1
|
Yan M, Yao J, Xie Y, Jiang P, Yan J, Li X. Bioreactor-based stem cell therapy for liver fibrosis. Biofabrication 2024; 16:025028. [PMID: 38442726 DOI: 10.1088/1758-5090/ad304d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/05/2024] [Indexed: 03/07/2024]
Abstract
Stem cell therapy, achieved using mesenchymal stem cells (MSCs), has been highlighted for the treatment of liver fibrosis. Infusion into the circulatory system is a traditional application of MSCs; however, this approach is limited by phenotypic drift, stem cell senescence, and vascular embolism. Maintaining the therapeutic phenotype of MSCs while avoiding adverse infusion-related reactions is the key to developing next-generation stem cell therapy technologies. Here, we propose a bioreactor-based MSCs therapy to avoid cell infusion. In this scheme, 5% liver fibrosis serum was used to induce the therapeutic phenotype of MSCs, and a fluid bioreactor carrying a co-culture system of hepatocytes and MSCs was constructed to produce the therapeutic medium. In a rat model of liver fibrosis, the therapeutic medium derived from the bioreactor significantly alleviated liver fibrosis. Therapeutic mechanisms include immune regulation, inhibition of hepatic stellate cell activation, establishment of hepatocyte homeostasis, and recovery of liver stem cell subsets. Overall, the bioreactor-based stem cell therapy (scheme) described here represents a promising new strategy for the treatment of liver fibrosis and will be beneficial for the development of 'cell-free' stem cell therapy.
Collapse
Affiliation(s)
- Mengchao Yan
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, People's Republic of China
- The Medical School, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Jia Yao
- The Medical School, Lanzhou University, Lanzhou 730000, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| | - Ye Xie
- The Medical School, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Pan Jiang
- State Key Laboratory of Solid Lubrication, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, People's Republic of China
| | - Jun Yan
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, People's Republic of China
- The Medical School, Lanzhou University, Lanzhou 730000, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, People's Republic of China
- The Medical School, Lanzhou University, Lanzhou 730000, People's Republic of China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, People's Republic of China
| |
Collapse
|
2
|
Li M, Luo S, Zhuo Z, Shu M. Two cases of pediatric primary immunodeficiency caused by a familial moesin(MSN)gene mutation. Clin Immunol 2024; 258:109858. [PMID: 38052292 DOI: 10.1016/j.clim.2023.109858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND We investigated two brothers who presented with repeated lung infections after 6 months of age. Lymphocytes and neutrophils were significantly decreased, and both had bronchiectasis and emphysema. OBJECTIVE We sought to characterize the complete picture of lung injury in some types of primary immunodeficiency disease, followed by verification and analysis. METHODS We performed immune function determination, a complete examination of the respiratory system, genetic analysis, and literature research. RESULTS The levels of lymphocytes, neutrophils, monocytes, and natural killer cells in the brothers were significantly decreased. The IgM and IgG levels of the older brother were decreased, while the IgM and IgA levels of the younger brother were decreased. Both brothers had bronchial wall erosion with a worm-eaten appearance and decreased lung function. Genetic testing revealed a hemizygous missense mutation (c.511C > T:p.R171W) in exon 5 of the MSN gene, which was inherited from the mother. A literature review showed that the primary immunodeficiency caused by MSN gene mutations is an X-linked recessive genetic disease with four known gene mutation sites, including nonsense and missense mutations. Nonsense mutations result in a higher incidence of autoimmune diseases and a lower degree of immune function impairment. Nonsense mutations closer to the front of the MSN gene may cause more severe disease. Neonatal disease screening can improve the early diagnosis rate, but hematopoietic stem cell transplantation (HSCT) treatment is controversial. CONCLUSION The primary immunodeficiency disease caused by MSN gene mutation is an X-linked recessive genetic disease that involves structural and functional damage to the respiratory system, and the worm-eaten appearance of the bronchial wall under endoscopy may be a relatively specific sign. The general manifestations of this disease are recurrent infections from 1 month to 6 months after birth, significantly reduced counts of lymphocytes and neutrophils, and decreased cellular and humoral immune function. Different types of MSN gene mutations and nonsense mutations at different sites have different clinical phenotypes. This study enriches the known spectrum of this disease.
Collapse
Affiliation(s)
- Muquan Li
- Department of Pediatrics, West China Xiamen Hospital (Research Institute), Sichuan University, Xiamen, China; The Xiamen Key Laboratory of Psychoradiology and Neuromodulation, Xiamen, China
| | - Shuanghong Luo
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | | | - Min Shu
- Department of Pediatrics, West China Xiamen Hospital (Research Institute), Sichuan University, Xiamen, China; The Xiamen Key Laboratory of Psychoradiology and Neuromodulation, Xiamen, China; Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
3
|
Kovács AL, Kárteszi J, Prohászka Z, Kalmár T, Késmárky G, Koltai K, Nagy Z, Sebők J, Vas T, Molnár K, Berki T, Böröcz K, Gyömörei C, Szalma J, Egyed M, Horváth S, Oláh P, Csuka D, Németh V, Gyulai R. Hemizygous nonsense variant in the moesin gene (MSN) leads to a new autoimmune phenotype of Immunodeficiency 50. Front Immunol 2022; 13:919411. [PMID: 36119109 PMCID: PMC9477008 DOI: 10.3389/fimmu.2022.919411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Here, we present the findings of an investigation involving two male siblings with juvenile total tooth loss, early-onset chronic leg ulcers, and autoimmune thyroiditis, as well as focal segmental glomerulosclerosis with associated pulmonary emphysema in one and diabetes mellitus in the other. The clinical picture and lupus anticoagulant, cryoglobulin, and cold agglutinin positivity suggested the diagnosis of antiphospholipid syndrome. Flow cytometry analysis showed immunophenotypes consistent with immune dysregulation: a low number of naive T cells, elevated CD4+ T cell counts, and decreased CD8+ T-cell counts were detected, and more than half of the T-helper population was activated. Considering the siblings' almost identical clinical phenotype, the genetic alteration was suspected in the background of the immunodeficiency. Whole exome sequencing identified a previously not described hemizygous nonsense variant (c.650G>A, p.W217X) within exon 6 of the moesin (MSN) gene localized on chromosome X, resulting in significantly decreased MSN mRNA expression compared to healthy controls. We present a putative new autoimmune phenotype of Immunodeficiency 50 (MIM300988) characterized by antiphospholipid syndrome, Hashimoto's thyroiditis, leg ulcers, and juvenile tooth loss, associated with W217X mutation of the MSN gene.
Collapse
Affiliation(s)
- András L. Kovács
- Department of Dermatology, Venereology and Oncodermatology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Judit Kárteszi
- Genetic Counseling, Saint Rafael Hospital of Zala County, Zalaegerszeg, Hungary
| | - Zoltán Prohászka
- Research Group for Immunology and Haematology, Eötvös Loránd Research Network (Office for Supported Research Groups), Semmelweis University, Budapest, Hungary
| | - Tibor Kalmár
- Genetic Diagnostic Laboratory, Department of Pediatrics and Pedriatic Health Center, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
| | - Gábor Késmárky
- Division of Angiology, 1st Department of Internal Medicine, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Katalin Koltai
- Division of Angiology, 1st Department of Internal Medicine, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Nagy
- Nephrological and Diabetological Center, 2nd Department of Internal Medicine, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Judit Sebők
- Nephrological and Diabetological Center, 2nd Department of Internal Medicine, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Tibor Vas
- Nephrological and Diabetological Center, 2nd Department of Internal Medicine, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Krisztián Molnár
- Department of Medical Imaging, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Katalin Böröcz
- Department of Immunology and Biotechnology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Csaba Gyömörei
- Department of Pathology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - József Szalma
- Oral and Maxillofacial Surgery, Department of Dentistry, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Miklós Egyed
- Department of Hematology, Somogy County Mór Kaposi General Hospital, Kaposvár, Hungary
| | - Szabina Horváth
- Department of Dermatology, Venereology and Oncodermatology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Péter Oláh
- Department of Dermatology, Venereology and Oncodermatology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
- Department of Dermatology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Dorottya Csuka
- Research Group for Immunology and Haematology, Eötvös Loránd Research Network (Office for Supported Research Groups), Semmelweis University, Budapest, Hungary
| | - Viktória Németh
- Department of Dermatology, Venereology and Oncodermatology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Rolland Gyulai
- Department of Dermatology, Venereology and Oncodermatology, Medical School, Clinical Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
4
|
Gómez-Henao W, Saavedra R, Chávez-Sánchez FR, Lascurain R, Zenteno E, Tenorio EP. Expression Dynamics of the O-Glycosylated Proteins Recognized by Amaranthus leucocarpus Lectin in T Lymphocytes and Its Relationship With Moesin as an Alternative Mechanism of Cell Activation. Front Immunol 2021; 12:788880. [PMID: 34917095 PMCID: PMC8669815 DOI: 10.3389/fimmu.2021.788880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
T lymphocyte activation begins with antigen/MHC recognition by the TCR/CD3 complex followed by a costimulatory signal provided by CD28. The search for novel costimulatory molecules has been extensive due to their potential use as immunotherapeutic targets. Although some molecules have been identified, they are unable to provide sustainable signaling to allow for proper T cell activation and proliferation. It has been shown that the Amaranthus leucocarpus lectin (ALL) can be used as an in vitro costimulator of CD4+ lymphocytes in the presence of anti-CD3 mAb; this lectin specifically recognizes O-glycans of the Galβ1-3GalNAc-O-Ser/Thr type, including a 70-kDa moesin-like protein that has been suggested as the costimulatory molecule. However, the identity of this molecule has not been confirmed and such costimulation has not been analyzed in CD8+ lymphocytes. We show herein that the expression kinetics of the glycoproteins recognized by ALL (gpALL) is different in CD4+ and CD8+ T cells, unlike moesin expression. Results from IP experiments demonstrate that the previously described 70-kDa moesin-like protein is an O-glycosylated form of moesin (O-moesin) and that in vitro stimulation with anti-CD3 and anti-moesin mAb induces expression of the activation molecules CD69 and CD25, proliferation and IL-2 production as efficiently as cells costimulated with ALL or anti-CD28. Overall, our results demonstrate that O-moesin is expressed in CD4+ and CD8+ T lymphocytes and that moesin provides a new costimulatory activation signal in both T cell subsets.
Collapse
Affiliation(s)
- Wilton Gómez-Henao
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rafael Saavedra
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Ricardo Lascurain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Eda Patricia Tenorio
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
5
|
Dupré L, Boztug K, Pfajfer L. Actin Dynamics at the T Cell Synapse as Revealed by Immune-Related Actinopathies. Front Cell Dev Biol 2021; 9:665519. [PMID: 34249918 PMCID: PMC8266300 DOI: 10.3389/fcell.2021.665519] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/06/2021] [Indexed: 01/21/2023] Open
Abstract
The actin cytoskeleton is composed of dynamic filament networks that build adaptable local architectures to sustain nearly all cellular activities in response to a myriad of stimuli. Although the function of numerous players that tune actin remodeling is known, the coordinated molecular orchestration of the actin cytoskeleton to guide cellular decisions is still ill defined. T lymphocytes provide a prototypical example of how a complex program of actin cytoskeleton remodeling sustains the spatio-temporal control of key cellular activities, namely antigen scanning and sensing, as well as polarized delivery of effector molecules, via the immunological synapse. We here review the unique knowledge on actin dynamics at the T lymphocyte synapse gained through the study of primary immunodeficiences caused by mutations in genes encoding actin regulatory proteins. Beyond the specific roles of individual actin remodelers, we further develop the view that these operate in a coordinated manner and are an integral part of multiple signaling pathways in T lymphocytes.
Collapse
Affiliation(s)
- Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Laurène Pfajfer
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France.,St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| |
Collapse
|
6
|
Neutrophils lacking ERM proteins polarize and crawl directionally but have decreased adhesion strength. Blood Adv 2021; 4:3559-3571. [PMID: 32761234 DOI: 10.1182/bloodadvances.2020002423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/28/2020] [Indexed: 12/19/2022] Open
Abstract
Ezrin/radixin/moesin (ERM) proteins are adaptors that link the actin cytoskeleton to the cytoplasmic domains of membrane proteins. Leukocytes express mostly moesin with lower levels of ezrin but no radixin. When leukocytes are activated, ERMs are postulated to redistribute membrane proteins from microvilli into uropods during polarization and to transduce signals that influence adhesion and other responses. However, these functions have not been tested in leukocytes lacking all ERMs. We used knockout (KO) mice with neutrophils lacking ezrin, moesin, or both proteins (double knockout [DKO]) to probe how ERMs modulate cell shape, adhesion, and signaling in vitro and in vivo. Surprisingly, chemokine-stimulated DKO neutrophils still polarized and redistributed ERM-binding proteins such as PSGL-1 and CD44 to the uropods. Selectin binding to PSGL-1 on moesin KO or DKO neutrophils activated kinases that enable integrin-dependent slow rolling but not those that generate neutrophil extracellular traps. Flowing neutrophils of all genotypes rolled normally on selectins and, upon chemokine stimulation, arrested on integrin ligands. However, moesin KO and DKO neutrophils exhibited defective integrin outside-in signaling and reduced adhesion strength. In vivo, DKO neutrophils displayed normal directional crawling toward a chemotactic gradient, but premature detachment markedly reduced migration from venules into inflamed tissues. Our results demonstrate that stimulated neutrophils do not require ERMs to polarize or to move membrane proteins into uropods. They also reveal an unexpected contribution of moesin to integrin outside-in signaling and adhesion strengthening.
Collapse
|
7
|
Zheng S, Huang K, Xia W, Shi J, Liu Q, Zhang X, Li G, Chen J, Wang T, Chen X, Xiang AP. Mesenchymal Stromal Cells Rapidly Suppress TCR Signaling-Mediated Cytokine Transcription in Activated T Cells Through the ICAM-1/CD43 Interaction. Front Immunol 2021; 12:609544. [PMID: 33692786 PMCID: PMC7937648 DOI: 10.3389/fimmu.2021.609544] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Cell-cell contact participates in the process of mesenchymal stromal cell (MSC)-mediated T cell modulation and thus contributes to MSC-based therapies for various inflammatory diseases, especially T cell-mediated diseases. However, the mechanisms underlying the adhesion interactions between MSCs and T cells are still poorly understood. In this study, we explored the interaction between MSCs and T cells and found that activated T cells could rapidly adhere to MSCs, leading to significant reduction of TNF-α and IFN-γ mRNA expression. Furthermore, TCR-proximal signaling in activated T cells was also dramatically suppressed in the MSC co-culture, resulting in weakened Ca2+ signaling. MSCs rapidly suppressed TCR signaling and its downstream signaling in a cell-cell contact-dependent manner, partially through the ICAM-1/CD43 adhesion interaction. Blockade of either ICAM-1 on MSCs or CD43 on T cells significantly reversed this rapid suppression of proinflammatory cytokine expression in T cells. Mechanistically, MSC-derived ICAM-1 likely disrupts CD43-mediated TCR microcluster formation to limit T cell activation. Taken together, our results reveal a fast mechanism of activated T cell inhibition by MSCs, which provides new clues to unravel the MSC-mediated immunoregulatory mechanism for aGVHD and other severe acute T cell-related diseases.
Collapse
Affiliation(s)
- Shuwei Zheng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Ke Huang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenjie Xia
- Guangzhou Blood Centre, Institute of Blood Transfusion, Guangzhou, China
| | - Jiahao Shi
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Qiuli Liu
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoran Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Gang Li
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Jieying Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Tao Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyong Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China.,Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Andy Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
8
|
Bisaria A, Hayer A, Garbett D, Cohen D, Meyer T. Membrane-proximal F-actin restricts local membrane protrusions and directs cell migration. Science 2020; 368:1205-1210. [PMID: 32527825 DOI: 10.1126/science.aay7794] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 02/24/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Abstract
Cell migration is driven by local membrane protrusion through directed polymerization of F-actin at the front. However, F-actin next to the plasma membrane also tethers the membrane and thus resists outgoing protrusions. Here, we developed a fluorescent reporter to monitor changes in the density of membrane-proximal F-actin (MPA) during membrane protrusion and cell migration. Unlike the total F-actin concentration, which was high in the front of migrating cells, MPA density was low in the front and high in the back. Back-to-front MPA density gradients were controlled by higher cofilin-mediated turnover of F-actin in the front. Furthermore, nascent membrane protrusions selectively extended outward from areas where MPA density was reduced. Thus, locally low MPA density directs local membrane protrusions and stabilizes cell polarization during cell migration.
Collapse
Affiliation(s)
- Anjali Bisaria
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Arnold Hayer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Damien Garbett
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Cohen
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
9
|
ERM Proteins at the Crossroad of Leukocyte Polarization, Migration and Intercellular Adhesion. Int J Mol Sci 2020; 21:ijms21041502. [PMID: 32098334 PMCID: PMC7073024 DOI: 10.3390/ijms21041502] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
Ezrin, radixin and moesin proteins (ERMs) are plasma membrane (PM) organizers that link the actin cytoskeleton to the cytoplasmic tail of transmembrane proteins, many of which are adhesion receptors, in order to regulate the formation of F-actin-based structures (e.g., microspikes and microvilli). ERMs also effect transmission of signals from the PM into the cell, an action mainly exerted through the compartmentalized activation of the small Rho GTPases Rho, Rac and Cdc42. Ezrin and moesin are the ERMs more highly expressed in leukocytes, and although they do not always share functions, both are mainly regulated through phosphatidylinositol 4,5-bisphosphate (PIP2) binding to the N-terminal band 4.1 protein-ERM (FERM) domain and phosphorylation of a conserved Thr in the C-terminal ERM association domain (C-ERMAD), exerting their functions through a wide assortment of mechanisms. In this review we will discuss some of these mechanisms, focusing on how they regulate polarization and migration in leukocytes, and formation of actin-based cellular structures like the phagocytic cup-endosome and the immune synapse in macrophages/neutrophils and lymphocytes, respectively, which represent essential aspects of the effector immune response.
Collapse
|
10
|
Abou-Fadel J, Vasquez M, Grajeda B, Ellis C, Zhang J. Systems-wide analysis unravels the new roles of CCM signal complex (CSC). Heliyon 2019; 5:e02899. [PMID: 31872111 PMCID: PMC6909108 DOI: 10.1016/j.heliyon.2019.e02899] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/17/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Cerebral cavernous malformations (CCMs) are characterized by abnormally dilated intracranial capillaries that result in increased susceptibility to stroke. Three genes have been identified as causes of CCMs; KRIT1 (CCM1), MGC4607 (CCM2) and PDCD10 (CCM3); one of them is disrupted in most CCM cases. It was demonstrated that both CCM1 and CCM3 bind to CCM2 to form a CCM signaling complex (CSC) to modulate angiogenesis. In this report, we deployed both RNA-seq and proteomic analysis of perturbed CSC after depletion of one of three CCM genes to generate interactomes for system-wide studies. Our results demonstrated a unique portrait detailing alterations in angiogenesis and vascular integrity. Interestingly, only in-direct overlapped alterations between RNA and protein levels were detected, supporting the existence of multiple layers of regulation in CSC cascades. Notably, this is the first report identifying that both β4 integrin and CAV1 signaling are downstream of CSC, conveying the angiogenic signaling. Our results provide a global view of signal transduction modulated by the CSC, identifies novel regulatory signaling networks and key cellular factors associated with CSC.
Collapse
Affiliation(s)
- Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Mariana Vasquez
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Brian Grajeda
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Cameron Ellis
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, TX, 79905, USA
| |
Collapse
|
11
|
Tangye SG, Bucciol G, Casas‐Martin J, Pillay B, Ma CS, Moens L, Meyts I. Human inborn errors of the actin cytoskeleton affecting immunity: way beyond WAS and WIP. Immunol Cell Biol 2019; 97:389-402. [DOI: 10.1111/imcb.12243] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/09/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Stuart G Tangye
- Immunology Division Garvan Institute of Medical Research Sydney NSW Australia
- Faculty of Medicine St Vincent's Clinical School UNSW Sydney Sydney NSW Australia
| | - Giorgia Bucciol
- Laboratory for Inborn Errors of Immunity Department of Microbiology and Immunology KU Leuven Leuven Belgium
- Department of Pediatrics University Hospitals Leuven Leuven Belgium
| | - Jose Casas‐Martin
- Laboratory for Inborn Errors of Immunity Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - Bethany Pillay
- Immunology Division Garvan Institute of Medical Research Sydney NSW Australia
- Faculty of Medicine St Vincent's Clinical School UNSW Sydney Sydney NSW Australia
| | - Cindy S Ma
- Immunology Division Garvan Institute of Medical Research Sydney NSW Australia
- Faculty of Medicine St Vincent's Clinical School UNSW Sydney Sydney NSW Australia
| | - Leen Moens
- Laboratory for Inborn Errors of Immunity Department of Microbiology and Immunology KU Leuven Leuven Belgium
| | - Isabelle Meyts
- Laboratory for Inborn Errors of Immunity Department of Microbiology and Immunology KU Leuven Leuven Belgium
- Department of Pediatrics University Hospitals Leuven Leuven Belgium
| |
Collapse
|
12
|
Rey-Gallardo A, Tomlins H, Joachim J, Rahman I, Kitscha P, Frudd K, Parsons M, Ivetic A. Sequential binding of ezrin and moesin to L-selectin regulates monocyte protrusive behaviour during transendothelial migration. J Cell Sci 2018; 131:jcs.215541. [PMID: 29777033 PMCID: PMC6051341 DOI: 10.1242/jcs.215541] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/02/2018] [Indexed: 01/21/2023] Open
Abstract
Leukocyte transendothelial migration (TEM) is absolutely fundamental to the inflammatory response, and involves initial pseudopod protrusion and subsequent polarised migration across inflamed endothelium. Ezrin/radixin/moesin (ERM) proteins are expressed in leukocytes and mediate cell shape changes and polarity. The spatio-temporal organisation of ERM proteins with their targets, and their individual contribution to protrusion during TEM, has never been explored. Here, we show that blocking binding of moesin to phosphatidylinositol 4,5-bisphosphate (PIP2) reduces its C-terminal phosphorylation during monocyte TEM, and that on–off cycling of ERM activity is essential for pseudopod protrusion into the subendothelial space. Reactivation of ERM proteins within transmigrated pseudopods re-establishes their binding to targets, such as L-selectin. Knockdown of ezrin, but not moesin, severely impaired the recruitment of monocytes to activated endothelial monolayers under flow, suggesting that this protein plays a unique role in the early recruitment process. Ezrin binds preferentially to L-selectin in resting cells and during early TEM. The moesin–L-selectin interaction increases within transmigrated pseudopods as TEM proceeds, facilitating localised L-selectin ectodomain shedding. In contrast, a non-cleavable L-selectin mutant binds selectively to ezrin, driving multi-pseudopodial extensions. Taken together, these results show that ezrin and moesin play mutually exclusive roles in modulating L-selectin signalling and shedding to control protrusion dynamics and polarity during monocyte TEM. Summary: Ezrin and moesin co-ordinate binding to L-selectin in monocytes to, respectively, regulate pseudopod protrusion and ectodomain shedding during transendothelial migration.
Collapse
Affiliation(s)
- Angela Rey-Gallardo
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Hannah Tomlins
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Justin Joachim
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Izajur Rahman
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Phoebe Kitscha
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Karen Frudd
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| | - Maddy Parsons
- School of Basic & Medical Biosciences, Randall Division of Cell & Molecular Biophysics, New Hunt's House, London, SE1 1UL, UK
| | - Aleksandar Ivetic
- School of Cardiovascular Medicine and Sciences, James Black Centre, BHF Centre of Research Excellence, 125 Coldharbour Lane, King's College London, London SE5 9NU, UK
| |
Collapse
|
13
|
Ma L, Liu YP, Geng CZ, Xing LX, Zhang XH. Low-dose epirubicin inhibits ezrin-mediated metastatic behavior of breast cancer cells. TUMORI JOURNAL 2018; 97:400-5. [DOI: 10.1177/030089161109700324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and background Overexpression of ezrin contributes to the progression and invasiveness of several human cancers; however, its role in breast cancer metastasis has not been investigated in detail. Methods Ezrin expression in tissue samples from patients with invasive ductal carcinoma of the breast was detected by immunohistochemistry. Ezrin expression in a breast cancer cell line was evaluated using Western blot and RT-PCR. Results Elevated expression of ezrin was associated with lymph node metastasis and poor prognosis in patients with invasive ductal carcinoma. Ezrin expression was related to the invasiveness of breast cancer cells in vitro. Low-dose epirubicin inhibited the migration of breast cancer cells in a concentration-dependent manner without promoting cytotoxicity in vitro and decreased the expression of ezrin in a concentration-dependent manner. Conclusions Low-dose epirubicin may be antimetastatic without promoting cytotoxic effects and could serve as a target for the development of therapeutics for breast carcinoma.
Collapse
Affiliation(s)
- Li Ma
- Breast Center, Institute of the Fourth Hospital of Hebei Medical University, Shijiazhuang
| | - Yue-Ping Liu
- Department of Pathology, Institute of the Fourth Hospital of Hebei Medical University, Shijiazhuang
| | - Cui-Zhi Geng
- Breast Center, Institute of the Fourth Hospital of Hebei Medical University, Shijiazhuang
| | - Ling-Xiao Xing
- Department of Pathological Laboratory, Institute of Basic Medical Science, Hebei Medical University, Shijiazhuang, PR China
| | - Xiang-Hong Zhang
- Department of Pathological Laboratory, Institute of Basic Medical Science, Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
14
|
Ivetic A. A head-to-tail view of L-selectin and its impact on neutrophil behaviour. Cell Tissue Res 2018; 371:437-453. [PMID: 29353325 PMCID: PMC5820395 DOI: 10.1007/s00441-017-2774-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/05/2017] [Indexed: 01/04/2023]
Abstract
L-selectin is a type I transmembrane cell adhesion molecule expressed on most circulating leukocytes, including neutrophils. Engagement of L-selectin with endothelial-derived ligands initiates neutrophil tethering and rolling behaviour along luminal walls of post-capillary venules, constituting the first step of the multi-step adhesion cascade. There is a large body of evidence to suggest that signalling downstream of L-selectin can influence neutrophil behaviour: adhesion, migration and priming. This review will cover aspects of L-selectin form and function and introduce the “triad of L-selectin regulation”, highlighting the inextricable links between adhesion, signalling and ectodomain shedding and also highlighting the cytosolic proteins that interconnect them. Recent advances in how L-selectin impacts priming, transendothelial migration (TEM) and cell polarity will also be discussed.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- BHF Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, James Black Centre 125, Coldharbour Lane, London, SE5 9NU, UK.
| |
Collapse
|
15
|
Lubart Q, Vitet H, Dalonneau F, Le Roy A, Kowalski M, Lourdin M, Ebel C, Weidenhaupt M, Picart C. Role of Phosphorylation in Moesin Interactions with PIP 2-Containing Biomimetic Membranes. Biophys J 2018; 114:98-112. [PMID: 29320700 PMCID: PMC5912500 DOI: 10.1016/j.bpj.2017.10.041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 10/08/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022] Open
Abstract
Moesin, a protein of the ezrin, radixin, and moesin family, which links the plasma membrane to the cytoskeleton, is involved in multiple physiological and pathological processes, including viral budding and infection. Its interaction with the plasma membrane occurs via a key phosphoinositide, the phosphatidyl(4,5)inositol-bisphosphate (PIP2), and phosphorylation of residue T558, which has been shown to contribute, in cellulo, to a conformationally open protein. We study the impact of a double phosphomimetic mutation of moesin (T235D, T558D), which mimics the phosphorylation state of the protein, on protein/PIP2/microtubule interactions. Analytical ultracentrifugation in the micromolar range showed moesin in the monomer and dimer forms, with wild-type (WT) moesin containing a slightly larger fraction (∼30%) of dimers than DD moesin (10-20%). Only DD moesin was responsive to PIP2 in its micellar form. Quantitative cosedimentation assays using large unilamellar vesicles and quartz crystal microbalance on supported lipid bilayers containing PIP2 reveal a specific cooperative interaction for DD moesin with an ability to bind two PIP2 molecules simultaneously, whereas WT moesin was able to bind only one. In addition, DD moesin could subsequently interact with microtubules, whereas WT moesin was unable to do so. Altogether, our results point to an important role of these two phosphorylation sites in the opening of moesin: since DD moesin is intrinsically in a more open conformation than WT moesin, this intermolecular interaction is reinforced by its binding to PIP2. We also highlight important differences between moesin and ezrin, which appear to be finely regulated and to exhibit distinct molecular behaviors.
Collapse
Affiliation(s)
- Quentin Lubart
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Helene Vitet
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Fabien Dalonneau
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Aline Le Roy
- Institut de Biologie Structurale (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Mathieu Kowalski
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Morgane Lourdin
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Christine Ebel
- Institut de Biologie Structurale (IBS), University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Marianne Weidenhaupt
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), University Grenoble Alpes, CEA, CNRS, Grenoble, France; Institut National Polytechnique de Grenoble, University Grenoble Alpes, CEA, CNRS, Grenoble, France.
| |
Collapse
|
16
|
Huang L, Zhang Y, Xu C, Gu X, Niu L, Wang J, Sun X, Bai X, Xuan X, Li Q, Shi C, Yu B, Miller H, Yang G, Westerberg LS, Liu W, Song W, Zhao X, Liu C. Rictor positively regulates B cell receptor signaling by modulating actin reorganization via ezrin. PLoS Biol 2017; 15:e2001750. [PMID: 28821013 PMCID: PMC5562439 DOI: 10.1371/journal.pbio.2001750] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/25/2017] [Indexed: 01/13/2023] Open
Abstract
As the central hub of the metabolism machinery, the mammalian target of rapamycin complex 2 (mTORC2) has been well studied in lymphocytes. As an obligatory component of mTORC2, the role of Rictor in T cells is well established. However, the role of Rictor in B cells still remains elusive. Rictor is involved in B cell development, especially the peripheral development. However, the role of Rictor on B cell receptor (BCR) signaling as well as the underlying cellular and molecular mechanism is still unknown. This study used B cell-specfic Rictor knockout (KO) mice to investigate how Rictor regulates BCR signaling. We found that the key positive and negative BCR signaling molecules, phosphorylated Brutons tyrosine kinase (pBtk) and phosphorylated SH2-containing inositol phosphatase (pSHIP), are reduced and enhanced, respectively, in Rictor KO B cells. This suggests that Rictor positively regulates the early events of BCR signaling. We found that the cellular filamentous actin (F-actin) is drastically increased in Rictor KO B cells after BCR stimulation through dysregulating the dephosphorylation of ezrin. The high actin-ezrin intensity area restricts the lateral movement of BCRs upon stimulation, consequently reducing BCR clustering and BCR signaling. The reduction in the initiation of BCR signaling caused by actin alteration is associated with a decreased humoral immune response in Rictor KO mice. The inhibition of actin polymerization with latrunculin in Rictor KO B cells rescues the defects of BCR signaling and B cell differentiation. Overall, our study provides a new pathway linking cell metablism to BCR activation, in which Rictor regulates BCR signaling via actin reorganization.
Collapse
Affiliation(s)
- Lu Huang
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjie Zhang
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Chenguang Xu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Xiaomei Gu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Linlin Niu
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Jinzhi Wang
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Sun
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoming Bai
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xingtian Xuan
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Qubei Li
- Children’s Hospital Respiratory Center of Chongqing Medical University, Chongqing, China
| | - Chunwei Shi
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Yu
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, United States of America
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lisa S. Westerberg
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Institute for Immunology, Tsinghua University, Beijing, China
| | - Wenxia Song
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, United States of America
| | - Xiaodong Zhao
- Chongqing Key Laboratory of Child Infection and Immunity, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
PSGL-1: A New Player in the Immune Checkpoint Landscape. Trends Immunol 2017; 38:323-335. [PMID: 28262471 DOI: 10.1016/j.it.2017.02.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/02/2017] [Accepted: 02/08/2017] [Indexed: 02/07/2023]
Abstract
P-selectin glycoprotein ligand-1 (PSGL-1) has long been studied as an adhesion molecule involved in immune cell trafficking and is recognized as a regulator of many facets of immune responses by myeloid cells. PSGL-1 also regulates T cell migration during homeostasis and inflammatory settings. However, recent findings indicate that PSGL-1 can also negatively regulate T cell function. Because T cell differentiation is finely tuned by multiple positive and negative regulatory signals that appropriately scale the magnitude of the immune response, PSGL-1 has emerged as an important checkpoint during this process. We summarize what is known regarding PSGL-1 structure and function and highlight how it may act as an immune checkpoint inhibitor in T cells.
Collapse
|
18
|
Hartzell CA, Jankowska KI, Burkhardt JK, Lewis RS. Calcium influx through CRAC channels controls actin organization and dynamics at the immune synapse. eLife 2016; 5. [PMID: 27440222 PMCID: PMC4956410 DOI: 10.7554/elife.14850] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/13/2016] [Indexed: 11/21/2022] Open
Abstract
T cell receptor (TCR) engagement opens Ca2+ release-activated Ca2+ (CRAC) channels and triggers formation of an immune synapse between T cells and antigen-presenting cells. At the synapse, actin reorganizes into a concentric lamellipod and lamella with retrograde actin flow that helps regulate the intensity and duration of TCR signaling. We find that Ca2+ influx is required to drive actin organization and dynamics at the synapse. Calcium acts by promoting actin depolymerization and localizing actin polymerization and the actin nucleation promotion factor WAVE2 to the periphery of the lamellipod while suppressing polymerization elsewhere. Ca2+-dependent retrograde actin flow corrals ER tubule extensions and STIM1/Orai1 complexes to the synapse center, creating a self-organizing process for CRAC channel localization. Our results demonstrate a new role for Ca2+ as a critical regulator of actin organization and dynamics at the synapse, and reveal potential feedback loops through which Ca2+ influx may modulate TCR signaling. DOI:http://dx.doi.org/10.7554/eLife.14850.001 An effective immune response requires the immune system to rapidly recognize and respond to foreign invaders. Immune cells known as T cells recognize infection through a protein on their surface known as the T cell receptor. The T cell receptor binds to foreign proteins displayed on the surface of other cells. This interaction initiates a chain of events, including the opening of calcium channels embedded in the T cell membrane known as CRAC channels, which allows calcium ions to flow into the cell. These events ultimately lead to the activation of the T cell, enabling it to mount an immune response against the foreign invader. As part of the activation process, the T cell spreads over the surface of the cell that is displaying foreign proteins to form an extensive interface known as an immune synapse. The movement of the T cell's internal skeleton (the cytoskeleton) is crucial for the formation and function of the synapse. Actin filaments, a key component of the cytoskeleton, flow from the edge of the synapse toward the center; these rearrangements of the actin cytoskeleton help to transport clusters of T cell receptors to the center of the synapse and enable the T cell receptors to transmit signals that lead to the T cell being activated. It is not entirely clear how the binding of T cell receptors to foreign proteins drives the actin rearrangements, but there is indirect evidence suggesting that calcium ions may be involved. Hartzell et al. have now investigated the interactions between calcium and the actin cytoskeleton at the immune synapse in human T cells. T cells were placed on glass so that they formed immune synapse-like connections with the surface, and actin movements at the synapse were visualized using a specialized type of fluorescence microscopy. When calcium ions were prevented from entering the T cell, the movement of actin stopped almost entirely. Thus, the flow of calcium ions into the T cell through CRAC channels is essential for driving the actin movements that underlie immune synapse development and T cell activation. In further experiments, Hartzell et al. tracked the movements of CRAC channels and actin at the synapse and found that actin filaments create a constricting “corral” that concentrates CRAC channels in the center of the synapse. Thus, by driving cytoskeleton movement, calcium ions also help to organize calcium channels at the immune synapse. Future work will focus on identifying the actin remodeling proteins that enable calcium ions to control this process. DOI:http://dx.doi.org/10.7554/eLife.14850.002
Collapse
Affiliation(s)
- Catherine A Hartzell
- Immunology Program, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| | - Katarzyna I Jankowska
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, United States.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Richard S Lewis
- Immunology Program, Stanford University, Stanford, United States.,Department of Molecular and Cellular Physiology, Stanford University, Stanford, United States
| |
Collapse
|
19
|
Mattila PK, Batista FD, Treanor B. Dynamics of the actin cytoskeleton mediates receptor cross talk: An emerging concept in tuning receptor signaling. J Cell Biol 2016; 212:267-80. [PMID: 26833785 PMCID: PMC4748574 DOI: 10.1083/jcb.201504137] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent evidence implicates the actin cytoskeleton in the control of receptor signaling. This may be of particular importance in the context of immune receptors, such as the B cell receptor, where dysregulated signaling can result in autoimmunity and malignancy. Here, we discuss the role of the actin cytoskeleton in controlling receptor compartmentalization, dynamics, and clustering as a means to regulate receptor signaling through controlling the interactions with protein partners. We propose that the actin cytoskeleton is a point of integration for receptor cross talk through modulation of protein dynamics and clustering. We discuss the implication of this cross talk via the cytoskeleton for both ligand-induced and low-level constitutive (tonic) signaling necessary for immune cell survival.
Collapse
Affiliation(s)
- Pieta K Mattila
- Institute of Biomedicine, MediCity, University of Turku, 20520 Turku, Finland
| | - Facundo D Batista
- Lymphocyte Interaction Laboratory, The Francis Crick Institute, Cancer Research UK, London WC2A 3LY, England, UK
| | - Bebhinn Treanor
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M5T 1C6, Canada
| |
Collapse
|
20
|
Wehbi VL, Taskén K. Molecular Mechanisms for cAMP-Mediated Immunoregulation in T cells - Role of Anchored Protein Kinase A Signaling Units. Front Immunol 2016; 7:222. [PMID: 27375620 PMCID: PMC4896925 DOI: 10.3389/fimmu.2016.00222] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
The cyclic AMP/protein kinase A (cAMP/PKA) pathway is one of the most common and versatile signal pathways in eukaryotic cells. A-kinase anchoring proteins (AKAPs) target PKA to specific substrates and distinct subcellular compartments providing spatial and temporal specificity for mediation of biological effects channeled through the cAMP/PKA pathway. In the immune system, cAMP is a potent negative regulator of T cell receptor-mediated activation of effector T cells (Teff) acting through a proximal PKA/Csk/Lck pathway anchored via a scaffold consisting of the AKAP Ezrin holding PKA, the linker protein EBP50, and the anchoring protein phosphoprotein associated with glycosphingolipid-enriched microdomains holding Csk. As PKA activates Csk and Csk inhibits Lck, this pathway in response to cAMP shuts down proximal T cell activation. This immunomodulating pathway in Teff mediates clinically important responses to regulatory T cell (Treg) suppression and inflammatory mediators, such as prostaglandins (PGs), adrenergic stimuli, adenosine, and a number of other ligands. A major inducer of T cell cAMP levels is PG E2 (PGE2) acting through EP2 and EP4 prostanoid receptors. PGE2 plays a crucial role in the normal physiological control of immune homeostasis as well as in inflammation and cancer immune evasion. Peripherally induced Tregs express cyclooxygenase-2, secrete PGE2, and elicit the immunosuppressive cAMP pathway in Teff as one tumor immune evasion mechanism. Moreover, a cAMP increase can also be induced by indirect mechanisms, such as intercellular transfer between T cells. Indeed, Treg, known to have elevated levels of intracellular cAMP, may mediate their suppressive function by transferring cAMP to Teff through gap junctions, which we speculate could also be regulated by PKA/AKAP complexes. In this review, we present an updated overview on the influence of cAMP-mediated immunoregulatory mechanisms acting through localized cAMP signaling and the therapeutical increasing prospects of AKAPs disruptors in T-cell immune function.
Collapse
Affiliation(s)
- Vanessa L. Wehbi
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Kjetil Taskén
- Nordic EMBL Partnership, Centre for Molecular Medicine Norway, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Inflammation Research Centre, Oslo University Hospital, Oslo, Norway
- Biotechnology Centre, Oslo University Hospital, University of Oslo, Oslo, Norway
- Jebsen Centre for Cancer Immunotherapy, Oslo University Hospital, Oslo, Norway
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
21
|
X-linked primary immunodeficiency associated with hemizygous mutations in the moesin (MSN) gene. J Allergy Clin Immunol 2016; 138:1681-1689.e8. [PMID: 27405666 DOI: 10.1016/j.jaci.2016.04.032] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/26/2016] [Accepted: 04/06/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND We investigated 7 male patients (from 5 different families) presenting with profound lymphopenia, hypogammaglobulinemia, fluctuating monocytopenia and neutropenia, a poor immune response to vaccine antigens, and increased susceptibility to bacterial and varicella zoster virus infections. OBJECTIVE We sought to characterize the genetic defect involved in a new form of X-linked immunodeficiency. METHODS We performed genetic analyses and an exhaustive phenotypic and functional characterization of the lymphocyte compartment. RESULTS We observed hemizygous mutations in the moesin (MSN) gene (located on the X chromosome and coding for MSN) in all 7 patients. Six of the latter had the same missense mutation, which led to an amino acid substitution (R171W) in the MSN four-point-one, ezrin, radixin, moesin domain. The seventh patient had a nonsense mutation leading to a premature stop codon mutation (R533X). The naive T-cell counts were particularly low for age, and most CD8+ T cells expressed the senescence marker CD57. This phenotype was associated with impaired T-cell proliferation, which was rescued by expression of wild-type MSN. MSN-deficient T cells also displayed poor chemokine receptor expression, increased adhesion molecule expression, and altered migration and adhesion capacities. CONCLUSION Our observations establish a causal link between an ezrin-radixin-moesin protein mutation and a primary immunodeficiency that could be referred to as X-linked moesin-associated immunodeficiency.
Collapse
|
22
|
Roles for lipid heterogeneity in immunoreceptor signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:830-836. [PMID: 26995463 DOI: 10.1016/j.bbalip.2016.03.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 11/22/2022]
Abstract
Immune receptors that specifically recognize foreign antigens to activate leukocytes in adaptive immune responses belong to a family of multichain cell surface proteins. All of these contain immunoreceptor tyrosine-based activation motifs in one or more subunits that initiate signaling cascades following stimulated tyrosine phosphorylation by Src-family kinases. As highlighted in this review, lipids participate in this initial activation step, as well as in more downstream signaling steps. We summarize evidence for cholesterol-dependent ordered lipids serving to regulate the store-operated Ca(2+) channel, Orai1, and we describe the sensitivity of Orai1 coupling to the ER Ca(2+) sensor, STIM1, to inhibition by polyunsaturated fatty acids. Phosphoinositides play key roles in regulating STIM1-Orai1 coupling, as well as in the stimulated Ca(2+) oscillations that are a consequence of IgE receptor signaling in mast cells. They also participate in the coupling between the plasma membrane and the actin cytoskeleton, which regulates immune receptor responses in T cells, B cells, and mast cells, both positively and negatively, depending on the cellular context. Recent studies show that other phospholipids with mostly saturated acylation also participate in coupling between receptors and the actin cytoskeleton. Lipid heterogeneity is a central feature of the intimate relationship between the plasma membrane and the actin cytoskeleton. The detailed nature of these interactions and how they are dynamically regulated to initiate and propagate receptor-mediated cell signaling are challenging questions for further investigation. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
|
23
|
Jiang H, Promchan K, Lin BR, Lockett S, Chen D, Marshall H, Badralmaa Y, Natarajan V. LZTFL1 Upregulated by All-Trans Retinoic Acid during CD4+ T Cell Activation Enhances IL-5 Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:1081-90. [PMID: 26700766 PMCID: PMC4724573 DOI: 10.4049/jimmunol.1500719] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 11/17/2015] [Indexed: 01/05/2023]
Abstract
Retinoic acids, which are metabolites of vitamin A, have been shown to be involved in multiple T cell effector responses through their binding to the retinoic acid receptor, a ligand-activated transcription factor. Because the molecular mechanism of regulation by retinoic acid is still not fully uncovered, we investigated the gene expression profile of all-trans retinoic acid (ATRA)-treated human CD4(+) T cells. Leucine zipper transcription factor-like 1 (LZTFL1) was upregulated by ATRA in a dose- and time-dependent manner. The expression of LZTFL1 depended on both ATRA and TCR signaling. LZTFL1 accumulated in the plasma membrane compartment of human CD4(+) T cells, and, during immunological synapse formation, it transiently redistributed to the T cell and APC contact zone, indicating its role in T cell activation. Live-cell imaging demonstrates that at the initial stage of immunological synapse formation, LZTFL1 is concentrated at the APC contact site, and, during later stages, it relocates to the distal pole. Knockdown of LZTFL1 reduced the basal- and ATRA-induced levels of IL-5 in CD4(+) T cells, and overexpression of LZTFL1 enhanced the TCR-mediated NFAT signaling, suggesting that LZTFL1 is an important regulator of ATRA-induced T cell response. Together, these data indicate that LZTFL1 modulates T cell activation and IL-5 levels.
Collapse
Affiliation(s)
- Hong Jiang
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Kanyarat Promchan
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Bor-Ruei Lin
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - De Chen
- Optical Microscopy and Analysis Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Heather Marshall
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Yunden Badralmaa
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| | - Ven Natarajan
- Laboratory of Molecular Cell Biology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702; and
| |
Collapse
|
24
|
Cascio G, Martín-Cófreces NB, Rodríguez-Frade JM, López-Cotarelo P, Criado G, Pablos JL, Rodríguez-Fernández JL, Sánchez-Madrid F, Mellado M. CXCL12 Regulates through JAK1 and JAK2 Formation of Productive Immunological Synapses. THE JOURNAL OF IMMUNOLOGY 2015; 194:5509-19. [PMID: 25917087 DOI: 10.4049/jimmunol.1402419] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/23/2015] [Indexed: 11/19/2022]
Abstract
The adaptive immune response requires interaction between T cells and APC to form a specialized structure termed the immune synapse (IS). Although the TCR is essential for IS organization, other factors such as chemokines participate in this process. In this study, we show that the chemokine CXCL12-mediated signaling contributes to correct IS organization and therefore influences T cell activation. CXCR4 downregulation or blockade on T cells caused defective actin polymerization at the contact site with APC, altered microtubule-organizing center polarization and the IS structure, and reduced T cell/APC contact duration. T cell activation was thus inhibited, as shown by reduced expression of CD25 and CD69 markers and of IL-2 mRNA levels. The results indicate that, through Gi and JAK1 and 2 kinases activation, CXCL12 signaling cooperates to build the IS and to maintain adhesive contacts between APC and T cells, required for continuous TCR signaling.
Collapse
Affiliation(s)
- Graciela Cascio
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, E-28049 Madrid, Spain
| | - Noa B Martín-Cófreces
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, E-28006 Madrid, Spain
| | - José Miguel Rodríguez-Frade
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, E-28049 Madrid, Spain
| | - Pilar López-Cotarelo
- Departamento de Biología Vascular e Inflamación, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, E-28029 Madrid, Spain; Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, E-28040 Madrid, Spain; and
| | - Gabriel Criado
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Sanitaria Hospital 12 de Octubre, E-28041 Madrid, Spain
| | - José L Pablos
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Sanitaria Hospital 12 de Octubre, E-28041 Madrid, Spain
| | - José Luis Rodríguez-Fernández
- Departamento de Biología Vascular e Inflamación, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, E-28029 Madrid, Spain; Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, E-28040 Madrid, Spain; and
| | - Francisco Sánchez-Madrid
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, E-28006 Madrid, Spain
| | - Mario Mellado
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Cientificas, E-28049 Madrid, Spain;
| |
Collapse
|
25
|
Surface expression of the hRSV nucleoprotein impairs immunological synapse formation with T cells. Proc Natl Acad Sci U S A 2014; 111:E3214-23. [PMID: 25056968 DOI: 10.1073/pnas.1400760111] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human respiratory syncytial virus (hRSV) is the leading cause of bronchiolitis and pneumonia in young children worldwide. The recurrent hRSV outbreaks and reinfections are the cause of a significant public health burden and associate with an inefficient antiviral immunity, even after disease resolution. Although several mouse- and human cell-based studies have shown that hRSV infection prevents naïve T-cell activation by antigen-presenting cells, the mechanism underlying such inhibition remains unknown. Here, we show that the hRSV nucleoprotein (N) could be at least partially responsible for inhibiting T-cell activation during infection by this virus. Early after infection, the N protein was expressed on the surface of epithelial and dendritic cells, after interacting with trans-Golgi and lysosomal compartments. Further, experiments on supported lipid bilayers loaded with peptide-MHC (pMHC) complexes showed that surface-anchored N protein prevented immunological synapse assembly by naive CD4(+) T cells and, to a lesser extent, by antigen-experienced T-cell blasts. Synapse assembly inhibition was in part due to reduced T-cell receptor (TCR) signaling and pMHC clustering at the T-cell-bilayer interface, suggesting that N protein interferes with pMHC-TCR interactions. Moreover, N protein colocalized with the TCR independently of pMHC, consistent with a possible interaction with TCR complex components. Based on these data, we conclude that hRSV N protein expression at the surface of infected cells inhibits T-cell activation. Our study defines this protein as a major virulence factor that contributes to impairing acquired immunity and enhances susceptibility to reinfection by hRSV.
Collapse
|
26
|
Soares H, Lasserre R, Alcover A. Orchestrating cytoskeleton and intracellular vesicle traffic to build functional immunological synapses. Immunol Rev 2014; 256:118-32. [PMID: 24117817 DOI: 10.1111/imr.12110] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunological synapses are specialized cell-cell contacts formed between T lymphocytes and antigen-presenting cells. They are induced upon antigen recognition and are crucial for T-cell activation and effector functions. The generation and function of immunological synapses depend on an active T-cell polarization process, which results from a finely orchestrated crosstalk between the antigen receptor signal transduction machinery, the actin and microtubule cytoskeletons, and controlled vesicle traffic. Although we understand how some of these particular events are regulated, we still lack knowledge on how these multiple cellular elements are harmonized to ensure appropriate T-cell responses. We discuss here our view on how T-cell receptor signal transduction initially commands cytoskeletal and vesicle traffic polarization, which in turn sets the immunological synapse molecular design that regulates T-cell activation. We also discuss how the human immunodeficiency virus (HIV-1) hijacks some of these processes impairing immunological synapse generation and function.
Collapse
Affiliation(s)
- Helena Soares
- Institut Pasteur, Department of Immunology, Lymphocyte Cell Biology Unit, Paris, France; CNRS, URA-1961, Paris, France
| | | | | |
Collapse
|
27
|
Babich A, Burkhardt JK. Coordinate control of cytoskeletal remodeling and calcium mobilization during T-cell activation. Immunol Rev 2014; 256:80-94. [PMID: 24117814 DOI: 10.1111/imr.12123] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
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.
Collapse
Affiliation(s)
- Alexander Babich
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
28
|
Ezrin is a component of the HIV-1 virological presynapse and contributes to the inhibition of cell-cell fusion. J Virol 2014; 88:7645-58. [PMID: 24760896 DOI: 10.1128/jvi.00550-14] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED During cell-to-cell transmission of HIV-1, viral and cellular proteins transiently accumulate at the contact zone between infected (producer) and uninfected (target) cells, forming the virological synapse. Rearrangements of the cytoskeleton in producer and target cells are required for proper targeting of viral and cellular components during synapse formation, yet little is known about how these processes are regulated, particularly within the producer cell. Since ezrin-radixin-moesin (ERM) proteins connect F-actin with integral and peripheral membrane proteins, are incorporated into virions, and interact with cellular components of the virological presynapse, we hypothesized that they play roles during the late stage of HIV-1 replication. Here we document that phosphorylated (i.e., active) ezrin specifically accumulates at the HIV-1 presynapse in T cell lines and primary CD4(+) lymphocytes. To investigate whether ezrin supports virus transmission, we sought to ablate ezrin expression in producer cells. While cells did not tolerate a complete knockdown of ezrin, even a modest reduction of ezrin expression (~50%) in HIV-1-producing cells led to the release of particles with impaired infectivity. Further, when cocultured with uninfected target cells, ezrin-knockdown producer cells displayed reduced accumulation of the tetraspanin CD81 at the synapse and fused more readily with target cells, thus forming syncytia. Such an outcome likely is not optimal for virus dissemination, as evidenced by the fact that, in vivo, only relatively few infected cells form syncytia. Thus, ezrin likely helps secure efficient virus spread not only by enhancing virion infectivity but also by preventing excessive membrane fusion at the virological synapse. IMPORTANCE While viruses, in principal, can propagate through successions of syncytia, HIV-1-infected cells in the majority of cases do not fuse with potential target cells during viral transmission. This mode of spread is coresponsible for key features of HIV-1 pathogenesis, including killing of bystander cells and establishment of latently infected T lymphocytes. Here we identify the ERM protein family member ezrin as a cellular factor that contributes to the inhibition of cell-cell fusion and thus to suppressing excessive syncytium formation. Our analyses further suggest that ezrin, which connects integral membrane proteins with actin, functions in concert with CD81, a member of the tetraspanin family of proteins. Additional evidence, documented here and elsewhere, suggests that ezrin and CD81 cooperate to prevent cytoskeleton rearrangements that need to take place during the fusion of cellular membranes.
Collapse
|
29
|
Niggli V. Insights into the mechanism for dictating polarity in migrating T-cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 312:201-70. [PMID: 25262243 DOI: 10.1016/b978-0-12-800178-3.00007-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This review is focused on mechanisms of chemokine-induced polarization of T-lymphocytes. Polarization involves, starting from spherical cells, formation of a morphologically and functionally different rear (uropod) and front (leading edge). This polarization is required for efficient random and directed T-cell migration. The addressed topics concern the specific location of cell organelles and of receptors, signaling molecules, and cytoskeletal proteins in chemokine-stimulated polarized T-cells. In chemokine-stimulated, polarized T-cells, specific proteins, signaling molecules and organelles show enrichment either in the rear, the midzone, or the front; different from the random location in spherical resting cells. Possible mechanisms involved in this asymmetric location will be discussed. A major topic is also the functional role of proteins and cell organelles in T-cell polarization and migration. Specifically, the roles of adhesion and chemokine receptors, cytoskeletal proteins, signaling molecules, scaffolding proteins, and membrane microdomains in these processes will be discussed. The polarity which is established during contact formation of T-cells with antigen-presenting cells is not discussed in detail.
Collapse
Affiliation(s)
- Verena Niggli
- Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
30
|
Huse M, Le Floc'h A, Liu X. From lipid second messengers to molecular motors: microtubule-organizing center reorientation in T cells. Immunol Rev 2013; 256:95-106. [PMID: 24117815 PMCID: PMC4595039 DOI: 10.1111/imr.12116] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In T lymphocytes, polarization of the microtubule-organizing center (MTOC) to the immunological synapse enables the directional secretion of cytokines, cytolytic factors, and other soluble molecules toward the antigen-presenting cell. This is likely to be crucial for maintaining the specificity of T-cell effector responses. Here, we review recent advances in our understanding of MTOC reorientation in T cells, focusing first on the importance of diacylglycerol and protein kinase C isozymes and then on the molecular motor proteins that function downstream to drive MTOC movement.
