T-Cell/physiology , Signal Transduction/immunology
20.
J Immunol ; 183(11): 7352-61, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19917685

ABSTRACT

Productive T cell activation requires efficient reorganization of the actin cytoskeleton. We showed previously that the actin-regulatory protein, hematopoietic lineage cell-specific protein 1 (HS1), is required for the stabilization of F-actin and Vav1 at the immunological synapse and for efficient calcium responses. The Tec family kinase IL-2-inducible T cell kinase (Itk) regulates similar aspects of T cell activation, suggesting that these proteins act in the same pathway. Using video microscopy, we show that T cells lacking Itk or HS1 exhibited similar defects in actin responses, extending unstable lamellipodial protrusions upon TCR stimulation. HS1 and Itk could be coimmunoprecipitated from T cell lysates, and GST-pulldown studies showed that Itk's Src homology 2 domain binds directly to two phosphotyrosines in HS1. In the absence of Itk, or in T cells overexpressing an Itk Src homology 2 domain mutant, HS1 failed to localize to the immunological synapse, indicating that Itk serves to recruit HS1 to sites of TCR engagement. Because Itk is required for phospholipase C (PLC)gamma1 phosphorylation and calcium store release, we examined the calcium signaling pathway in HS1(-/-) T cells in greater detail. In response to TCR engagement, T cells lacking HS1 exhibited diminished calcium store release, but TCR-dependent PLCgamma1 phosphorylation was intact, indicating that HS1's role in calcium signaling is distinct from that of Itk. HS1-deficient T cells exhibited defective cytoskeletal association of PLCgamma1 and altered formation of PLCgamma1 microclusters. We conclude that HS1 functions as an effector of Itk in the T cell actin-regulatory pathway, and directs the spatial organization of PLCgamma1 signaling complexes.


Subject(s)
Granulocyte Colony-Stimulating Factor/immunology , Immunological Synapses/immunology , Phospholipase C gamma/immunology , Protein-Tyrosine Kinases/immunology , T-Lymphocytes/immunology , Actins/metabolism , Animals , Blotting, Western , Calcium Signaling/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Immunological Synapses/metabolism , Immunoprecipitation , Jurkat Cells , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Phospholipase C gamma/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Pseudopodia/metabolism , Pseudopodia/pathology , RNA Interference , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL