1
|
Hwang IY, Kim JS, Harrison KA, Park C, Shi CS, Kehrl JH. Chemokine-mediated F-actin dynamics, polarity, and migration in B lymphocytes depend on WNK1 signaling. Sci Signal 2024; 17:eade1119. [PMID: 39190707 PMCID: PMC11542683 DOI: 10.1126/scisignal.ade1119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 05/01/2023] [Accepted: 08/02/2024] [Indexed: 08/29/2024]
Abstract
Ligand-engaged chemokine receptors trigger nucleotide exchange in heterotrimeric Gαi proteins, which stimulates cytoskeletal reorganization and cell polarity changes. To better understand the signaling events responsible for these cellular changes, we focused on early changes in F-actin dynamics after engagement of the chemokine receptor CXCR5 in murine splenic B cells. Within 10 seconds of exposure to the CXCR5 ligand CXCL13, three-dimensional lamellar-like pseudopods and F-actin-rich ridges appeared. The transient F-actin increase depended on Gαi2/3 signaling, the PI3K/AKT pathway, ERK activation, phospholipase C activity, and Rac1/2 activation mediated by Dock2 (dedicator of cytokinesis 2). Immunoblot analyses identified the kinase WNK1 (with no lysine kinase 1) as a potential early AKT effector. Treating B cells with specific WNK inhibitors disrupted F-actin dynamics and impaired B cell polarity, motility, and chemotaxis. These changes were mimicked in a murine B cell line by CRISPR-Cas9 gene editing of Wnk1, which also suggested that WNK1 contributed to B cell proliferation. Administration of a single dose of a WNK inhibitor transiently reduced B cell motility and polarity in the lymph nodes of live mice. These results indicate that WNK1 signaling maintains B cell responsiveness to CXCL13 and suggest that pharmacological inhibition of WNK1, which is involved in cancer progression and blood pressure regulation, may affect humoral immunity.
Collapse
Affiliation(s)
- Il-Young Hwang
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Ji Sung Kim
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Kathleen A. Harrison
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Chung Park
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Chong Shan Shi
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - John H. Kehrl
- B-cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
2
|
Abstract
T cell activation is initiated by the recognition of specific antigenic peptides and subsequently accomplished by complex signaling cascades. These aspects have been extensively studied for decades as pivotal factors in the establishment of adaptive immunity. However, how receptors or signaling molecules are organized in the resting state prior to encountering antigens has received less attention. Recent advancements in super-resolution microscopy techniques have revealed topographically controlled pre-formed organization of key molecules involved in antigen recognition and signal transduction on microvillar projections of T cells before activation and substantial effort has been dedicated to characterizing the topological structure of resting T cells over the past decade. This review will summarize our current understanding of how key surface receptors are pre-organized on the T-cell plasma membrane and discuss the potential role of these receptors, which are preassembled prior to ligand binding in the early activation events of T cells.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Nano-Biomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science, Seoul, Republic of Korea
| |
Collapse
|
3
|
Chen J, Wang M, Zhang Y, Zhu F, Xu Y, Yi G, Zheng R, Wu B. Platelet extracellular vesicles: Darkness and light of autoimmune diseases. Int Rev Immunol 2023; 43:63-73. [PMID: 37350464 DOI: 10.1080/08830185.2023.2225551] [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: 01/11/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
Autoimmune diseases are characterized by a breakdown of immune tolerance, leading to inflammation and irreversible end-organ tissue damage. Platelet extracellular vesicles are cellular elements that are important in blood circulation and actively participate in inflammatory and immune responses through intercellular communication and interactions between inflammatory cells, immune cells, and their secreted factors. Therefore, platelet extracellular vesicles are the "accelerator" in the pathological process of autoimmune diseases; however, this robust set of functions of platelet extracellular vesicles has also prompted new advances in therapeutic strategies for autoimmune diseases. In this review, we update fundamental mechanisms based on platelet extracellular vesicles communication function in autoimmune diseases. We also focus on the potential role of platelet extracellular vesicles for the treatment of autoimmune diseases. Some recent studies have found that antiplatelet aggregation drugs, specific biological agents can reduce the release of platelet extracellular vesicles. Platelet extracellular vesicles can also serve as vehicles to deliver drugs to targeted cells. It seems that we can try to silence or inhibit microRNA carried by platelet extracellular vesicles transcription and regulate the target cells to treat autoimmune diseases as platelet extracellular vesicles can transfer microRNA to other cells to regulate immune-inflammatory responses. Hopefully, the information presented here will provide hope for patients with autoimmune diseases.
Collapse
Affiliation(s)
- Jingru Chen
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, P.R. China
| | - Miao Wang
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Ying Zhang
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Fenglin Zhu
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Yanqiu Xu
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Guoxiang Yi
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
| | - Runxiu Zheng
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, P.R. China
| | - Bin Wu
- Department of Rheumatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, P.R. China
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan, P.R. China
| |
Collapse
|
4
|
Toffali L, D'Ulivo B, Giagulli C, Montresor A, Zenaro E, Delledonne M, Rossato M, Iadarola B, Sbarbati A, Bernardi P, Angelini G, Rossi B, Lopez N, Linke WA, Unger A, Di Silvestre D, Benazzi L, De Palma A, Motta S, Constantin G, Mauri P, Laudanna C. An isoform of the giant protein titin is a master regulator of human T lymphocyte trafficking. Cell Rep 2023; 42:112516. [PMID: 37204926 DOI: 10.1016/j.celrep.2023.112516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/24/2023] [Accepted: 05/01/2023] [Indexed: 05/21/2023] Open
Abstract
Response to multiple microenvironmental cues and resilience to mechanical stress are essential features of trafficking leukocytes. Here, we describe unexpected role of titin (TTN), the largest protein encoded by the human genome, in the regulation of mechanisms of lymphocyte trafficking. Human T and B lymphocytes express five TTN isoforms, exhibiting cell-specific expression, distinct localization to plasma membrane microdomains, and different distribution to cytosolic versus nuclear compartments. In T lymphocytes, the LTTN1 isoform governs the morphogenesis of plasma membrane microvilli independently of ERM protein phosphorylation status, thus allowing selectin-mediated capturing and rolling adhesions. Likewise, LTTN1 controls chemokine-triggered integrin activation. Accordingly, LTTN1 mediates rho and rap small GTPases activation, but not actin polymerization. In contrast, chemotaxis is facilitated by LTTN1 degradation. Finally, LTTN1 controls resilience to passive cell deformation and ensures T lymphocyte survival in the blood stream. LTTN1 is, thus, a critical and versatile housekeeping regulator of T lymphocyte trafficking.
Collapse
Affiliation(s)
- Lara Toffali
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Beatrice D'Ulivo
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Cinzia Giagulli
- Department of Molecular and Translational Medicine, University of Brescia; 25123 Brescia, Lombardia, Italy
| | - Alessio Montresor
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy; The Center for Biomedical Computing (CBMC), University of Verona; 37134 Verona, Veneto, Italy
| | - Elena Zenaro
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Massimo Delledonne
- Department of Biotechnology, University of Verona; 37134 Verona, Veneto, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona; 37134 Verona, Veneto, Italy
| | - Barbara Iadarola
- Department of Biotechnology, University of Verona; 37134 Verona, Veneto, Italy
| | - Andrea Sbarbati
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona; 37134 Verona, Veneto, Italy
| | - Paolo Bernardi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona; 37134 Verona, Veneto, Italy
| | - Gabriele Angelini
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Barbara Rossi
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Nicola Lopez
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, and Heart Center, University Medicine; 37075 Göttingen, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Muenster, and Heart Center, University Medicine; 37075 Göttingen, Germany
| | - Dario Di Silvestre
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Louise Benazzi
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Antonella De Palma
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Sara Motta
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Gabriela Constantin
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy; The Center for Biomedical Computing (CBMC), University of Verona; 37134 Verona, Veneto, Italy
| | - Pierluigi Mauri
- Institute of Biomedical Technologies (ITB) CNR; 20090 Milan, Lombardia, Italy
| | - Carlo Laudanna
- Department of Medicine, Division of General Pathology, Laboratory of Cell Trafficking and Signal Transduction, University of Verona; 37134 Verona, Veneto, Italy; The Center for Biomedical Computing (CBMC), University of Verona; 37134 Verona, Veneto, Italy.
| |
Collapse
|
5
|
Molon B, Liboni C, Viola A. CD28 and chemokine receptors: Signalling amplifiers at the immunological synapse. Front Immunol 2022; 13:938004. [PMID: 35983040 PMCID: PMC9379342 DOI: 10.3389/fimmu.2022.938004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/08/2022] [Indexed: 01/14/2023] Open
Abstract
T cells are master regulators of the immune response tuning, among others, B cells, macrophages and NK cells. To exert their functions requiring high sensibility and specificity, T cells need to integrate different stimuli from the surrounding microenvironment. A finely tuned signalling compartmentalization orchestrated in dynamic platforms is an essential requirement for the proper and efficient response of these cells to distinct triggers. During years, several studies have depicted the pivotal role of the cytoskeleton and lipid microdomains in controlling signalling compartmentalization during T cell activation and functions. Here, we discuss mechanisms responsible for signalling amplification and compartmentalization in T cell activation, focusing on the role of CD28, chemokine receptors and the actin cytoskeleton. We also take into account the detrimental effect of mutations carried by distinct signalling proteins giving rise to syndromes characterized by defects in T cell functionality.
Collapse
Affiliation(s)
- Barbara Molon
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- *Correspondence: Barbara Molon,
| | - Cristina Liboni
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Antonella Viola
- Pediatric Research Institute “Città della Speranza”, Corso Stati Uniti, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
6
|
De Belly H, Paluch EK, Chalut KJ. Interplay between mechanics and signalling in regulating cell fate. Nat Rev Mol Cell Biol 2022; 23:465-480. [PMID: 35365816 DOI: 10.1038/s41580-022-00472-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/11/2022]
Abstract
Mechanical signalling affects multiple biological processes during development and in adult organisms, including cell fate transitions, cell migration, morphogenesis and immune responses. Here, we review recent insights into the mechanisms and functions of two main routes of mechanical signalling: outside-in mechanical signalling, such as mechanosensing of substrate properties or shear stresses; and mechanical signalling regulated by the physical properties of the cell surface itself. We discuss examples of how these two classes of mechanical signalling regulate stem cell function, as well as developmental processes in vivo. We also discuss how cell surface mechanics affects intracellular signalling and, in turn, how intracellular signalling controls cell surface mechanics, generating feedback into the regulation of mechanosensing. The cooperation between mechanosensing, intracellular signalling and cell surface mechanics has a profound impact on biological processes. We discuss here our understanding of how these three elements interact to regulate stem cell fate and development.
Collapse
Affiliation(s)
- Henry De Belly
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ewa K Paluch
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Kevin J Chalut
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Wellcome/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
7
|
Abstract
Ras homology (RHO) GTPases are signalling proteins that have crucial roles in triggering multiple immune functions. Through their interactions with a broad range of effectors and kinases, they regulate cytoskeletal dynamics, cell polarity and the trafficking and proliferation of immune cells. The activity and localization of RHO GTPases are highly controlled by classical families of regulators that share consensus motifs. In this Review, we describe the recent discovery of atypical modulators and partners of RHO GTPases, which bring an additional layer of regulation and plasticity to the control of RHO GTPase activities in the immune system. Furthermore, the development of large-scale genetic screening has now enabled researchers to identify dysregulation of RHO GTPase signalling pathways as a cause of many immune system-related diseases. We discuss the mutations that have been identified in RHO GTPases and their signalling circuits in patients with rare diseases. The discoveries of new RHO GTPase partners and genetic mutations in RHO GTPase signalling hubs have uncovered unsuspected layers of crosstalk with other signalling pathways and may provide novel therapeutic opportunities for patients affected by complex immune or broader syndromes.
Collapse
|
8
|
Jung Y, Wen L, Altman A, Ley K. CD45 pre-exclusion from the tips of T cell microvilli prior to antigen recognition. Nat Commun 2021; 12:3872. [PMID: 34162836 PMCID: PMC8222282 DOI: 10.1038/s41467-021-23792-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
The tyrosine phosphatase CD45 is a major gatekeeper for restraining T cell activation. Its exclusion from the immunological synapse (IS) is crucial for T cell receptor (TCR) signal transduction. Here, we use expansion super-resolution microscopy to reveal that CD45 is mostly pre-excluded from the tips of microvilli (MV) on primary T cells prior to antigen encounter. This pre-exclusion is diminished by depleting cholesterol or by engineering the transmembrane domain of CD45 to increase its membrane integration length, but is independent of the CD45 extracellular domain. We further show that brief MV-mediated contacts can induce Ca2+ influx in mouse antigen-specific T cells engaged by antigen-pulsed antigen presenting cells (APC). We propose that the scarcity of CD45 phosphatase activity at the tips of MV enables or facilitates TCR triggering from brief T cell-APC contacts before formation of a stable IS, and that these MV-mediated contacts represent the earliest step in the initiation of a T cell adaptive immune response.
Collapse
Affiliation(s)
- Yunmin Jung
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | - Lai Wen
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Amnon Altman
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Ghosh S, Di Bartolo V, Tubul L, Shimoni E, Kartvelishvily E, Dadosh T, Feigelson SW, Alon R, Alcover A, Haran G. ERM-Dependent Assembly of T Cell Receptor Signaling and Co-stimulatory Molecules on Microvilli prior to Activation. Cell Rep 2021; 30:3434-3447.e6. [PMID: 32160548 DOI: 10.1016/j.celrep.2020.02.069] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/16/2019] [Accepted: 02/18/2020] [Indexed: 01/25/2023] Open
Abstract
T cell surfaces are covered with microvilli, actin-rich and flexible protrusions. We use super-resolution microscopy to show that ≥90% of T cell receptor (TCR) complex molecules TCRαβ and TCRζ, as well as the co-receptor CD4 (cluster of differentiation 4) and the co-stimulatory molecule CD2, reside on microvilli of resting human T cells. Furthermore, TCR proximal signaling molecules involved in the initial stages of the immune response, including the protein tyrosine kinase Lck (lymphocyte-specific protein tyrosine kinase) and the key adaptor LAT (linker for activation of T cells), are also enriched on microvilli. Notably, phosphorylated proteins of the ERM (ezrin, radixin, and moesin) family colocalize with TCRαβ as well as with actin filaments, implying a role for one or more ERMs in linking the TCR complex to the actin cytoskeleton within microvilli. Our results establish microvilli as key signaling hubs, in which the TCR complex and its proximal signaling molecules and adaptors are preassembled prior to activation in an ERM-dependent manner, facilitating initial antigen sensing.
Collapse
Affiliation(s)
- Shirsendu Ghosh
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Liron Tubul
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eyal Shimoni
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elena Kartvelishvily
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Dadosh
- Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Andres Alcover
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France
| | - Gilad Haran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
10
|
Kwon W, Freeman SA. Phagocytosis by the Retinal Pigment Epithelium: Recognition, Resolution, Recycling. Front Immunol 2020; 11:604205. [PMID: 33281830 PMCID: PMC7691529 DOI: 10.3389/fimmu.2020.604205] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Tissue-resident phagocytes are responsible for the routine binding, engulfment, and resolution of their meals. Such populations of cells express appropriate surface receptors that are tailored to recognize the phagocytic targets of their niche and initiate the actin polymerization that drives internalization. Tissue-resident phagocytes also harbor enzymes and transporters along the endocytic pathway that orchestrate the resolution of ingested macromolecules from the phagolysosome. Solutes fluxed from the endocytic pathway and into the cytosol can then be reutilized by the phagocyte or exported for their use by neighboring cells. Such a fundamental metabolic coupling between resident phagocytes and the tissue in which they reside is well-emphasized in the case of retinal pigment epithelial (RPE) cells; specialized phagocytes that are responsible for the turnover of photoreceptor outer segments (POS). Photoreceptors are prone to photo-oxidative damage and their long-term health depends enormously on the disposal of aged portions of the outer segment. The phagocytosis of the POS by the RPE is the sole means of this turnover and clearance. RPE are themselves mitotically quiescent and therefore must resolve the ingested material to prevent their toxic accumulation in the lysosome that otherwise leads to retinal disorders. Here we describe the sequence of events underlying the healthy turnover of photoreceptors by the RPE with an emphasis on the signaling that ensures the phagocytosis of the distal POS and on the transport of solutes from the phagosome that supersedes its resolution. While other systems may utilize different receptors and transporters, the biophysical and metabolic manifestations of such events are expected to apply to all tissue-resident phagocytes that perform regular phagocytic programs.
Collapse
Affiliation(s)
- Whijin Kwon
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
12
|
Orbach R, Su X. Surfing on Membrane Waves: Microvilli, Curved Membranes, and Immune Signaling. Front Immunol 2020; 11:2187. [PMID: 33013920 PMCID: PMC7516127 DOI: 10.3389/fimmu.2020.02187] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/11/2020] [Indexed: 01/22/2023] Open
Abstract
Microvilli are finger-like membrane protrusions, supported by the actin cytoskeleton, and found on almost all cell types. A growing body of evidence suggests that the dynamic lymphocyte microvilli, with their highly curved membranes, play an important role in signal transduction leading to immune responses. Nevertheless, challenges in modulating local membrane curvature and monitoring the high dynamicity of microvilli hampered the investigation of the curvature-generation mechanism and its functional consequences in signaling. These technical barriers have been partially overcome by recent advancements in adapted super-resolution microscopy. Here, we review the up-to-date progress in understanding the mechanisms and functional consequences of microvillus formation in T cell signaling. We discuss how the deformation of local membranes could potentially affect the organization of signaling proteins and their biochemical activities. We propose that curved membranes, together with the underlying cytoskeleton, shape microvilli into a unique compartment that sense and process signals leading to lymphocyte activation.
Collapse
Affiliation(s)
- Ron Orbach
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
- Yale Cancer Center, Yale University, New Haven, CT, United States
| |
Collapse
|
13
|
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: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [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
|
14
|
Wurzer H, Hoffmann C, Al Absi A, Thomas C. Actin Cytoskeleton Straddling the Immunological Synapse between Cytotoxic Lymphocytes and Cancer Cells. Cells 2019; 8:cells8050463. [PMID: 31100864 PMCID: PMC6563383 DOI: 10.3390/cells8050463] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
The immune system is a fundamental part of the tumor microenvironment. In particular, cytotoxic lymphocytes, such as cytolytic T cells and natural killer cells, control tumor growth and disease progression by interacting and eliminating tumor cells. The actin cytoskeleton of cytotoxic lymphocytes engaged in an immunological synapse has received considerable research attention. It has been recognized as a central mediator of the formation and maturation of the immunological synapse, and its signaling and cytolytic activities. In comparison, fewer studies have explored the organization and function of actin filaments on the target cancer cell side of the immunological synapse. However, there is growing evidence that the actin cytoskeleton of cancer cells also undergoes extensive remodeling upon cytotoxic lymphocyte attack, and that such remodeling can alter physical and functional interactions at the immunological synapse. In this article, we review the current knowledge of actin organization and functions at both sides of the immunological synapse between cytotoxic lymphocytes and cancer cells, with particular focus on synapse formation, signaling and cytolytic activity, and immune evasion.
Collapse
Affiliation(s)
- Hannah Wurzer
- Cytoskeleton and Cancer Progression; Laboratory of Experimental Cancer Research, Department of Oncology 84 Val Fleuri, L-1526 Luxembourg City, Luxembourg.
- University of Luxembourg, Faculty of Science, Technology and Communication, 2 Avenue de l'Université, L-4365 Esch-sur-Alzette, Luxembourg.
| | - Céline Hoffmann
- Cytoskeleton and Cancer Progression; Laboratory of Experimental Cancer Research, Department of Oncology 84 Val Fleuri, L-1526 Luxembourg City, Luxembourg.
| | - Antoun Al Absi
- Cytoskeleton and Cancer Progression; Laboratory of Experimental Cancer Research, Department of Oncology 84 Val Fleuri, L-1526 Luxembourg City, Luxembourg.
- University of Strasbourg, 67081 Strasbourg, France.
| | - Clément Thomas
- Cytoskeleton and Cancer Progression; Laboratory of Experimental Cancer Research, Department of Oncology 84 Val Fleuri, L-1526 Luxembourg City, Luxembourg.
| |
Collapse
|
15
|
Ivetic A, Hoskins Green HL, Hart SJ. L-selectin: A Major Regulator of Leukocyte Adhesion, Migration and Signaling. Front Immunol 2019; 10:1068. [PMID: 31139190 PMCID: PMC6527602 DOI: 10.3389/fimmu.2019.01068] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
L-selectin (CD62L) is a type-I transmembrane glycoprotein and cell adhesion molecule that is expressed on most circulating leukocytes. Since its identification in 1983, L-selectin has been extensively characterized as a tethering/rolling receptor. There is now mounting evidence in the literature to suggest that L-selectin plays a role in regulating monocyte protrusion during transendothelial migration (TEM). The N-terminal calcium-dependent (C-type) lectin domain of L-selectin interacts with numerous glycans, including sialyl Lewis X (sLex) for tethering/rolling and proteoglycans for TEM. Although the signals downstream of L-selectin-dependent adhesion are poorly understood, they will invariably involve the short 17 amino acid cytoplasmic tail. In this review we will detail the expression of L-selectin in different immune cell subsets, and its influence on cell behavior. We will list some of the diverse glycans known to support L-selectin-dependent adhesion, within luminal and abluminal regions of the vessel wall. We will describe how each domain within L-selectin contributes to adhesion, migration and signal transduction. A significant focus on the L-selectin cytoplasmic tail and its proposed contribution to signaling via the ezrin-radixin-moesin (ERM) family of proteins will be outlined. Finally, we will discuss how ectodomain shedding of L-selectin during monocyte TEM is essential for the establishment of front-back cell polarity, bestowing emigrated cells the capacity to chemotax toward sites of damage.
Collapse
Affiliation(s)
- Aleksandar Ivetic
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Hannah Louise Hoskins Green
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| | - Samuel James Hart
- King's College London, School of Cardiovascular Medicine and Sciences, BHF Center of Research Excellence, London, United Kingdom
| |
Collapse
|
16
|
Yin LM, Duan TT, Ulloa L, Yang YQ. Ezrin Orchestrates Signal Transduction in Airway Cells. Rev Physiol Biochem Pharmacol 2019; 174:1-23. [PMID: 28702704 DOI: 10.1007/112_2017_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Ezrin is a critical structural protein that organizes receptor complexes and orchestrates their signal transduction. In this study, we review the ezrin-meditated regulation of critical receptor complexes, including the epidermal growth factor receptor (EGFR), CD44, vascular cell adhesion molecule (VCAM), and the deleted in colorectal cancer (DCC) receptor. We also analyze the ezrin-meditated regulation of critical pathways associated with asthma, such as the RhoA, Rho-associated protein kinase (ROCK), and protein kinase A (cAMP/PKA) pathways. Mounting evidence suggests that ezrin plays a role in controlling airway cell function and potentially contributes to respiratory diseases. Ezrin can participate in asthma pathogenesis by affecting bronchial epithelium repair, T lymphocyte regulation, and the contraction of the airway smooth muscle cells. These studies provide new insights for the design of novel therapeutic strategies for asthma treatment.
Collapse
Affiliation(s)
- Lei-Miao Yin
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and Meridian, Yue Yang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Ting-Ting Duan
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and Meridian, Yue Yang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Luis Ulloa
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and Meridian, Yue Yang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China. .,Department of Surgery, Center of Immunology and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, Newark, NJ, 07101, USA.
| | - Yong-Qing Yang
- Laboratory of Molecular Biology, Shanghai Research Institute of Acupuncture and Meridian, Yue Yang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| |
Collapse
|
17
|
Abstract
Platelets play important roles in blood clotting, hemostasis and wound repair, while more and more research show that platelets also have significant contributions in the process of inflammation. Rheumatoid arthritis is a chronic systemic inflammatory autoimmune disease. Platelet microparticles, which are membrane vesicles shed by activated platelets, are reported to amplify inflammation in Rheumatoid arthritis. Here we show that either platelet-specific deletion of Rac1 (Rac1-/-) or Rac1-specific inhibitor NSC23766 dramatically inhibit platelet-derived microparticles formation. As we all know, collagen-induced arthritis (CIA) mouse model is the most common autoimmune model of rheumatoid arthritis. Interestingly, NSC23766 alleviated the process of collagen-induced arthritis of DBA mice in vivo, including the reduced hind paw thickness and ankle stiffness, the reduction of arthritic scores and incidence of arthritis. Our work also found that NSC23766-treated CIA mouse spleen is less swollen and contains less enlarged white pulp than PBS control. The histological analysis shows that NSC23766-treated but not solvent control improve the cartilage erosion symptom in the joint of CIA mouse. Interestingly, platelet microparticles in the peripheral blood of NSC23766-treated CIA mice were decreased significantly compared with PBS-treated CIA mice. In conclusion, our work demonstrated that Rac1 inhibition alleviates collagen-induced arthritis through the decrease of platelet microparticles' release. In short, Rac1 aggravate the rheumatoid arthritis deterioration through the regulation of platelet microparticles formation.
Collapse
Affiliation(s)
- Xue Chen
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai, China
| |
Collapse
|
18
|
Megrelis L, El Ghoul E, Moalli F, Versapuech M, Cassim S, Ruef N, Stein JV, Mangeney M, Delon J. Fam65b Phosphorylation Relieves Tonic RhoA Inhibition During T Cell Migration. Front Immunol 2018; 9:2001. [PMID: 30254631 PMCID: PMC6141708 DOI: 10.3389/fimmu.2018.02001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/14/2018] [Indexed: 11/13/2022] Open
Abstract
We previously identified Fam65b as an atypical inhibitor of the small G protein RhoA. Using a conditional model of a Fam65b-deficient mouse, we first show that Fam65b restricts spontaneous RhoA activation in resting T lymphocytes and regulates intranodal T cell migration in vivo. We next aimed at understanding, at the molecular level, how the brake that Fam65b exerts on RhoA can be relieved upon signaling to allow RhoA activation. Here, we show that chemokine stimulation phosphorylates Fam65b in T lymphocytes. This post-translational modification decreases the affinity of Fam65b for RhoA and favors Fam65b shuttling from the plasma membrane to the cytosol. Functionally, we show that the degree of Fam65b phosphorylation controls some cytoskeletal alterations downstream active RhoA such as actin polymerization, as well as T cell migration in vitro. Altogether, our results show that Fam65b expression and phosphorylation can finely tune the amount of active RhoA in order to favor optimal T lymphocyte motility.
Collapse
Affiliation(s)
- Laura Megrelis
- Infection, Immunity, Inflammation, Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Elyas El Ghoul
- Infection, Immunity, Inflammation, Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Federica Moalli
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Margaux Versapuech
- Infection, Immunity, Inflammation, Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Shamir Cassim
- Infection, Immunity, Inflammation, Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nora Ruef
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Marianne Mangeney
- Infection, Immunity, Inflammation, Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jérôme Delon
- Infection, Immunity, Inflammation, Inserm, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
19
|
T cell microvilli constitute immunological synaptosomes that carry messages to antigen-presenting cells. Nat Commun 2018; 9:3630. [PMID: 30194420 PMCID: PMC6128830 DOI: 10.1038/s41467-018-06090-8] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/17/2018] [Indexed: 12/22/2022] Open
Abstract
Microvilli on T cells have been proposed to survey surfaces of antigen-presenting cells (APC) or facilitate adhesion under flow; however, whether they serve essential functions during T cell activation remains unclear. Here we show that antigen-specific T cells deposit membrane particles derived from microvilli onto the surface of cognate antigen-bearing APCs. Microvilli carry T cell receptors (TCR) at all stages of T cell activation and are released as large TCR-enriched, T cell microvilli particles (TMP) in a process of trogocytosis. These microvilli exclusively contain protein arrestin-domain-containing protein 1, which is directly involved in membrane budding and, in combination with vacuolar protein-sorting-associated protein 4, transforms large TMPs into smaller, exosome-sized TMPs. Notably, TMPs from CD4+ T cells are enriched with LFA-2/CD2 and various cytokines involved in activating dendritic cells. Collectively, these results demonstrate that T cell microvilli constitute “immunological synaptosomes” that carry T cell messages to APCs. Microvilli can participate in adhesion or migration of T cells, but whether they are involved in function regulation is unclear. Here the authors show that T cell microvilli form budding vesicles containing T cell signalling components for deposition onto antigen presenting cells (APC) and modulation of APC functions.
Collapse
|
20
|
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: 81] [Impact Index Per Article: 11.6] [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
|
21
|
Kurdi AT, Bassil R, Olah M, Wu C, Xiao S, Taga M, Frangieh M, Buttrick T, Orent W, Bradshaw EM, Khoury SJ, Elyaman W. Tiam1/Rac1 complex controls Il17a transcription and autoimmunity. Nat Commun 2016; 7:13048. [PMID: 27725632 PMCID: PMC5062600 DOI: 10.1038/ncomms13048] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/30/2016] [Indexed: 12/14/2022] Open
Abstract
RORγt is a master transcription factor of Th17 cells and considered as a promising drug target for the treatment of autoimmune diseases. Here, we show the guanine nucleotide exchange factor, Tiam1, and its cognate Rho-family G protein, Rac1, regulate interleukin (IL)17A transcription and autoimmunity. Whereas Tiam1 genetic deficiency weakens IL-17A expression partially and inhibits the development of experimental autoimmune encephalomyelitis (EAE), deletion of Rac1 in T cells exhibits more robust effects on Th17 cells and EAE. We demonstrate Tiam1 and Rac1 form a complex with RORγt in the nuclear compartment of Th17 cells, and together bind and activate the Il17 promoter. The clinical relevance of these findings is emphasized by pharmacological targeting of Rac1 that suppresses both murine and human Th17 cells as well as EAE. Thus, our findings highlight a regulatory pathway of Tiam1/Rac1 in Th17 cells and suggest that it may be a therapeutic target in multiple sclerosis. Tiam1 is a guanine nucleotide exchange factor for the Rho-family GTPase Rac1. Here, the authors show that nuclear Tiam1 and Rac1 bind to RORγt on the IL-17 promoter, activating its transcription, and that inhibiting Tiam1/Rac1 is beneficial in a mouse model of autoimmunity.
Collapse
Affiliation(s)
- Ahmed T Kurdi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ribal Bassil
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Marta Olah
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Chuan Wu
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sheng Xiao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mariko Taga
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Michael Frangieh
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Thomas Buttrick
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - William Orent
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Elizabeth M Bradshaw
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| | - Samia J Khoury
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Abu Haidar Neuroscience Institute, American University of Beirut Medical Center, Riad El Solh, Beirut 1107 2020, Lebanon
| | - Wassim Elyaman
- Program in Translational NeuroPsychiatric Genomics, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Broad Institute at Harvard University and MIT, Boston, Massachusetts 02115, USA
| |
Collapse
|
22
|
Jung Y, Riven I, Feigelson SW, Kartvelishvily E, Tohya K, Miyasaka M, Alon R, Haran G. Three-dimensional localization of T-cell receptors in relation to microvilli using a combination of superresolution microscopies. Proc Natl Acad Sci U S A 2016; 113:E5916-E5924. [PMID: 27647916 PMCID: PMC5056101 DOI: 10.1073/pnas.1605399113] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leukocyte microvilli are flexible projections enriched with adhesion molecules. The role of these cellular projections in the ability of T cells to probe antigen-presenting cells has been elusive. In this study, we probe the spatial relation of microvilli and T-cell receptors (TCRs), the major molecules responsible for antigen recognition on the T-cell membrane. To this end, an effective and robust methodology for mapping membrane protein distribution in relation to the 3D surface structure of cells is introduced, based on two complementary superresolution microscopies. Strikingly, TCRs are found to be highly localized on microvilli, in both peripheral blood human T cells and differentiated effector T cells, and are barely found on the cell body. This is a decisive demonstration that different types of T cells universally localize their TCRs to microvilli, immediately pointing to these surface projections as effective sensors for antigenic moieties. This finding also suggests how previously reported membrane clusters might form, with microvilli serving as anchors for specific T-cell surface molecules.
Collapse
Affiliation(s)
- Yunmin Jung
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Inbal Riven
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sara W Feigelson
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Kazuo Tohya
- Department of Anatomy, Kansai University of Health Sciences, Kumatori, Osaka 590-0482, Japan
| | - Masayuki Miyasaka
- Institute for Academic Initiatives, Osaka University, Suita, Osaka 565-0871, Japan; MediCity Research Laboratory, University of Turku, FI-20521, Turku, Finland
| | - Ronen Alon
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Gilad Haran
- Department of Chemical Physics, Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
23
|
Abstract
Pharmacogenetic studies have been performed for almost all classes of drugs that have been used in IBD but very few have generated consistent findings or have been replicated. The genetic test that has been approved for clinical practice is TPMT testing prior to starting treatment with thiopurine drugs. Research in IBD pharmacogenetics has focused on prediction of drug efficacy and toxicity by identifying polymorphisms in the genes encoding enzymes that are involved in metabolic pathways. Recent research has mainly focused on therapeutic agents such as azathioprine, methotrexate, aminosalicylates, corticosteroids, infliximab and adalimumab. Future pharmaceutical trials should include pharmacogenetic research to test appropriate candidate genes in a prospective manner and correlate genetic associations with trial outcomes and relevant functional data.
Collapse
|
24
|
Tu Z, Wang Q, Cui T, Wang J, Ran H, Bao H, Lu J, Wang B, Lydon JP, DeMayo F, Zhang S, Kong S, Wu X, Wang H. Uterine RAC1 via Pak1-ERM signaling directs normal luminal epithelial integrity conducive to on-time embryo implantation in mice. Cell Death Differ 2015; 23:169-81. [PMID: 26184908 DOI: 10.1038/cdd.2015.98] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 06/05/2015] [Accepted: 06/12/2015] [Indexed: 12/18/2022] Open
Abstract
Successful embryo implantation requires functional luminal epithelia to establish uterine receptivity and blastocyst-uterine adhesion. During the configuration of uterine receptivity from prereceptive phase, the luminal epithelium undergoes dynamic membrane reorganization and depolarization. This timely regulated epithelial membrane maturation and precisely maintained epithelial integrity are critical for embryo implantation in both humans and mice. However, it remained largely unexplored with respect to potential signaling cascades governing this functional epithelial transformation prior to implantation. Using multiple genetic and cellular approaches combined with uterine conditional Rac1 deletion mouse model, we demonstrated herein that Rac1, a small GTPase, is spatiotemporally expressed in the periimplantation uterus, and uterine depletion of Rac1 induces premature decrease of epithelial apical-basal polarity and defective junction remodeling, leading to disrupted uterine receptivity and implantation failure. Further investigations identified Pak1-ERM as a downstream signaling cascade upon Rac1 activation in the luminal epithelium necessary for uterine receptivity. In addition, we also demonstrated that Rac1 via P38 MAPK signaling ensures timely epithelial apoptotic death at postimplantation. Besides uncovering a potentially important molecule machinery governing uterine luminal integrity for embryo implantation, our finding has high clinical relevance, because Rac1 is essential for normal endometrial functions in women.
Collapse
Affiliation(s)
- Z Tu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China.,University of the Chinese Academy of Sciences, Beijing 100039, PR China
| | - Q Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - T Cui
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China.,University of the Chinese Academy of Sciences, Beijing 100039, PR China
| | - J Wang
- Department of Pharmacology, Zhejiang University, Hangzhou 310058, PR China
| | - H Ran
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100039, PR China
| | - H Bao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100039, PR China
| | - J Lu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - B Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - J P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - F DeMayo
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - S Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - S Kong
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - X Wu
- Department of Pharmacology, Zhejiang University, Hangzhou 310058, PR China
| | - H Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| |
Collapse
|
25
|
Akiyama T, Niyonsaba F, Kiatsurayanon C, Nguyen TT, Ushio H, Fujimura T, Ueno T, Okumura K, Ogawa H, Ikeda S. The human cathelicidin LL-37 host defense peptide upregulates tight junction-related proteins and increases human epidermal keratinocyte barrier function. J Innate Immun 2014; 6:739-53. [PMID: 24862212 DOI: 10.1159/000362789] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 04/10/2014] [Indexed: 12/30/2022] Open
Abstract
Both psoriasis and atopic dermatitis (AD) are not only associated with an impaired stratum corneum barrier, but also with abnormal expression of the tight junction (TJ) proteins. Because host defense peptides, including LL-37, are overexpressed in lesional psoriatic skin but are downregulated in lesional AD skin, we hypothesized that LL-37 might regulate the TJ function in keratinocytes. We demonstrated that LL-37 selectively increased the expression of several claudins and occludin, and enhanced their membrane distribution. Furthermore, LL-37 elevated the transepithelial electrical resistance while reducing the paracellular permeability of keratinocyte layers, and this activity was weakened by the claudin inhibitor ochratoxin A. A characterization of the molecular mechanism underlying the regulation of the TJ barrier by LL-37 revealed that LL-37 induced the activation of the Rac1, atypical PKC, glycogen synthase kinase-3 and PI3K pathways, and the specific inhibition of these pathways reversed the LL-37-mediated regulation of TJ function. In addition, LL-37 enhanced the expression of differentiation markers under the control of ochratoxin A, suggesting an association between LL-37-induced TJ function and keratinocyte differentiation. These data provide novel evidence that, in addition to its antimicrobial and other immunoregulatory functions, LL-37 contributes to cutaneous immunity by strengthening the skin's barrier function.
Collapse
Affiliation(s)
- Toshihiro Akiyama
- Department of Dermatology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ren L, Khanna C. Role of ezrin in osteosarcoma metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:181-201. [PMID: 24924175 DOI: 10.1007/978-3-319-04843-7_10] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The cause of death for the vast majority of cancer patients is the development of metastases at sites distant from that of the primary tumor. For most pediatric sarcoma patients such as those with osteosarcoma (OS), despite successful management of the primary tumor through multimodality approaches, the development of metastases, commonly to the lungs, is the cause of death. Significant improvements in long-term outcome for these patients have not been seen in more than 30 years. Furthermore, the long-term outcome for patients who present with metastatic disease is grave [1-5]. New treatment options are needed.Opportunities to improve outcomes for patients who present with metastases and those at-risk for progression and metastasis require an improved understanding of cancer progression and metastasis. With this goal in mind we and others have identified ezrin as a metastasis-associated protein that associated with OS and other cancers. Ezrin is the prototypical ERM (Ezrin/Radixin/Moesin) protein family member. ERMs function as linker proteins connecting the actin cytoskeleton and the plasma membrane. Since our initial identification of ezrin in pediatric sarcoma, an increasing understanding the role of ezrin in metastasis has emerged. Briefly, ezrin appears to allow metastatic cells to overcome a number of stresses experienced during the metastatic cascade, most notably the stress experienced as cells interact with the microenvironment of the secondary site. Cells must rapidly adapt to this environment in order to survive. Evidence now suggests a connection between ezrin expression and a variety of mechanisms linked to this important cellular adaptation including the ability of metastatic cells to initiate the translation of new proteins and to allow the efficient generation of ATP through a variety of sources. This understanding of the role of ezrin in the biology of metastasis is now sufficient to consider ezrin as an important therapeutic target in osteosarcoma patients. This chapter reviews our understanding of ezrin and the related ERM proteins in normal tissues and physiology, summarizes the expression of ezrin in human cancers and associations with clinical parameters of disease progression, reviews reports that detail a biological understanding of ezrin's role in metastatic progression, and concludes with a rationale that may be considered to target ezrin and ezrin biology in osteosarcoma.
Collapse
Affiliation(s)
- Ling Ren
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Dr., Rm 2144, Bethesda, MD, 20892, USA,
| | | |
Collapse
|
27
|
Abrogation of chronic rejection in rat model system involves modulation of the mTORC1 and mTORC2 pathways. Transplantation 2013; 96:782-90. [PMID: 23985719 DOI: 10.1097/tp.0b013e3182a2034f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Current immunosuppressive regimens fail to avert chronic rejection (CR) of transplanted organs; however, selective targeting of actin-cytoskeletal regulators decreases T-cell motility and abrogates CR in rat model system. Administration of mutated class I major histocompatibility complex molecules or selective targeting of the RhoA pathway, which controls T-cell cytoskeletal activity, using Y27632 (a selective Rock1 inhibitor) resulted in reduced T-cell infiltration and abrogation of CR as judged from the neointimal index (13.9±19.7 vs. 45±37.5; P<0.001) and the number of affected vessels (30% vs. 60%; P<0.01). Here, we examined the role of mammalian target of rapamycin (mTOR) pathway in inhibition of CR. METHODS A mutated class I major histocompatibility complex molecule that eliminates CR was delivered into ACI recipients of Wistar-Furth hearts at the time of transplantation with subtherapeutic cyclosporine (10 mg/kg on days 0-2). Controls included untreated and cyclosporine A-treated (10 mg/kg on days 0-2) heart allograft recipients. RESULTS Western blotting and immunostaining showed that rat heart allografts with abolished CR exhibited down-regulation of the RAPA-sensitive mTORC1 components such as mTOR and Raptor and down-regulation of the RAPA-insensitive mTORC2 elements Rictor and Sin1. The mTOR regulator Deptor and its downstream target Rac1 were also inhibited. CONCLUSIONS Abrogation of CR in rat model system involves modulation of two mTOR pathways: a RAPA-sensitive mTORC1 pathway regulating cellular proliferation and a RAPA-insensitive mTORC2 pathway regulating T-cell motility. Selective targeting of T-cell actin cytoskeletal pathways shows potential for pathway-targeted immunosuppression therapies.
Collapse
|
28
|
Srivastava N, Sudan R, Kerr WG. Role of inositol poly-phosphatases and their targets in T cell biology. Front Immunol 2013; 4:288. [PMID: 24069021 PMCID: PMC3779868 DOI: 10.3389/fimmu.2013.00288] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Accepted: 09/03/2013] [Indexed: 11/13/2022] Open
Abstract
T lymphocytes play a critical role in host defense in all anatomical sites including mucosal surfaces. This not only includes the effector arm of the immune system, but also regulation of immune responses in order to prevent autoimmunity. Genetic targeting of PI3K isoforms suggests that generation of PI(3,4,5)P3 by PI3K plays a critical role in promoting effector T cell responses. Consequently, the 5'- and 3'-inositol poly-phosphatases SHIP1, SHIP2, and phosphatase and tensin homolog capable of targeting PI(3,4,5)P3 are potential genetic determinants of T cell effector functions in vivo. In addition, the 5'-inositol poly-phosphatases SHIP1 and 2 can shunt PI(3,4,5)P3 to the rare but potent signaling phosphoinositide species PI(3,4)P2 and thus these SHIP1/2, and the INPP4A/B enzymes that deplete PI(3,4)P2 may have precise roles in T cell biology to amplify or inhibit effectors of PI3K signaling that are selectively recruited to and activated by PI(3,4)P2. Here we summarize recent genetic and chemical evidence that indicates the inositol poly-phosphatases have important roles in both the effector and regulatory functions of the T cell compartment. In addition, we will discuss future genetic studies that might be undertaken to further elaborate the role of these enzymes in T cell biology as well as potential pharmaceutical manipulation of these enzymes for therapeutic purposes in disease settings where T cell function is a key in vivo target.
Collapse
Affiliation(s)
- Neetu Srivastava
- Department of Microbiology and Immunology, SUNY Upstate Medical University , Syracuse, NY , USA
| | | | | |
Collapse
|
29
|
Adada M, Canals D, Hannun YA, Obeid LM. Sphingolipid regulation of ezrin, radixin, and moesin proteins family: implications for cell dynamics. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:727-37. [PMID: 23850862 DOI: 10.1016/j.bbalip.2013.07.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 06/30/2013] [Accepted: 07/02/2013] [Indexed: 12/13/2022]
Abstract
A key but poorly studied domain of sphingolipid functions encompasses endocytosis, exocytosis, cellular trafficking, and cell movement. Recently, the ezrin, radixin and moesin (ERM) family of proteins emerged as novel potent targets regulated by sphingolipids. ERMs are structural proteins linking the actin cytoskeleton to the plasma membrane, also forming a scaffold for signaling pathways that are used for cell proliferation, migration and invasion, and cell division. Opposing functions of the bioactive sphingolipid ceramide and sphingosine-1-phosphate (S1P), contribute to ERM regulation. S1P robustly activates whereas ceramide potently deactivates ERM via phosphorylation/dephosphorylation, respectively. This recent dimension of cytoskeletal regulation by sphingolipids opens up new avenues to target cell dynamics, and provides further understanding of some of the unexplained biological effects mediated by sphingolipids. In addition, these studies are providing novel inroads into defining basic mechanisms of regulation and action of bioactive sphingolipids. This review describes the current understanding of sphingolipid regulation of the cytoskeleton, it also describes the biologies in which ERM proteins have been involved, and finally how these two large fields have started to converge. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Mohamad Adada
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel Canals
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yusuf A Hannun
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- The Department of Medicine, Stony Brook University, Stony Brook, NY 11794, USA; The Northport VA Medical Center, Northport, NY 11768, USA.
| |
Collapse
|
30
|
Chaturvedi S, Yuen DA, Bajwa A, Huang YW, Sokollik C, Huang L, Lam GY, Tole S, Liu GY, Pan J, Chan L, Sokolskyy Y, Puthia M, Godaly G, John R, Wang C, Lee WL, Brumell JH, Okusa MD, Robinson LA. Slit2 prevents neutrophil recruitment and renal ischemia-reperfusion injury. J Am Soc Nephrol 2013; 24:1274-87. [PMID: 23766538 DOI: 10.1681/asn.2012090890] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Neutrophils recruited to the postischemic kidney contribute to the pathogenesis of ischemia-reperfusion injury (IRI), which is the most common cause of renal failure among hospitalized patients. The Slit family of secreted proteins inhibits chemotaxis of leukocytes by preventing activation of Rho-family GTPases, suggesting that members of this family might modulate the recruitment of neutrophils and the resulting IRI. Here, in static and microfluidic shear assays, Slit2 inhibited multiple steps required for the infiltration of neutrophils into tissue. Specifically, Slit2 blocked the capture and firm adhesion of human neutrophils to inflamed vascular endothelial barriers as well as their subsequent transmigration. To examine whether these observations were relevant to renal IRI, we administered Slit2 to mice before bilateral clamping of the renal pedicles. Assessed at 18 hours after reperfusion, Slit2 significantly inhibited renal tubular necrosis, neutrophil and macrophage infiltration, and rise in plasma creatinine. In vitro, Slit2 did not impair the protective functions of neutrophils, including phagocytosis and superoxide production, and did not inhibit neutrophils from killing the extracellular pathogen Staphylococcus aureus. In vivo, administration of Slit2 did not attenuate neutrophil recruitment or bacterial clearance in mice with ascending Escherichia coli urinary tract infections and did not increase the bacterial load in the livers of mice infected with the intracellular pathogen Listeria monocytogenes. Collectively, these results suggest that Slit2 may hold promise as a strategy to combat renal IRI without compromising the protective innate immune response.
Collapse
Affiliation(s)
- Swasti Chaturvedi
- Division of Nephrology, Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Dehapiot B, Halet G. Ran GTPase promotes oocyte polarization by regulating ERM (Ezrin/Radixin/Moesin) inactivation. Cell Cycle 2013; 12:1672-8. [PMID: 23656777 DOI: 10.4161/cc.24901] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Asymmetric meiotic divisions in mammalian oocytes are driven by the eccentric positioning of the spindle, along with a dramatic reorganization of the overlying cortex, including a loss of microvilli and formation of a thick actin cap. Actin polarization relies on a Ran-GTP gradient centered on metaphase chromosomes; however, the downstream signaling cascade is not completely understood. In a recent study, we have shown that Ran promotes actin cap formation via the polarized activation of Cdc42. The related GTPase Rac is also activated in a polarized fashion in the oocyte cortex and co-localizes with active Cdc42. In other cells, microvilli collapse can be triggered by inactivation of the ERM (Ezrin/Radixin/Moesin) family of actin-membrane crosslinkers under the control of Rac. Accordingly, we show here that Ran-GTP promotes a substantial loss of phosphorylated ERMs in the cortex overlying the spindle in mouse oocytes. However, this polarized phospho-ERM exclusion zone was unaffected by Rac or Cdc42 inhibition. Therefore, we suggest that Ran activates two distinct pathways to regulate actin cap formation and microvilli disassembly in the polarized cortex of mouse oocytes. The possibility of a crosstalk between Rho GTPase and ERM signaling and a role for ERM inactivation in promoting cortical actin dynamics are also discussed.
Collapse
Affiliation(s)
- Benoit Dehapiot
- Institut de Génétique et Développement de Rennes, CNRS UMR 6290; Rennes, France
| | | |
Collapse
|
32
|
Chen EJH, Shaffer MH, Williamson EK, Huang Y, Burkhardt JK. Ezrin and moesin are required for efficient T cell adhesion and homing to lymphoid organs. PLoS One 2013; 8:e52368. [PMID: 23468835 PMCID: PMC3585410 DOI: 10.1371/journal.pone.0052368] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 11/12/2012] [Indexed: 01/13/2023] Open
Abstract
T cell trafficking between the blood and lymphoid organs is a complex, multistep process that requires several highly dynamic and coordinated changes in cyto-architecture. Members of the ezrin, radixin and moesin (ERM) family of actin-binding proteins have been implicated in several aspects of this process, but studies have yielded conflicting results. Using mice with a conditional deletion of ezrin in CD4+ cells and moesin-specific siRNA, we generated T cells lacking ERM proteins, and investigated the effect on specific events required for T cell trafficking. ERM-deficient T cells migrated normally in multiple in vitro and in vivo assays, and could undergo efficient diapedesis in vitro. However, these cells were impaired in their ability to adhere to the β1 integrin ligand fibronectin, and to polarize appropriately in response to fibronectin and VCAM-1 binding. This defect was specific for β1 integrins, as adhesion and polarization in response to ICAM-1 were normal. In vivo, ERM-deficient T cells showed defects in homing to lymphoid organs. Taken together, these results show that ERM proteins are largely dispensable for T cell chemotaxis, but are important for β1 integrin function and homing to lymphoid organs.
Collapse
Affiliation(s)
- Emily J. H. Chen
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Meredith H. Shaffer
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Edward K. Williamson
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Yanping Huang
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Janis K. Burkhardt
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
33
|
ARF6-mediated endosomal transport of Telencephalin affects dendritic filopodia-to-spine maturation. EMBO J 2012; 31:3252-69. [PMID: 22781129 PMCID: PMC3411082 DOI: 10.1038/emboj.2012.182] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 06/11/2012] [Indexed: 01/21/2023] Open
Abstract
The GTPase Arf6 and its exchange factor EFA6A promote internalization of the Ig-like molecule Telencephalin in hippocampal neurons leading to the maturation of filopodia into dendritic spines, important for synapse formation. Dendritic filopodia are dynamic structures thought to be the precursors of spines during synapse development. Morphological maturation to spines is associated with the stabilization and strengthening of synapses, and can be altered in various neurological disorders. Telencephalin (TLN/intercellular adhesion molecule-5 (ICAM5)) localizes to dendritic filopodia, where it facilitates their formation/maintenance, thereby slowing spine morphogenesis. As spines are largely devoid of TLN, its exclusion from the filopodia surface appears to be required in this maturation process. Using HeLa cells and primary hippocampal neurons, we demonstrate that surface removal of TLN involves internalization events mediated by the small GTPase ADP-ribosylation factor 6 (ARF6), and its activator EFA6A. This endocytosis of TLN affects filopodia-to-spine transition, and requires Rac1-mediated dephosphorylation/release of actin-binding ERM proteins from TLN. At the somato-dendritic surface, TLN and EFA6A are confined to distinct, flotillin-positive membrane subdomains. The co-distribution of TLN with this lipid raft marker also persists during its endosomal targeting to CD63-positive late endosomes. This suggests a specific microenvironment facilitating ARF6-mediated mobilization of TLN that contributes to promotion of dendritic spine development.
Collapse
|
34
|
Ren L, Hong SH, Chen QR, Briggs J, Cassavaugh J, Srinivasan S, Lizardo MM, Mendoza A, Xia AY, Avadhani N, Khan J, Khanna C. Dysregulation of ezrin phosphorylation prevents metastasis and alters cellular metabolism in osteosarcoma. Cancer Res 2011; 72:1001-12. [PMID: 22147261 DOI: 10.1158/0008-5472.can-11-0210] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Ezrin links the plasma membrane to the actin cytoskeleton where it plays a pivotal role in the metastatic progression of several human cancers; however, the precise mechanistic basis for its role remains unknown. Here, we define transitions between active (phosphorylated open) and inactive (dephosphorylated closed) forms of Ezrin that occur during metastatic progression in osteosarcoma. In our evaluation of these conformations we expressed C-terminal mutant forms of Ezrin that are open (phosphomimetic T567D) or closed (phosphodeficient T567A) and compared their biologic characteristics to full-length wild-type Ezrin in osteosarcoma cells. Unexpectedly, cells expressing open, active Ezrin could form neither primary orthotopic tumors nor lung metastases. In contrast, cells expressing closed, inactive Ezrin were also deficient in metastasis but were unaffected in their capacity for primary tumor growth. By imaging single metastatic cells in the lung, we found that cells expressing either open or closed Ezrin displayed increased levels of apoptosis early after their arrival in the lung. Gene expression analysis suggested dysregulation of genes that are functionally linked to carbohydrate and amino acid metabolism. In particular, cells expressing closed, inactive Ezrin exhibited reduced lactate production and basal or ATP-dependent oxygen consumption. Collectively, our results suggest that dynamic regulation of Ezrin phosphorylation at amino acid T567 that controls structural transitions of this protein plays a pivotal role in tumor progression and metastasis, possibly in part by altering cellular metabolism.
Collapse
Affiliation(s)
- Ling Ren
- Tumor and Metastasis Biology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Harris SJ, Parry RV, Foster JG, Blunt MD, Wang A, Marelli-Berg F, Westwick J, Ward SG. Evidence That the Lipid Phosphatase SHIP-1 Regulates T Lymphocyte Morphology and Motility. THE JOURNAL OF IMMUNOLOGY 2011; 186:4936-45. [DOI: 10.4049/jimmunol.1002350] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
36
|
Parameswaran N, Matsui K, Gupta N. Conformational switching in ezrin regulates morphological and cytoskeletal changes required for B cell chemotaxis. THE JOURNAL OF IMMUNOLOGY 2011; 186:4088-97. [PMID: 21339367 DOI: 10.4049/jimmunol.1001139] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
B cell chemotaxis occurs in response to specific chemokine gradients and is critical for homeostasis and immune response. The molecular regulation of B cell membrane-actin interactions during migration is poorly understood. In this study, we report a role for ezrin, a member of the membrane-cytoskeleton cross-linking ezrin-radixin-moesin proteins, in the regulation of the earliest steps of B cell polarization and chemotaxis. We visualized chemokine-induced changes in murine B cell morphology using scanning electron microscopy and spatiotemporal dynamics of ezrin in B cells using epifluorescence and total internal reflection microscopy. Upon chemokine stimulation, ezrin is transiently dephosphorylated to assume an inactive conformation and localizes to the lamellipodia. B cells expressing a phosphomimetic conformationally active mutant of ezrin or those in which ezrin dephosphorylation was pharmacologically inhibited displayed impaired microvillar dynamics, morphological polarization, and chemotaxis. Our data suggest a 2-fold involvement of ezrin in B cell migration, whereby it first undergoes chemokine-induced dephosphorylation to facilitate membrane flexibility, followed by relocalization to the actin-rich lamellipodia for dynamic forward protrusion of the cells.
Collapse
Affiliation(s)
- Neetha Parameswaran
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | |
Collapse
|
37
|
Garcia GG, Miller RA. Age-related defects in the cytoskeleton signaling pathways of CD4 T cells. Ageing Res Rev 2011; 10:26-34. [PMID: 19941976 DOI: 10.1016/j.arr.2009.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 11/18/2009] [Accepted: 11/18/2009] [Indexed: 11/25/2022]
Abstract
It has been postulated that the cytoskeleton controls many aspects of T cell function, including activation, proliferation and apoptosis. Recent advances in our understanding of F-actin polymerization and the Ezrin-Radixin-Moesin (ERM) family of cytoskeleton signal proteins have provided new insights into immunological synapse formation during T cell activation. During aging there is a significant decline of T cell function largely attributable to declines in activation of CD4 T cells and defects in the formation of the immunological synapse. Here we discuss recent progress in the understanding of how aging alters F-actin and ERM proteins in mouse CD4 T cells, and the implications of these changes for the T cell activation process.