Collapse
Affiliation(s)
- Morgan Huse
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Audrey Le Floc'h
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Xin Liu
- Immunology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
31
|
Abstract
It has been over 30 years since the reorganization of both the microtubule network and a 'peculiar actin polarization' was reported at the contact area of cytotoxic T lymphocytes interacting with target cells. Since that time, hundreds of studies have been published in an effort to elucidate the structure and function of the microtubule network and the actin cytoskeleton in T-cell activation, migration, and effector function at the interface between a T cell and its cognate antigen-presenting cell or target cell. This interface has become known as the immunological synapse, and this review examines some of the roles played by the cytoskeleton at the synapse.
Collapse
Affiliation(s)
- Alex T Ritter
- Cambridge Institute for Medical Research, University of Cambridge Biomedical CampusCambridge, UK
| | - Karen L Angus
- Cambridge Institute for Medical Research, University of Cambridge Biomedical CampusCambridge, UK
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, University of Cambridge Biomedical CampusCambridge, UK
| |
Collapse
|
32
|
Viswanatha R, Wayt J, Ohouo PY, Smolka MB, Bretscher A. Interactome analysis reveals ezrin can adopt multiple conformational states. J Biol Chem 2013; 288:35437-51. [PMID: 24151071 DOI: 10.1074/jbc.m113.505669] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ezrin, a member of the ezrin-radixin-moesin family (ERM), is an essential regulator of the structure of microvilli on the apical aspect of epithelial cells. Ezrin provides a linkage between membrane-associated proteins and F-actin, oscillating between active/open and inactive/closed states, and is regulated in part by phosphorylation of a C-terminal threonine. In the open state, ezrin can bind a number of ligands, but in the closed state the ligand-binding sites are inaccessible. In vitro analysis has proposed that there may be a third hyperactivated form of ezrin. To gain a better understanding of ezrin, we conducted an unbiased proteomic analysis of ezrin-binding proteins in an epithelial cell line, Jeg-3. We refined our list of interactors by comparing the interactomes using quantitative mass spectrometry between wild-type ezrin, closed ezrin, open ezrin, and hyperactivated ezrin. The analysis reveals several novel interactors confirmed by their localization to microvilli, as well as a significant class of proteins that bind closed ezrin. Taken together, the data indicate that ezrin can exist in three different conformational states, and different ligands "perceive" ezrin conformational states differently.
Collapse
Affiliation(s)
- Raghuvir Viswanatha
- From the Department of Molecular Biology and Genetics and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853
| | | | | | | | | |
Collapse
|
33
|
Parameswaran N, Gupta N. Re-defining ERM function in lymphocyte activation and migration. Immunol Rev 2013; 256:63-79. [DOI: 10.1111/imr.12104] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Neetha Parameswaran
- Department of Immunology; Lerner Research Institute; Cleveland Clinic; Cleveland OH USA
| | - Neetu Gupta
- Department of Immunology; Lerner Research Institute; Cleveland Clinic; Cleveland OH USA
| |
Collapse
|
34
|
Baaten BJG, Cooper AM, Swain SL, Bradley LM. Location, location, location: the impact of migratory heterogeneity on T cell function. Front Immunol 2013; 4:311. [PMID: 24115949 PMCID: PMC3792444 DOI: 10.3389/fimmu.2013.00311] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/16/2013] [Indexed: 01/13/2023] Open
Abstract
T cell migration is crucial for an effective adaptive immune response to invading pathogens. Naive and memory T cells encounter pathogen antigens, become activated, and differentiate into effector cells in secondary lymphoid tissues, and then migrate to the site(s) of infection where they exert effector activities that control and eliminate pathogens. To achieve activation, efficient effector function, and good memory formation, T cells must traffic between lymphoid and non-lymphoid tissues within the body. This complex process is facilitated by chemokine receptors, selectins, CD44, and integrins that mediate the interactions of T cells with the environment. The expression patterns of these migration receptors (MR) dictate the tissues into which the effector T cells migrate and enable them to occupy specific niches within the tissue. While MR have been considered primarily to facilitate cell movement, we highlight how the heterogeneity of signaling through these receptors influences the function and fate of T cells in situ. We explore what drives MR expression heterogeneity, how this affects migration, and how this impacts T cell effector function and memory formation.
Collapse
Affiliation(s)
- Bas J G Baaten
- Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute , La Jolla, CA , USA
| | | | | | | |
Collapse
|
35
|
Adyshev DM, Dudek SM, Moldobaeva N, Kim KM, Ma SF, Kasa A, Garcia JGN, Verin AD. Ezrin/radixin/moesin proteins differentially regulate endothelial hyperpermeability after thrombin. Am J Physiol Lung Cell Mol Physiol 2013; 305:L240-55. [PMID: 23729486 DOI: 10.1152/ajplung.00355.2012] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Endothelial cell (EC) barrier disruption induced by inflammatory agonists such as thrombin leads to potentially lethal physiological dysfunction such as alveolar flooding, hypoxemia, and pulmonary edema. Thrombin stimulates paracellular gap and F-actin stress fiber formation, triggers actomyosin contraction, and alters EC permeability through multiple mechanisms that include protein kinase C (PKC) activation. We previously have shown that the ezrin, radixin, and moesin (ERM) actin-binding proteins differentially participate in sphingosine-1 phosphate-induced EC barrier enhancement. Phosphorylation of a conserved threonine residue in the COOH-terminus of ERM proteins causes conformational changes in ERM to unmask binding sites and is considered a hallmark of ERM activation. In the present study we test the hypothesis that ERM proteins are phosphorylated on this critical threonine residue by thrombin-induced signaling events and explore the role of the ERM family in modulating thrombin-induced cytoskeletal rearrangement and EC barrier function. Thrombin promotes ERM phosphorylation at this threonine residue (ezrin Thr567, radixin Thr564, moesin Thr558) in a PKC-dependent fashion and induces translocation of phosphorylated ERM to the EC periphery. Thrombin-induced ERM threonine phosphorylation is likely synergistically mediated by protease-activated receptors PAR1 and PAR2. Using the siRNA approach, depletion of either moesin alone or of all three ERM proteins significantly attenuates thrombin-induced increase in EC barrier permeability (transendothelial electrical resistance), cytoskeletal rearrangements, paracellular gap formation, and accumulation of phospho-myosin light chain. In contrast, radixin depletion exerts opposing effects on these indexes. These data suggest that ERM proteins play important differential roles in the thrombin-induced modulation of EC permeability, with moesin promoting barrier dysfunction and radixin opposing it.
Collapse
Affiliation(s)
- Djanybek M Adyshev
- Institute for Personalized Respiratory Medicine, Department of Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, COMRB 3154, MC 719, 909 S. Wolcott Ave., Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Loss of TCR-beta F1 and/or EZRIN expression is associated with unfavorable prognosis in nodal peripheral T-cell lymphomas. Blood Cancer J 2013; 3:e111. [PMID: 23599023 PMCID: PMC3641318 DOI: 10.1038/bcj.2013.10] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Nodal peripheral T-cell lymphoma (nodal PTCL) has an unfavorable prognosis, and specific pathogenic alterations have not been fully identified. The biological and clinical relevance of the expression of CD30/T-cell receptor (TCR) genes is a topic under active investigation. One-hundred and ninety-three consecutive nodal PTCLs (89 angioimmunoblastic T-cell lymphomas (AITL) and 104 PTCL-unspecified (PTCL-not otherwise specified (NOS)) cases) were analyzed for the immunohistochemical expression of 19 molecules, involving TCR/CD30 pathways and the associations with standard prognostic indices. Mutually exclusive expression was found between CD3 and TCR-beta F1 with CD30 expression. Taking all PTCL cases together, logistic regression identified a biological score (BS) including TCR molecules (TCR-beta F1 and EZRIN) that separates two subgroups of patients with a median survival of 34.57 and 5.20 months (P<0.001). Multivariate analysis identified BS and the prognostic index for PTCL (PIT) score as independent prognostic factors. This BS maintained its significance in multivariate analysis only for the PTCL-NOS subgroup of tumors. In AITL cases, only a high level of ki67 expression was related to prognosis. A BS including molecules involved in the TCR signaling pathway proved to be an independent prognostic factor of poor outcome in a multivariate analysis, specifically in PTCL-NOS patients. Nevertheless, validation in an independent series of homogeneously treated PTCL patients is required to confirm these data.
Collapse
|
37
|
Parameswaran N, Enyindah-Asonye G, Bagheri N, Shah NB, Gupta N. Spatial coupling of JNK activation to the B cell antigen receptor by tyrosine-phosphorylated ezrin. THE JOURNAL OF IMMUNOLOGY 2013; 190:2017-26. [PMID: 23338238 DOI: 10.4049/jimmunol.1201292] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ezrin-radixin-moesin proteins regulate B lymphocyte activation via their effect on BCR diffusion and microclustering. This relies on their ability to dynamically tether the plasma membrane with actin filaments that is in turn facilitated by phosphorylation of the conserved threonine residue in the actin-binding domain. In this study, we describe a novel function of ezrin in regulating JNK activation that is mediated by phosphorylation of a tyrosine (Y353) residue that is unconserved with moesin and radixin. BCR, but not CD40, TLR4, or CXCR5 stimulation, induced phosphorylation of ezrin at Y353 in mouse splenic B cells. Ezrin existed in a preformed complex with Syk in unstimulated B cells and underwent Syk-dependent phosphorylation upon anti-IgM stimulation. Y353-phosphorylated ezrin colocalized with the BCR within minutes of stimulation and cotrafficked with the endocytosed BCRs through the early and late endosomes. The T567 residue of ezrin was rephosphorylated in late endosomes and at the plasma membrane at later times of BCR stimulation. Expression of a nonphosphorylatable Y353F mutant of ezrin specifically impaired JNK activation. BCR crosslinking induced the association of Y353-phosphorylated ezrin with JNK and its kinase MAPKK7, as well as spatial colocalization with phosphorylated JNK in the endosomes. The yellow fluorescent protein-tagged Y353F mutant displayed reduced colocalization with the endocytosed BCR as compared with wild-type ezrin-yellow fluorescent protein. Taken together, our data identify a novel role for ezrin as a spatial adaptor that couples JNK signaling components to the BCR signalosome, thus facilitating JNK activation.
Collapse
Affiliation(s)
- Neetha Parameswaran
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
38
|
Maniti O, Carvalho K, Picart C. Model membranes to shed light on the biochemical and physical properties of ezrin/radixin/moesin. Biochimie 2013; 95:3-11. [PMID: 23041444 PMCID: PMC4112940 DOI: 10.1016/j.biochi.2012.09.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/28/2012] [Indexed: 10/27/2022]
Abstract
Ezrin, radixin and moesin (ERM) proteins are now more and more recognized to play a key role in a large number of important physiological processes such as morphogenesis, cancer metastasis and virus infection. Several recent reviews extensively discuss their biological functions [1 -4 ]. In this review, we will first remind the main features of this family of proteins, which are now known as linkers and regulators of the plasma membrane/cytoskeleton linkage. We will then briefly review their implication in pathological processes such as cancer and viral infection. In a second part, we will focus on biochemical and biophysical approaches to study ERM interaction with lipid membranes and conformational change in well-defined environments. In vitro studies using biomimetic lipid membranes, especially large unilamellar vesicles (LUVs), giant unilamellar vesicles (GUVs) and supported lipid bilayers (SLBs) and recombinant proteins help to understand the molecular mechanism of conformational activation of ERM proteins. These tools are aimed to decorticate the different steps of the interaction, to simplify the experiments performed in vivo in much more complex biological environments.
Collapse
Affiliation(s)
- Ofélia Maniti
- CNRS UMR 5628 (LMGP), Grenoble Institute of Technology and CNRS, 3 parvis Louis Néel, F-38016 Grenoble Cedex, France
- Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires, UMR 5246, CNRS, Université de Lyon, Université Lyon 1, INSA-Lyon, CPE-Lyon, 43 Bd du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Kevin Carvalho
- Institut Curie, centre de recherche and CNRS UMR 168, 11 rue Pierre et Marie Curie, Paris, F-75248 cedex 5
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), Grenoble Institute of Technology and CNRS, 3 parvis Louis Néel, F-38016 Grenoble Cedex, France
| |
Collapse
|
39
|
Single-cell force spectroscopy: mechanical insights into the functional impacts of interactions between antigen-presenting cells and T cells. Immunol Res 2012; 53:108-14. [PMID: 22434515 DOI: 10.1007/s12026-012-8290-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Antigen recognition and discrimination by T lymphocyte are essential in initiating appropriate immune responses. The mechanisms underlying exquisite sensitivity and specificity of antigen discrimination are not fully elucidated but involved physical intercellular interactions between T cell and antigen-presenting cell (APC). The specificity of T-cell activation is tightly regulated by T-cell receptor (TCR) recognition of antigenic peptides in complex with major histocompatibility complex (pMHC) glycoproteins on the cell surface of APC. Antigen recognition via TCR/pMHC interactions, together with other co-receptors and co-stimulatory molecules, are spatially organized into the two-dimensional contact zone between T cells and APC, resulting in the formation of an immune synapse (IS). Here, we will review current implementations and applications of a cutting-edge biophysical technique, namely single-cell force spectroscopy (SCFS) that allows us to quantify mechanical forces of IS at APC/T cell-cell contact. The functional impacts of the mechanical strength in regulating T-cell functional activity will be discussed. We will also describe limitations of SCFS techniques, and outline recent investigations focusing on the functional roles of IS as mechanotransducer in regulating T-cell activities.
Collapse
|
40
|
Maniti O, Khalifat N, Goggia K, Dalonneau F, Guérin C, Blanchoin L, Ramos L, Picart C. Binding of moesin and ezrin to membranes containing phosphatidylinositol (4,5) bisphosphate: a comparative study of the affinity constants and conformational changes. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1818:2839-49. [PMID: 22813867 PMCID: PMC4111548 DOI: 10.1016/j.bbamem.2012.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2012] [Revised: 06/20/2012] [Accepted: 07/09/2012] [Indexed: 11/28/2022]
Abstract
The plasma membrane-cytoskeleton interface is a dynamic structure participating in a variety of cellular events. Moesin and ezrin, proteins from the ezrin/radixin/moesin (ERM) family, provide a direct linkage between the cytoskeleton and the membrane via their interaction with phosphatidylinositol 4,5-bisphosphate (PIP(2)). PIP(2) binding is considered as a prerequisite step in ERM activation. The main objective of this work was to compare moesin and ezrin interaction with PIP(2)-containing membranes in terms of affinity and to analyze secondary structure modifications leading eventually to ERM activation. For this purpose, we used two types of biomimetic model membranes, large and giant unilamellar vesicles. The dissociation constant between moesin and PIP(2)-containing large unilamellar vesicles or PIP(2)-containing giant unilamellar vesicles was found to be very similar to that between ezrin and PIP(2)-containing large unilamellar vesicles or PIP(2)-containing giant unilamellar vesicles. In addition, both proteins were found to undergo conformational changes after binding to PIP(2)-containing large unilamellar vesicles. Changes were evidenced by an increased sensitivity to proteolysis, modifications in the fluorescence intensity of the probe attached to the C-terminus and in the proportion of secondary structure elements.
Collapse
Affiliation(s)
- Ofelia Maniti
- Grenoble Institute of Technology and CNRS, Grenoble Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
41
|
McRobert EA, Young AN, Bach LA. Advanced glycation end-products induce calpain-mediated degradation of ezrin. FEBS J 2012; 279:3240-50. [PMID: 22805611 DOI: 10.1111/j.1742-4658.2012.08710.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Advanced glycation end-products (AGEs) are important mediators of diabetic complications via incompletely understood pathways. AGEs bind to intracellular ERM proteins (ezrin, radixin and moesin) that modulate cell shape, motility, adhesion and signal transduction. AGEs bind to the N-terminal domain of ezrin but not full-length ezrin. The AGE binding site may be made accessible either by proteolysis releasing an N-terminal fragment or ezrin activation by phosphorylation. Increased intracellular calcium is a primary event in cell activation by high glucose or AGEs. Calpain activity is increased concomitantly, and ezrin is a calpain substrate. The present study assessed whether glycated proteins affect ezrin cleavage and activation in renal tubule epithelial cells. After 7 days, AGE-BSA decreased ezrin levels in MDCK renal tubular cells to 66 ± 4% of control. AGE-RNAse, ribosylated fetal bovine serum and methylglyoxal-BSA all had similar effects. The AGE-BSA-induced decrease in ezrin was abolished by calpastatin peptide, a specific calpain inhibitor, and 1,2-bis-aminophenoxyethane-tetraacetic acid acetoxymethyl ester (BAPTA-AM), a calcium chelator. Ezrin breakdown products were increased in AGE-BSA-treated cells, with a main fragment of ∼ 43 kDa. In vitro, calpain 1 cleaved recombinant human ezrin, generating breakdown fragments including an N-terminal fragment of ∼ 43 kDa. Studies with ezrin mutants showed that non-phosphorylated ezrin was more susceptible to calpain cleavage. AGE-BSA decreased phosphorylated ERM levels to 31 ± 12% in MDCK cells. Thus, AGE-BSA promotes calpain-mediated proteolysis of ezrin in MDCK cells by both increasing calpain activity and reducing phosphorylation. Therapies targeting both glycated proteins and calpain may provide protection against diabetic complications.
Collapse
|
42
|
Lam Hui K, Wang C, Grooman B, Wayt J, Upadhyaya A. Membrane dynamics correlate with formation of signaling clusters during cell spreading. Biophys J 2012; 102:1524-33. [PMID: 22500752 DOI: 10.1016/j.bpj.2012.02.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 01/31/2012] [Accepted: 02/08/2012] [Indexed: 10/28/2022] Open
Abstract
The morphology and duration of contacts between cells and adhesive surfaces play a key role in several biological processes, such as cell migration, cell differentiation, and the immune response. The interaction of receptors on the cell membrane with ligands on the adhesive surface leads to triggering of signaling pathways, which allow cytoskeletal rearrangement, and large-scale deformation of the cell membrane, which allows the cell to spread over the substrate. Despite numerous studies of cell spreading, the nanometer-scale dynamics of the membrane during formation of contacts, spreading, and initiation of signaling are not well understood. Using interference reflection microscopy, we study the kinetics of cell spreading at the micron scale, as well as the topography and fluctuations of the membrane at the nanometer scale during spreading of Jurkat T cells on antibody-coated substrates. We observed two modes of spreading, which were characterized by dramatic differences in membrane dynamics and topography. Formation of signaling clusters was closely related to the movement and morphology of the membrane in contact with the activating surface. Our results suggest that cell membrane morphology may be a critical constraint on signaling at the cell-substrate interface.
Collapse
Affiliation(s)
- King Lam Hui
- Department of Physics, University of Maryland, College Park, Maryland, USA
| | | | | | | | | |
Collapse
|
43
|
Nzounza P, Chazal M, Guedj C, Schmitt A, Massé JM, Randriamampita C, Pique C, Ramirez BC. The scaffolding protein Dlg1 is a negative regulator of cell-free virus infectivity but not of cell-to-cell HIV-1 transmission in T cells. PLoS One 2012; 7:e30130. [PMID: 22272285 PMCID: PMC3260186 DOI: 10.1371/journal.pone.0030130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 12/13/2011] [Indexed: 12/28/2022] Open
Abstract
Background Cell-to-cell virus transmission of Human immunodeficiency virus type-1 (HIV-1) is predominantly mediated by cellular structures such as the virological synapse (VS). The VS formed between an HIV-1-infected T cell and a target T cell shares features with the immunological synapse (IS). We have previously identified the human homologue of the Drosophila Discs Large (Dlg1) protein as a new cellular partner for the HIV-1 Gag protein and a negative regulator of HIV-1 infectivity. Dlg1, a scaffolding protein plays a key role in clustering protein complexes in the plasma membrane at cellular contacts. It is implicated in IS formation and T cell signaling, but its role in HIV-1 cell-to-cell transmission was not studied before. Methodology/Principal Findings Kinetics of HIV-1 infection in Dlg1-depleted Jurkat T cells show that Dlg1 modulates the replication of HIV-1. Single-cycle infectivity tests show that this modulation does not take place during early steps of the HIV-1 life cycle. Immunofluorescence studies of Dlg1-depleted Jurkat T cells show that while Dlg1 depletion affects IS formation, it does not affect HIV-1-induced VS formation. Co-culture assays and quantitative cell-to-cell HIV-1 transfer analyses show that Dlg1 depletion does not modify transfer of HIV-1 material from infected to target T cells, or HIV-1 transmission leading to productive infection via cell contact. Dlg1 depletion results in increased virus yield and infectivity of the viral particles produced. Particles with increased infectivity present an increase in their cholesterol content and during the first hours of T cell infection these particles induce higher accumulation of total HIV-1 DNA. Conclusion Despite its role in the IS formation, Dlg1 does not affect the VS and cell-to-cell spread of HIV-1, but plays a role in HIV-1 cell-free virus transmission. We propose that the effect of Dlg1 on HIV-1 infectivity is at the stage of virus entry.
Collapse
Affiliation(s)
- Patrycja Nzounza
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maxime Chazal
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Chloé Guedj
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alain Schmitt
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Marc Massé
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Clotilde Randriamampita
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Claudine Pique
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertha Cecilia Ramirez
- INSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
44
|
Complementary costimulation of human T-cell subpopulations by cluster of differentiation 28 (CD28) and CD81. Proc Natl Acad Sci U S A 2012; 109:1613-8. [PMID: 22307619 DOI: 10.1073/pnas.1121307109] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cluster of differentiation 81 (CD81) is a widely expressed tetraspanin molecule that physically associates with CD4 and CD8 on the surface of human T cells. Coengagement of CD81 and CD3 results in the activation and proliferation of T cells. CD81 also costimulated mouse T cells that lack CD28, suggesting either a redundant or a different mechanism of action. Here we show that CD81 and CD28 have a preference for different subsets of T cells: Primary human naïve T cells are better costimulated by CD81, whereas the memory T-cell subsets and Tregs are better costimulated by CD28. The more efficient activation of naïve T cells by CD81 was due to prolonged signal transduction compared with that by CD28. We found that IL-6 played a role in the activation of the naïve T-cell subset by CD81. Combined costimulation through both CD28 and CD81 resulted in an additive effect on T-cell activation. Thus, these two costimulatory molecules complement each other both in the strength of signal transduction and in T-cell subset inclusions. Costimulation via CD81 might be useful for expansion of T cells for adoptive immunotherapy to allow the inclusion of naïve T cells with their broad repertoire.