Collapse
|
38
|
Faroudi M, Hons M, Zachacz A, Dumont C, Lyck R, Stein JV, Tybulewicz VLJ. Critical roles for Rac GTPases in T-cell migration to and within lymph nodes. Blood 2010; 116:5536-47. [PMID: 20870900 PMCID: PMC3368586 DOI: 10.1182/blood-2010-08-299438] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 09/06/2010] [Indexed: 12/28/2022] Open
Abstract
Naive T cells continuously recirculate between secondary lymphoid tissue via the blood and lymphatic systems, a process that maximizes the chances of an encounter between a T cell and its cognate antigen. This recirculation depends on signals from chemokine receptors, integrins, and the sphingosine-1-phosphate receptor. The authors of previous studies in other cell types have shown that Rac GTPases transduce signals leading to cell migration and adhesion; however, their roles in T cells are unknown. By using both 3-dimensional intravital and in vitro approaches, we show that Rac1- and Rac2-deficient T cells have multiple defects in this recirculation process. Rac-deficient T cells home very inefficiently to lymph nodes and the white pulp of the spleen, show reduced interstitial migration within lymph node parenchyma, and are defective in egress from lymph nodes. These mutant T cells show defective chemokine-induced chemotaxis, chemokinesis, and adhesion to integrin ligands. They have reduced lateral motility on endothelial cells and transmigrate in-efficiently. These multiple defects stem from critical roles for Rac1 and Rac2 in transducing chemokine and sphingosine-1-phosphate receptor 1 signals leading to motility and adhesion.
Collapse
Affiliation(s)
- Mustapha Faroudi
- Medical Research Council National Institute for Medical Research, London, UK
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The concept of an immunological synapse goes back to the early 1980s with the discovery of the relationship between T-cell antigen receptor mediated Ca(2+) signaling, adhesion, and directed secretion. However, this concept did not gain traction until images were published starting in 1998 that revealed a specific molecular pattern in the interface between T cells and model antigen-presenting cells or supported planar bilayers. The dominant pattern, a ring of adhesion molecules surrounding a central cluster of antigen receptors, was observed in both model systems. Analysis of the origins of this pattern over the past 10 years has presented a solution for a difficult problem in lymphocyte biology--how a highly motile cell can suddenly stop when it encounters a signal delivered by just a few antigenic ligands on the surface of another cell without disabling the sensory machinery of the motile cell. The T lymphocyte actively assembles the immunological synapse pattern following a modular design with roots in actin-myosin-based motility.
Collapse
|
40
|
Rac activation by the T-cell receptor inhibits T cell migration. PLoS One 2010; 5:e12393. [PMID: 20811629 PMCID: PMC2928276 DOI: 10.1371/journal.pone.0012393] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Accepted: 08/02/2010] [Indexed: 12/12/2022] Open
Abstract
Background T cell migration is essential for immune responses and inflammation. Activation of the T-cell receptor (TCR) triggers a migration stop signal to facilitate interaction with antigen-presenting cells and cell retention at inflammatory sites, but the mechanisms responsible for this effect are not known. Methodology/Principal Findings Migrating T cells are polarized with a lamellipodium at the front and uropod at the rear. Here we show that transient TCR activation induces prolonged inhibition of T-cell migration. TCR pre-activation leads to cells with multiple lamellipodia and lacking a uropod even after removal of the TCR signal. A similar phenotype is induced by expression of constitutively active Rac1, and TCR signaling activates Rac1. TCR signaling acts via Rac to reduce phosphorylation of ezrin/radixin/moesin proteins, which are required for uropod formation, and to increase stathmin phosphorylation, which regulates microtubule stability. T cell polarity and migration is partially restored by inhibiting Rac or by expressing constitutively active moesin. Conclusions/Significance We propose that transient TCR signaling induces sustained inhibition of T cell migration via Rac1, increased stathmin phosphorylation and reduced ERM phosphorylation which act together to inhibit T-cell migratory polarity.
Collapse
|
41
|
Fonseca AV, Freund D, Bornhäuser M, Corbeil D. Polarization and migration of hematopoietic stem and progenitor cells rely on the RhoA/ROCK I pathway and an active reorganization of the microtubule network. J Biol Chem 2010; 285:31661-71. [PMID: 20682776 DOI: 10.1074/jbc.m110.145037] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Understanding the physiological migration of hematopoietic progenitors is important, not only for basic stem cell research, but also in view of their therapeutic relevance. Here, we investigated the role of the Rho kinase pathway in the morphology and migration of hematopoietic progenitors using an ex vivo co-culture consisting of human primary CD34(+) progenitors and mesenchymal stromal cells. The addition of the Rho kinase inhibitor Y-27632 led to the abolishment of the uropod and microvillar-like structures of hematopoietic progenitors, concomitant with a redistribution of proteins found therein (prominin-1 and ezrin). Y-27632-treated cells displayed a deficiency in migration. Time-lapse video microscopy revealed impairment of the rear pole retraction. Interestingly, the knockdown of ROCK I, but not ROCK II, using RNA interference (RNAi) was sufficient to cause the referred morphological and migrational changes. Unexpectedly, the addition of nocodazole to either Y-27632- or ROCK I RNAi-treated cells could restore their polarized morphology and migration suggesting an active role for the microtubule network in tail retraction. Finally, we could demonstrate using RNAi that RhoA, the upstream regulator of ROCK, is involved in these processes. Collectively, our data provide new insights regarding the role of RhoA/ROCK I and the microtubules in the migration of stem cells.
Collapse
|
42
|
Skelton TS, Tejpal N, Gong Y, Kloc M, Ghobrial RM. Downregulation of RhoA and changes in T cell cytoskeleton correlate with the abrogation of allograft rejection. Transpl Immunol 2010; 23:185-93. [PMID: 20619345 DOI: 10.1016/j.trim.2010.06.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/15/2010] [Accepted: 06/16/2010] [Indexed: 12/15/2022]
Abstract
Proper actin cytoskeleton architecture and dynamics are indispensable for events in the immunological response such as T cell migration, redistribution of T cell receptors, and interaction with antigen presenting cells. Thus, T cell activation, downstream signaling events and effector functions are all actin-dependent. Actin cytoskeleton architecture and dynamics are regulated by proteins belonging to the superfamily of small GTP-binding proteins, such as RhoA GTPase. We previously showed that the administration of an MHC class I allochimeric molecule [alpha1h1/u]-RT1.Aa, which contains donor-type (Wistar Furth, WF; RT1u) immunogenic epitopes displayed on recipient-type (ACI, RT1a) sequences, to the ACI recipient of heterotopic WF heart resulted in the restriction of the TCR repertoire, inhibition of T cell infiltration into the heterotopic cardiac allografts, abrogation of acute and chronic rejection, and induction of indefinite survival of the allograft. Here we show that the allochimeric molecule treatment caused downregulation of RhoA GTPase in T cells. This resulted in dramatic changes in the distribution of actin and the actin-binding protein, Hip55, in these cells, which in turn, inhibited T cell infiltration into the graft. This indicates that the immunosuppressive activity of the allochimeric molecule is achieved via downregulation of the RhoA pathway and disruption of the proper organization of T cell actin cytoskeleton to inhibit T cell functions such as motility and/or TCR signaling events.
Collapse
Affiliation(s)
- T Spencer Skelton
- Department of Surgery, The Methodist Hospital and The Methodist Hospital Research Institute, Houston TX, USA
| | | | | | | | | |
Collapse
|
43
|
Takesono A, Heasman SJ, Wojciak-Stothard B, Garg R, Ridley AJ. Microtubules regulate migratory polarity through Rho/ROCK signaling in T cells. PLoS One 2010; 5:e8774. [PMID: 20098744 PMCID: PMC2808253 DOI: 10.1371/journal.pone.0008774] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Accepted: 12/29/2009] [Indexed: 01/03/2023] Open
Abstract
Background Migrating leukocytes normally have a polarized morphology with an actin-rich lamellipodium at the front and a uropod at the rear. Microtubules (MTs) are required for persistent migration and chemotaxis, but how they affect cell polarity is not known. Methodology/Principal Findings Here we report that T cells treated with nocodazole to disrupt MTs are unable to form a stable uropod or lamellipodium, and instead often move by membrane blebbing with reduced migratory persistence. However, uropod-localized receptors and ezrin/radixin/moesin proteins still cluster in nocodazole-treated cells, indicating that MTs are required specifically for uropod stability. Nocodazole stimulates RhoA activity, and inhibition of the RhoA target ROCK allows nocodazole-treated cells to re-establish lamellipodia and uropods and persistent migratory polarity. ROCK inhibition decreases nocodazole-induced membrane blebbing and stabilizes MTs. The myosin inhibitor blebbistatin also stabilizes MTs, indicating that RhoA/ROCK act through myosin II to destabilize MTs. Conclusions/Significance Our results indicate that RhoA/ROCK signaling normally contributes to migration by affecting both actomyosin contractility and MT stability. We propose that regulation of MT stability and RhoA/ROCK activity is a mechanism to alter T-cell migratory behavior from lamellipodium-based persistent migration to bleb-based migration with frequent turning.
Collapse
Affiliation(s)
- Aya Takesono
- University College London, Department of Biochemistry and Molecular Biology and Ludwig Institute for Cancer Research, London, United Kingdom
- School of Biosciences, University of Exeter, Exeter, United Kingdom
| | - Sarah J. Heasman
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Beata Wojciak-Stothard
- Department of Experimental Medicine and Toxicology, Imperial College London, London, United Kingdom
| | - Ritu Garg
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Anne J. Ridley
- University College London, Department of Biochemistry and Molecular Biology and Ludwig Institute for Cancer Research, London, United Kingdom
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
44
|
Abreu JR, Dontje W, Krausz S, de Launay D, van Hennik PB, van Stalborch AM, Ten Klooster JP, Sanders ME, Reedquist KA, Vervoordeldonk MJ, Hordijk PL, Tak PP. A Rac1 inhibitory peptide suppresses antibody production and paw swelling in the murine collagen-induced arthritis model of rheumatoid arthritis. Arthritis Res Ther 2010; 12:R2. [PMID: 20053277 PMCID: PMC2875627 DOI: 10.1186/ar2900] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 11/13/2009] [Accepted: 01/06/2010] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The Rho family GTPase Rac1 regulates cytoskeletal rearrangements crucial for the recruitment, extravasation and activation of leukocytes at sites of inflammation. Rac1 signaling also promotes the activation and survival of lymphocytes and osteoclasts. Therefore, we assessed the ability of a cell-permeable Rac1 carboxy-terminal inhibitory peptide to modulate disease in mice with collagen-induced arthritis (CIA). METHODS CIA was induced in DBA/1 mice, and in either early or chronic disease, mice were treated three times per week by intraperitoneal injection with control peptide or Rac1 inhibitory peptide. Effects on disease progression were assessed by measurement of paw swelling. Inflammation and joint destruction were examined by histology and radiology. Serum levels of anti-collagen type II antibodies were measured by enzyme-linked immunosorbent assay. T-cell phenotypes and activation were assessed by fluorescence-activated cell sorting analysis. Results were analyzed using Mann-Whitney U and unpaired Student t tests. RESULTS Treatment of mice with Rac1 inhibitory peptide resulted in a decrease in paw swelling in early disease and to a lesser extent in more chronic arthritis. Of interest, while joint destruction was unaffected by Rac1 inhibitory peptide, anti-collagen type II antibody production was significantly diminished in treated mice, in both early and chronic arthritis. Ex vivo, Rac1 inhibitory peptide suppressed T-cell receptor/CD28-dependent production of tumor necrosis factor alpha, interferon gamma and interleukin-17 by T cells from collagen-primed mice, and reduced induction of ICOS and CD154, T-cell costimulatory proteins important for B-cell help. CONCLUSIONS The data suggest that targeting of Rac1 with the Rac1 carboxy-terminal inhibitory peptide may suppress T-cell activation and autoantibody production in autoimmune disease. Whether this could translate into clinically meaningful improvement remains to be shown.
Collapse
Affiliation(s)
- Joana Rf Abreu
- Division of Clinical Immunology and Rheumatology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
L-selectin is a cell adhesion molecule (CAM) that is essential for the tethering and subsequent rolling of naïve lymphocytes along the luminal wall of postcapillary venules entering lymph nodes. As with many CAMs, L-selectin has the capacity to transduce intracellular signals in response to ligand binding. This implicates CAMs involved in tethering and rolling as contributors to intracellular signals that lead to the transition from rolling to arrest. In addition, studies in L: -selectin-null mice have also revealed a role for L: -selectin in chemokine-directed cell migration of leucocytes in tissues. The Ras homology (Rho) family of small GTPases are intracellular proteins that respond to signals received from the surrounding environment. The RhoGTPases typically activate downstream targets involved in the remodelling of the actin cytoskeleton, which is essential for continued progression through the multi-step adhesion cascade. This chapter will focus on how to prepare, perform and monitor RhoGTPase activation assays in response to L: -selectin stimulation. Although this section focuses on L: -selectin stimulation, the methods outlined here can be applied to analysing RhoGTPase activity in response to stimulating other receptors involved in tethering/rolling such as CD44, P-selectin glycoprotein ligand-1 and E-selectin ligand-1.
Collapse
Affiliation(s)
- Marouan Zarrouk
- Cytoskeleton Research Group, Division of Cardiovascular Sciences, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, Hammersmith Hospital Campus, DuCane Road, London, UK
| | | | | |
Collapse
|
46
|
Bunnell SC. Multiple microclusters: diverse compartments within the immune synapse. Curr Top Microbiol Immunol 2010; 340:123-54. [PMID: 19960312 DOI: 10.1007/978-3-642-03858-7_7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The activation of classical alphabeta T cells is initiated when the T cell receptor (TCR) recognizes peptide antigens presented by major histocompatibility complex (pMHC) molecules. This recognition always occurs at the junction of a T cell and antigen-presenting cell (APC). Existing models of T-cell activation accurately explain the sensitivity and selectivity of antigen recognition within the immunological synapse. However, these models have not fully incorporated the diverse microcluster types revealed by current imaging technologies. It is increasingly clear that a better understanding of T-cell activation will require an appreciation of the diverse signaling assemblies arising within the immune synapse, the interrelationships between these structures, and the mechanisms by which underlying cytoskeletal systems govern their assembly and fate. Here, we will provide a brief framework for understanding these issues, review our contributions to current knowledge, and provide perspectives on the future of this rapidly advancing field.
Collapse
Affiliation(s)
- Stephen C Bunnell
- Department of Pathology, Tufts University Medical School, Boston, MA 02111, USA.
| |
Collapse
|
47
|
Rollason R, Korolchuk V, Hamilton C, Jepson M, Banting G. A CD317/tetherin-RICH2 complex plays a critical role in the organization of the subapical actin cytoskeleton in polarized epithelial cells. ACTA ACUST UNITED AC 2009; 184:721-36. [PMID: 19273615 PMCID: PMC2686410 DOI: 10.1083/jcb.200804154] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
CD317/tetherin is a lipid raft–associated integral membrane protein with a novel topology. It has a short N-terminal cytosolic domain, a conventional transmembrane domain, and a C-terminal glycosyl-phosphatidylinositol anchor. We now show that CD317 is expressed at the apical surface of polarized epithelial cells, where it interacts indirectly with the underlying actin cytoskeleton. CD317 is linked to the apical actin network via the proteins RICH2, EBP50, and ezrin. Knocking down expression of either CD317 or RICH2 gives rise to the same phenotype: a loss of the apical actin network with concomitant loss of apical microvilli, an increase in actin bundles at the basal surface, and a reduction in cell height without any loss of tight junctions, transepithelial resistance, or the polarized targeting of apical and basolateral membrane proteins. Thus, CD317 provides a physical link between lipid rafts and the apical actin network in polarized epithelial cells and is crucial for the maintenance of microvilli in such cells.
Collapse
Affiliation(s)
- Ruth Rollason
- Department of Biochemistry, University of Bristol, Bristol BS8 1TD, England, UK
| | | | | | | | | |
Collapse
|
48
|
Abstract
Renewed interest in cell shape has been prompted by a recent flood of evidence that indicates that cell polarity is essential for the biology of motile cells. The uropod, a protrusion at the rear of amoeboid motile cells such as leukocytes, exemplifies the importance of morphology in cell motility. Remodelling of cell shape by uropod-interfering agents disturbs cell migration. But even though the mechanisms by which uropods regulate cell migration are beginning to emerge, their functional significance remains enigmatic.
Collapse
|
49
|
|
50
|
Belkina NV, Liu Y, Hao JJ, Karasuyama H, Shaw S. LOK is a major ERM kinase in resting lymphocytes and regulates cytoskeletal rearrangement through ERM phosphorylation. Proc Natl Acad Sci U S A 2009; 106:4707-12. [PMID: 19255442 PMCID: PMC2660762 DOI: 10.1073/pnas.0805963106] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2008] [Indexed: 11/18/2022] Open
Abstract
ERM (ezrin-radixin-moesin) proteins mediate linkage of actin cytoskeleton to plasma membrane in many cells. ERM activity is regulated in part by phosphorylation at a C-terminal threonine, but the identity of ERM kinases is unknown in lymphocytes and incompletely defined in other mammalian cells. Our studies show that lymphocyte-oriented kinase (LOK) is an ERM kinase in vitro and in vivo. Mass spectrometric analysis indicates LOK is abundant at the lymphocyte plasma membrane and immunofluorescence studies show LOK enrichment at the plasma membrane near ERM. In vitro peptide specificity analyses characterize LOK as a basophilic kinase whose optimal substrate sequence resembles the ERM site, including unusual preference for tyrosine at P-2. LOK's activity on moesin peptide and protein was comparable to reported ERM kinases ROCK and PKC but unlike them LOK displayed preferential specificity for moesin compared to traditional basophilic kinase substrates. Two genetic approaches demonstrate a role for LOK in ERM phosphorylation: cell transfection with LOK kinase domain augments ERM phosphorylation and lymphocytes from LOK knockout mice have >50% reduction in ERM phosphorylation. The findings on localization and specificity argue that LOK is a direct ERM kinase. The knockout mice have normal hematopoietic cell development but notably lymphocyte migration and polarization in response to chemokine are enhanced. These functional alterations fit the current understanding of the role of ERM phosphorylation in regulating cortical reorganization. Thus, these studies identify a new ERM kinase of importance in lymphocytes and confirm the role of ERM phosphorylation in regulating cell shape and motility.
Collapse
Affiliation(s)
- Natalya V. Belkina
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Yin Liu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Jian-Jiang Hao
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Hajime Karasuyama
- Department of Immune Regulation, Tokyo Medical and Dental University Graduate School, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Stephen Shaw
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|