Collapse
|
45
|
Mosenden R, Moltu K, Ruppelt A, Berge T, Taskén K. Effects of type I protein kinase A modulation on the T cell distal pole complex. Scand J Immunol 2011; 74:568-73. [PMID: 21854406 DOI: 10.1111/j.1365-3083.2011.02611.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The distal pole complex (DPC) assembles signalling proteins at the T cell pole opposite the immunological synapse (IS) and is thought to facilitate T cell activation by sequestering negative regulatory molecules away from the T cell receptor-proximal signalling machinery. Here, we report the translocation of type I protein kinase A (PKA) to the DPC in a fraction of T cells following activation and the localization of type I PKA with known components of the DPC. We propose that sequestration of type I PKA and concomitant loss of cAMP-mediated negative regulation at the IS may be necessary to allow full T cell activation. Moreover, composition of the DPC appears to be modulated by type I PKA activity, as the antagonist Rp-8-Br-cAMPS inhibited translocation of type I PKA and other DPC proteins.
Collapse
Affiliation(s)
- R Mosenden
- The Biotechnology Centre, University of Oslo, Oslo, Norway
| | | | | | | | | |
Collapse
|
46
|
Constitutively active ezrin increases membrane tension, slows migration, and impedes endothelial transmigration of lymphocytes in vivo in mice. Blood 2011; 119:445-53. [PMID: 22106344 DOI: 10.1182/blood-2011-07-368860] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
ERM (ezrin, radixin moesin) proteins in lymphocytes link cortical actin to plasma membrane, which is regulated in part by ERM protein phosphorylation. To assess whether phosphorylation of ERM proteins regulates lymphocyte migration and membrane tension, we generated transgenic mice whose T-lymphocytes express low levels of ezrin phosphomimetic protein (T567E). In these mice, T-cell number in lymph nodes was reduced by 27%. Lymphocyte migration rate in vitro and in vivo in lymph nodes decreased by 18% to 47%. Lymphocyte membrane tension increased by 71%. Investigations of other possible underlying mechanisms revealed impaired chemokine-induced shape change/lamellipod extension and increased integrin-mediated adhesion. Notably, lymphocyte homing to lymph nodes was decreased by 30%. Unlike most described homing defects, there was not impaired rolling or sticking to lymph node vascular endothelium but rather decreased migration across that endothelium. Moreover, decreased numbers of transgenic T cells in efferent lymph suggested defective egress. These studies confirm the critical role of ERM dephosphorylation in regulating lymphocyte migration and transmigration. Of particular note, they identify phospho-ERM as the first described regulator of lymphocyte membrane tension, whose increase probably contributes to the multiple defects observed in the ezrin T567E transgenic mice.
Collapse
|
47
|
Sun Y, Dandekar RD, Mao YS, Yin HL, Wülfing C. Phosphatidylinositol (4,5) bisphosphate controls T cell activation by regulating T cell rigidity and organization. PLoS One 2011; 6:e27227. [PMID: 22096541 PMCID: PMC3214035 DOI: 10.1371/journal.pone.0027227] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/12/2011] [Indexed: 11/19/2022] Open
Abstract
Here we investigate the role of Phosphatidylinositol (4,5) bisphosphate (PIP(2)) in the physiological activation of primary murine T cells by antigen presenting cells (APC) by addressing two principal challenges in PIP(2) biology. First, PIP(2) is a regulator of cytoskeletal dynamics and a substrate for second messenger generation. The relative importance of these two processes needs to be determined. Second, PIP(2) is turned over by multiple biosynthetic and metabolizing enzymes. The joint effect of these enzymes on PIP(2) distributions needs to be determined with resolution in time and space. We found that T cells express four isoforms of the principal PIP(2)-generating enzyme phosphatidylinositol 4-phosphate 5-kinase (PIP5K) with distinct spatial and temporal characteristics. In the context of a larger systems analysis of T cell signaling, these data identify the T cell/APC interface and the T cell distal pole as sites of differential PIP(2) turnover. Overexpression of different PIP5K isoforms, as corroborated by knock down and PIP(2) blockade, yielded an increase in PIP(2) levels combined with isoform-specific changes in the spatiotemporal distributions of accessible PIP(2). It rigidified the T cell, likely by impairing the inactivation of Ezrin Moesin Radixin, delayed and diminished the clustering of the T cell receptor at the cellular interface, reduced the efficiency of T cell proximal signaling and IL-2 secretion. These effects were consistently more severe for distal PIP5K isoforms. Thus spatially constrained cytoskeletal roles of PIP(2) in the control of T cell rigidity and spatiotemporal organization dominate the effects of PIP(2) on T cell activation.
Collapse
Affiliation(s)
- Yi Sun
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Radhika D. Dandekar
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yuntao S. Mao
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Helen L. Yin
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christoph Wülfing
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
48
|
Haynes J, Srivastava J, Madson N, Wittmann T, Barber DL. Dynamic actin remodeling during epithelial-mesenchymal transition depends on increased moesin expression. Mol Biol Cell 2011; 22:4750-64. [PMID: 22031288 PMCID: PMC3237619 DOI: 10.1091/mbc.e11-02-0119] [Citation(s) in RCA: 185] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
LifeAct-GFP, a fluorescent reporter for actin filaments, is used to uncover the dynamics of actin cytoskeleton remodeling in real time during TGF-β–induced EMT. Efficient actin filament remodeling and complete transition to a mesenchymal phenotype depend on an increase in expression of the ERM protein moesin. Remodeling of actin filaments is necessary for epithelial–mesenchymal transition (EMT); however, understanding of how this is regulated in real time is limited. We used an actin filament reporter and high-resolution live-cell imaging to analyze the regulated dynamics of actin filaments during transforming growth factor-β–induced EMT of mammary epithelial cells. Progressive changes in cell morphology were accompanied by reorganization of actin filaments from thin cortical bundles in epithelial cells to thick, parallel, contractile bundles that disassembled more slowly but remained dynamic in transdifferentiated cells. We show that efficient actin filament remodeling during EMT depends on increased expression of the ezrin/radixin/moesin (ERM) protein moesin. Cells suppressed for moesin expression by short hairpin RNA had fewer, thinner, and less stable actin bundles, incomplete morphological transition, and decreased invasive capacity. These cells also had less α-smooth muscle actin and phosphorylated myosin light chain in cortical patches, decreased abundance of the adhesion receptor CD44 at membrane protrusions, and attenuated autophosphorylation of focal adhesion kinase. Our findings suggest that increased moesin expression promotes EMT by regulating adhesion and contractile elements for changes in actin filament organization. We propose that the transciptional program driving EMT controls progressive remodeling of actin filament architectures.
Collapse
Affiliation(s)
- Jennifer Haynes
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
49
|
Lorentzen A, Bamber J, Sadok A, Elson-Schwab I, Marshall CJ. An ezrin-rich, rigid uropod-like structure directs movement of amoeboid blebbing cells. J Cell Sci 2011; 124:1256-67. [PMID: 21444753 DOI: 10.1242/jcs.074849] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Melanoma cells can switch between an elongated mesenchymal-type and a rounded amoeboid-type migration mode. The rounded 'amoeboid' form of cell movement is driven by actomyosin contractility resulting in membrane blebbing. Unlike elongated A375 melanoma cells, rounded A375 cells do not display any obvious morphological front-back polarisation, although polarisation is thought to be a prerequisite for cell movement. We show that blebbing A375 cells are polarised, with ezrin (a linker between the plasma membrane and actin cytoskeleton), F-actin, myosin light chain, plasma membrane, phosphatidylinositol (4,5)-bisphosphate and β1-integrin accumulating at the cell rear in a uropod-like structure. This structure does not have the typical protruding shape of classical leukocyte uropods, but, as for those structures, it is regulated by protein kinase C. We show that the ezrin-rich uropod-like structure (ERULS) is an inherent feature of polarised A375 cells and not a consequence of cell migration, and is necessary for cell invasion. Furthermore, we demonstrate that membrane blebbing is reduced at this site, leading to a model in which the rigid ezrin-containing structure determines the direction of a moving cell through localised inhibition of membrane blebbing.
Collapse
Affiliation(s)
- Anna Lorentzen
- Institute of Cancer Research, Cancer Research UK, Cancer Research UK Tumour Cell Signalling Unit, 237 Fulham Road, London SW3 6JB, UK
| | | | | | | | | |
Collapse
|
50
|
Adyshev DM, Moldobaeva NK, Elangovan VR, Garcia JGN, Dudek SM. Differential involvement of ezrin/radixin/moesin proteins in sphingosine 1-phosphate-induced human pulmonary endothelial cell barrier enhancement. Cell Signal 2011; 23:2086-96. [PMID: 21864676 DOI: 10.1016/j.cellsig.2011.08.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 07/30/2011] [Accepted: 08/01/2011] [Indexed: 01/07/2023]
Abstract
Endothelial cell (EC) barrier dysfunction induced by inflammatory agonists is a frequent pathophysiologic event in multiple diseases. The platelet-derived phospholipid sphingosine-1 phosphate (S1P) reverses this dysfunction by potently enhancing the EC barrier through a process involving Rac GTPase-dependent cortical actin rearrangement as an integral step. In this study we explored the role of the ezrin, radixin, and moesin (ERM) family of actin-binding linker protein in modulating S1P-induced human pulmonary EC barrier enhancement. S1P induces ERM translocation to the EC periphery and promotes ERM phosphorylation on a critical threonine residue (Ezrin-567, Radixin-564, Moesin-558). This phosphorylation is dependent on activation of PKC isoforms and Rac1. The majority of ERM phosphorylation on these critical threonine residues after S1P occurs in moesin and ezrin. Baseline radixin phosphorylation is higher than in the other two ERM proteins but does not increase after S1P. S1P-induced moesin and ezrin threonine phosphorylation is not mediated by the barrier enhancing receptor S1PR1 because siRNA downregulation of S1PR1 fails to inhibit these phosphorylation events, while stimulation of EC with the S1PR1-specific agonist SEW2871 fails to induce these phosphorylation events. Silencing of either all ERM proteins or radixin alone (but not moesin alone) reduced S1P-induced Rac1 activation and phosphorylation of the downstream Rac1 effector PAK1. Radixin siRNA alone, or combined siRNA for all three ERM proteins, dramatically attenuates S1P-induced EC barrier enhancement (measured by transendothelial electrical resistance (TER), peripheral accumulation of di-phospho-MLC, and cortical cytoskeletal rearrangement. In contrast, moesin depletion has the opposite effects on these parameters. Ezrin silencing partially attenuates S1P-induced EC barrier enhancement and cytoskeletal changes. Thus, despite structural similarities and reported functional redundancy, the ERM proteins differentially modulate S1P-induced alterations in lung EC cytoskeleton and permeability. These results suggest that ERM activation is an important regulatory event in EC barrier responses to S1P.
Collapse
Affiliation(s)
- Djanybek M Adyshev
- Institute for Personalized Respiratory Medicine, Department of Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|